Presidential Symposium
S100: QUIZARTINIB PROLONGED SURVIVAL VS PLACEBO PLUS INTENSIVE INDUCTION AND CONSOLIDATION
THERAPY FOLLOWED BY SINGLE-AGENT CONTINUATION IN PATIENTS AGED 18-75 YEARS WITH NEWLY
DIAGNOSED FLT3-ITD+ AML
H. Erba1,*, P. Montesinos2, R. Vrhovac3, E. Patkowska4, H.-J. Kim5, P. Zak6, P.-N.
Wang7, T. Mitov8, J. Hanyok9, L. Liu9, A. Benzohra9, A. Lesegretain9, J. Cortes10,
A. Perl11, M. Sekeres12, H. Dombret13, S. Amadori14, J. Wang15, M. Levis16, R. Schlenk17
1Duke Cancer Institute, Durham, NC, United States of America; 2La Fe University and
Polytechnic Hospital, Valencia, Spain; 3University Hospital Centre Zagreb, Zagreb,
Croatia; 4Institute of Hematology and Blood Transfusion, Warsaw, Poland; 5The Catholic
University of Korea, Seoul St. Mary’s Hospital, Seoul, South Korea; 6University Hospital
Hradec Kralove, Hradec Kralove, Czechia; 7Chang Gung Medical Foundation, Linkou, Taiwan;
8Daiichi Sankyo UK Ltd, Uxbridge, United Kingdom; 9Daiichi Sankyo, Inc, Basking Ridge,
NJ; 10Augusta University Medical Center, Augusta, GA; 11University of Pennsylvania,
Philadelphia, PA; 12University of Miami Health System, Miami, FL, United States of
America; 13Saint Louis Hospital, University of Paris, Paris, France; 14Tor Vergata
Polyclinic Hospital Rome, Rome, Italy; 15Institute of Hematology and Blood Diseases
Hospital, Tianjin, China; 16Johns Hopkins University, Baltimore, MD, United States
of America; 17Heidelberg University Hospital and German Cancer Research Center, Heidelberg,
Germany
Background: Quizartinib (Quiz) is an oral, highly potent, and selective type II FLT3
inhibitor with single-agent activity in relapsed/refractory FLT3–internal tandem duplication
positive (FLT3-ITD+) acute myeloid leukemia (AML). This is the first report of the
global, randomized, double-blind, placebo (PBO)-controlled phase 3 QuANTUM-First trial
(NCT02668653).
Aims: QuANTUM-First aimed to determine if the addition of Quiz to standard induction
(IND) and post remission (including allogeneic hematopoietic cell transplant [allo-HCT])
in first complete remission [CR1]) consolidation followed by single-agent continuation
therapy for up to 3 years improved survival compared with chemotherapy alone in patients
(pts) with newly diagnosed FLT3-ITD+ AML.
Methods: Pts aged 18-75 y with newly diagnosed AML were centrally screened for FLT3-ITD
prior to initiation of IND with cytarabine 100 mg/m2/day (200 mg/m2/day if institutional
standard) for 7 days and anthracycline (daunorubicin 60 mg/m2/day or idarubicin 12 mg/m2/day)
for 3 days. Pts at 193 sites in 26 countries who were FLT3-ITD+ provided informed
consent and were randomized to Quiz (40 mg/day days 8-21) or PBO and were stratified
by region (North America, Europe, and Asia/Other regions), age (<60 y, ≥60 y), and
white blood cell count (<40×109/L, ≥40×109/L) at diagnosis. A second IND was allowed
if residual AML was noted at the post-IND marrow exam. Pts who achieved CR or CR with
incomplete hematologic recovery (CRi) received up to 4 cycles of high-dose cytarabine
plus Quiz (40 mg/day) or PBO and/or allo-HCT followed by up to 3 y of continuation
therapy with Quiz (30-60 mg/day) or PBO. The primary endpoint was overall survival
(OS).
Results: Between September 2016 and August 2019, 3468 pts were screened, and 539 pts
with FLT3-ITD+ AML were randomized to Quiz (n=268) or PBO (n=271). The median age
was 56 y (range, 20-75 y). Baseline pt and disease characteristics, including FLT3-ITD
variant allele frequency, were balanced between the 2 arms. At data cutoff (August
2021), the median follow-up was 39.2 months and 58 pts remained on continuation therapy.
OS was significantly longer in the Quiz arm than the PBO arm (hazard ratio [HR], 0.776;
95% CI, 0.615-0.979; 2-sided P=.0324). Median OS was 31.9 mo with Quiz vs 15.1 mo
with PBO (Figure). CR/CRi rates were 71.6% and 64.9%, respectively. Allo-HCT in CR1
was performed in 157 pts (Quiz, 31%; PBO, 27%). When censored for allo-HCT, OS trended
longer with Quiz vs PBO (HR, 0.752; 95% CI, 0.562-1.008; 2-sided P=0.055). Relapse-free
survival was longer with Quiz than PBO (HR, 0.733; 95% CI, 0.554-0.969). Although
rates of grade ≥3 adverse events (AEs) were similar across arms, grade ≥3 neutropenia
was more frequent in the Quiz arm (18.1% vs 8.6%). Discontinuations due to AEs occurred
in 20.4% of Quiz and 8.6% of PBO pts. A total of 56 treatment-emergent AEs were associated
with a fatal outcome (Quiz, 11.3%; PBO, 9.7%), mostly due to infections. Grade 3/4
electrocardiogram QT prolonged occurred in 3.0% of Quiz vs 1.1% of PBO pts.
Image:
Summary/Conclusion: These pivotal findings show that the addition of Quiz to standard
chemotherapy and up to 3 years of continuation therapy yielded statistically significant
and clinically meaningful improvements to OS in adults with newly diagnosed FLT3-ITD+
AML up to age 75 y. The manageable safety profile further supports use of Quiz in
combination with standard therapy, including allo-HCT, in FLT3-ITD+ AML.
S101: GENETIC AND EPIGENETIC FACTORS DRIVING PRIMARY MEDIASTINAL B-CELL LYMPHOMA PATHOGENESIS
AND OUTCOME
D. Noerenberg1,*, F. Briest1, C. Hennch1, K. Yoshida2,3, J. Nimo1, R. Hablesreiter1,
Y. Takeuchi2, D. Sasca4, H. Ueno2, L. Mansouri5, Y. Inoue2, L. Wiegand1, A. M. Staiger6,7,
B. Casadei8,9, M. Ziepert10, F. Asmar11, P. Korkolopoulou12, M. Kirchner13, P. Mertins13,
J. Weiner14, E. Toth15, T. Weber16, A. Warth17, T. Schneider18, R.-M. Amini19, W.
Klapper20, M. Hummel21,22, V. Poeschel23, G. Kanellis24, A. Rosenwald25, G. Held23,26,
E. Campo27,28,29, K. Stamatopoulos5,30, I. Anagnostopoulos21,25, L. Bullinger1,22,
N. Goldschmidt31, P. L. Zinzani9,32, C. Bödor33, R. Rosenquist5,34, T. P. Vassilakopoulos35,
G. Ott6, S. Ogawa2,36,37, F. Damm1,22
1Department of Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin
Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin,
Berlin, Germany; 2Department of Pathology and Tumor Biology, Graduate School of Medicine,
Kyoto University, Kyoto, Japan; 3Wellcome Trust Sanger Institute, Hinxton, United
Kingdom; 4Department of Hematology, Oncology, and Pulmonary Medicine, University Medical
Center, Johannes Gutenberg-University, Mainz, Germany; 5Department of Molecular Medicine
and Surgery, Karolinska Institutet, Stockholm, Sweden; 6Department of Clinical Pathology,
Robert-Bosch-Krankenhaus; 7Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology
Stuttgart, and University of Tuebingen, Stuttgart, Germany; 8Istituto di Ematologia
“Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna; 9Dipartimento di
Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna,
Italy; 10Institute of Medical Informatics, Statistics and Epidemiology, University
of Leipzig, Leipzig, Germany; 11Department of Hematology, Copenhagen University Hospital,
Rigshospitalet, Copenhagen, Denmark; 12First Department of Pathology, National and
Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece; 13Core
Unit Proteomics, Berlin Institute of Health, Charité - Universitätsmedizin Berlin
and Max-Delbrück-Center for Molecular Medicine; 14Core Unit Bioinformatics, Berlin
Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; 15National
Institute of Oncology, Budapest, Hungary; 16Department of Internal Medicine IV, Haematology
and Oncology, University Hospital Halle (Saale), Martin-Luther-University Halle-Wittenberg,
Halle; 17Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany;
18National Institute of Oncology, Budapest, Hungary; 19Department of Immunology, Genetics
and Pathology, Uppsala University and University Hospital, Uppsala, Sweden; 20Department
of Pathology, Hematopathology Section and Lymph Node Registry, Universitätsklinikum
Schleswig-Holstein, Kiel; 21Department of Pathology, Charité – Universitätsmedizin
Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin,
Berlin; 22German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ),
Heidelberg; 23Department of Internal Medicine 1 (Oncology, Hematology, Clinical Immunology,
and Rheumatology), Saarland University Medical School, Homburg/Saar, Germany; 24Department
of Hematopathology, Evangelismos General Hospital, Athens, Greece; 25Institute of
Pathology, University of Würzburg and Comprehensive Cancer Center (CCC) Mainfranken,
Würzburg; 26Department Internal Medicine I, Westpfalzklinikum Kaiserslautern, Kaiserslautern,
Germany; 27Centro de Investigacion Biomedica en Red en Oncologia (CIBERONC), Madrid;
28Hospital Clinic of Barcelona, University of Barcelona; 29Institut d’Investigacions
Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; 30Institute of Applied
Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece; 31Hadassah-Hebrew
University Medical Center, Jerusalem, Israel; 32Istituto di Ematologia “Seràgnoli”,
IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; 33HCEMM-SE Momentum
Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental
Cancer Research, Semmelweis University, Budapest, Hungary; 34Clinical Genetics, Karolinska
University Laboratory, Karolinska University Hospital, Stockholm, Sweden; 35Department
of Hematology and Bone Marrow Transplantation, National and Kapodistrian University
of Athens, Laikon General Hospital, Athens, Greece; 36Institute for the Advanced Study
of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan; 37Department of Medicine,
Centre for Haematology and Regenerative Medicine, Karolinska Institutet, Stockholm,
Sweden
Background: Primary mediastinal large B-cell lymphoma (PMBCL) is an aggressive lymphoma
affecting predominantly young female patients. Previous studies in this rare entity
have focused on single genes or were limited in cohort size.
Aims: To unravel the underlying genetic pathogenesis and its impact on outcome, we
embarked on a comprehensive large-scale genetic investigation.
Methods: Specimens of 486 previously untreated PMBCL patients were analyzed by paired
tumor/normal whole-genome (WGS, n=14), whole-exome (WES, n=78) and targeted sequencing
(TS, n=486). To understand the consequences of highly recurrent mutations in the chromatin-modifying
gene ZNF217, we conducted functional and multi-omics analyses in CRISPR/Cas9 engineered
cell lines.
Results: WGS/WES revealed a complex genomic landscape in PMBCL with a median of 85
structural variants, a mutational burden of 5 mutations/Mb, 12 mutated coding candidate
driver genes (CDG) and 4 focal somatic copy-number aberrations per sample (Fig.1a).
Besides known targets, significant breakpoints were identified in genes previously
not implicated in B-lymphomagenesis such as TOX and TP73 (36% and 21%). In addition,
non-coding mutations clustered within the PAX5 enhancer region. With the identification
of 50 recurrently mutated CDGs, we significantly expand the repertoire of known PMBCL
drivers. The 10 most frequently mutated CDGs were SOCS1 (86%), B2M (67%), ITPKB (64%),
ACTB (58%), STAT6 (58%), IGLL5 (56%), TNFAIP3 (53%), NFKBIE (49%), GNA13 (47%), and
ZNF217 (36%), respectively. The operative mutational processes were attributed to
aging, AID/APOBEC activity, defective MMR, and an unexpected infidelity of DNA-Polymerase
ε. Next, we performed TS in 486 samples using a PMBCL-specific 106-gene panel. Recurrent
lesions in 25 epigenetic modifiers were found in >90%, with ZNF217 being among the
most frequently mutated genes (Fig.1b). After knockdown of ZNF217 in Karpas1106P and
L428 cells, we demonstrated altered proliferation, migration, and apoptosis. Using
mass spectrometry, we showed that ZNF217 is acting in a LSD1, CoREST and HDAC containing
histone modifier complex. Accordingly, knockout of ZNF217 led to global changes in
chromatin accessibility with an enrichment of differentially accessible motifs for
crucial lymphoma-associated transcription factors, especially of the NF-κB, BATF/AP1,
and IRF family, but also of CTCF, a major regulator of global 3D chromatin architecture.
Resulting gene expression was characterized by changes in interferon-responsive genes
and inflammation-associated transcription (Fig 1c). Clinical data were available for
329 cases, including 84 cases from clinical trials. Multivariate analysis using an
IPI-corrected Cox regression model was performed. The estimated 5-year PFS and OS
were 77% and 86%. Among the genetic lesions with the strongest association for poor
outcome, we identified patients with mutatedCD58 having a significantly shorter survival
(PFS: HR 2.96; p<.001; OS: HR 2.55; p=.006). In contrast, mutated DUSP2 indicated
longer survival (PFS: HR 0.28; p=.002; OS: HR 0.15; p=.011) (Fig1d). Notably, DUSP2
mutated patients (25%) showed a similar outcome for CR rate, PFS and OS when comparing
CHOP-like and intensified treatment regimens, suggesting no further benefit from treatment
intensification in this very-low risk patient population.
Image:
Summary/Conclusion: Here, we present the genetic landscape of PMBCL highlighting a
previously underappreciated role of chromatin modifying genes, identify novel treatment
targets and provide a solid basis for guiding precision medicine approaches.
S102: COMPREHENSIVE GENOME CHARACTERIZATION REVEALS NEW SUBTYPES AND MECHANISMS OF
ONCOGENE DEREGULATION IN CHILDHOOD T-ALL
P. Pölönen1,*, A. Elsayed1,2, L. Montefiori1, S. Kimura1, J. Myers3, D. Hedges3, J.
Xu4, Y. Hui3, Z. Cheng3, Y. Fan3, Y. Chang1, R. Shraim5, M. Devidas6, S. Winter7,
K. Dunsmore8, J. J. Yang9, T. L. Vincent10, K. Tan10,11,12,13,14, C. Chen10, H. Newman15,
M. Loh16, E. Raetz17, S. P. Hunger18, E. Rampersaud3, T.-C. Chang3, G. Wu3, S. Pounds2,
C. G. Mullighan1,19, D. T. Teachey5,12,20
1Pathology; 2Biostatistics; 3Center for Applied Bioinformatics, St. Jude Children’s
Research Hospital, Memphis; 4Perelman School of Medicine at the University of Pennsylvania;
5Department of Pediatrics and the Center for Childhood Cancer Research, Children’s
Hospital of Philadelphia, Philadelphia; 6Global Pediatric Medicine, St. Jude Children’s
Research Hospital, Memphis; 7Minnesota Research Institute and Cancer and Blood Disorders
Program, Children’s Minnesota Research Institute, Minneapolis; 8University of Virginia
Children’s Hospital, Charlottesville; 9Pharmaceutical Sciences, St. Jude Children’s
Research Hospital, Memphis; 10Division of Oncology and Center for Childhood Cancer
Research; 11Department of Biomedical and Health Informatics, Children’s Hospital of
Philadelphia; 12Perelman School of Medicine; 13Institute for regenerative medicine;
14Penn Epigenetics Institute, University of Pennsylvania; 15Division of Oncology and
Center for Childhood Cancer, Children’s Hospital of Philadelphia, Philadelphia; 16Department
of Pediatrics, Benioff Children’s Hospital and Helen Diller Family Comprehensive Cancer
Center, University of California San Francisco, San Francisco; 17Department of Pediatrics
and Perlmutter Cancer Center, NYU Langone Health, New York; 18Department of Pediatrics
and the Center for Childhood Cancer Research, Children’s Hospital of Philadelphia
and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia;
19Hematological Malignancies Program, St. Jude Children’s Research Hospital, Memphis;
20Divisions of Hematology and Oncology, Children’s Hospital of Philadelphia, Philadelphia,
United States of America
Background: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematopoietic
malignancy including leukemias of early T cell precursor acute lymphoblastic leukemia
(ETP-ALL) and transformed thymocytes. Prior genomic studies of T-ALL had limited cohort
size, excluded refractory disease and focused on alterations in coding parts of the
genome.
Aims: Investigate the genomic basis of T-ALL by identifying both coding and non-coding
alterations and defining T-ALL subtypes.
Methods: We performed whole-genome sequencing (WGS), whole-exome sequencing (WES),
RNA-sequencing of 1,313 cases enrolled on the Children’s Oncology Group AALL0434 trial.
Results: Uniform Manifold Approximation and Projection (UMAP) and gene expression
clustering analyses of RNA-seq data identified 16 subtypes, of which 4 have not been
reported previously. Furthermore, we could divide existing subtypes into smaller subgroups
with common subtype-defining alterations, such as structural variation (SV) and copy
number variation (CNV). TLX1 activation was linked with TLX1-TCR SVs and deletions
in the TLX1 chromosomal domain, whereas TLX3 deregulation was associated with TLX3-BCL11B
enhancer hijacking, but also through TLX3-TCR, TLX3-CDK6 rearrangements. We discovered
two separate NKX2-1 deregulated groups, one characterized by TCR rearrangements and
RPL10 mutations and the other by NKX2-1 CNVs, chromosome 14 chromothripsis, or MYB-TCR
rearrangements. We also observed a distinct group of 9 cases aged 1-2 years, with
recurrent STAG2-LMO2 rearrangements and a patient with inactivating STAG2 mutation
and CELF1 enhancer hijacking by LMO2. Moreover, we found a group of 22 cases that
were highly enriched for ETP-ALL with the following hallmark lesions: BCL11B enhancer
amplification, BCL11B locus SVs involving enhancer hijacking of ARID1B, CCDC26, and
novel CD34+ enhancer hijacking of lincRNA locus in chromosome 6.
Gene expression-based clustering was unable to stratify patients based on TAL1, TAL2,
LMO1, LMO2, LYL1 expression, and their respective activation mechanisms. However,
WGS enabled further characterization of these patients, by identifying several types
of activation mechanisms and co-occurring alterations for each oncogene. We identified
canonical events, such as TAL1-STIL fusions, TCR rearrangements, and activation by
TAL1/LMO1/LMO2 regulatory region indels, but also novel events, such as CD34 specific
enhancer duplications downstream of TAL1 and BCL11B enhancer hijacking by LMO2. We
also observed two smaller clusters with TAL1/LMO2 activation, where one with 39 cases
was associated with TAL1/LMO2 activation and RPL10 mutations and the other with 8
cases had refractory disease (Day 29 MRD >5%). HOXA gene expression-associated subtypes
were defined by fusions involving KMT2A or MLLT10 and PICALM/DDX3X or HOXA9-TCR SVs.
Furthermore, our analysis revealed segregation of patients by HOXA13 or ZFP36L2 rearrangements
and NUP98/NUP214 fusions. Interestingly, HOXA locus breakpoints involving HOXA13 and
HOXA9 were in different chromatin compartments and were associated with mutually exclusive
activation of either HOXA13 or other HOXA genes. Interestingly, HOXA13-deregulation
was associated with ETP-ALL and frequent BCL11B enhancer hijacking, whereas HOXA9
breakpoints typically involved TCRg and were non-ETP.
Image:
Summary/Conclusion: Large-scale analysis of all children enrolled on the AALL0434
study has identified new subtype-defining lesions in T-ALL, including candidate novel
enhancer hijacking events and enhancer duplications that are likely to result in oncogene
deregulation in T-ALL.
S103: EFFICACY AND SAFETY OF ARI0002H, AN ACADEMIC BCMA-DIRECTED CAR-T CELL THERAPY
WITH FRACTIONATED INITIAL THERAPY AND BOOSTER DOSE IN PATIENTS WITH RELAPSED/REFRACTORY
MULTIPLE MYELOMA
C. Fernandez De Larrea1,*, V. González-Calle2, A. Oliver-Caldés1, V. Cabañas3, P.
Rodríguez-Otero4, M. Español-Rego1, J. L. Reguera5, L. López-Corral2, B. Martin-Antonio1,
B. Paiva4, S. Inogés4, L. Rosiñol1, A. López-Díaz de Cerio4, N. Tovar1, M. López-Parra2,
L. G. Rodríguez-Lobato1, A. Sánchez-Salinas3, S. Varea1, V. Ortiz-Maldonado1, J. A.
Pérez Simón5, F. Prósper4, M. Juan1, J. M. Moraleda3, M. V. Mateos2, M. Pascal1, A.
Urbano-Ispizua1
1Hospital Clínic de Barcelona. IDIBAPS. University of Barcelona, Barcelona; 2Hospital
Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca (IBSAL),
Centro de Investigación del Cancer (IBMCC-USAL, CSIC), Salamanca; 3Hospital Clínico
Universitario Virgen de la Arrixaca. IMIB-Arrixaca. University of Murcia, Murcia;
4Clínica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), IDISNA,
CIBERONC Pamplona, Pamplona; 5Hospital Universitario Virgen del Rocío, Instituto de
Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), University of Sevilla, Sevilla, Spain
Background: ARI0002h is a lentiviral autologous CAR T-cell product with a 4-1BB co-stimulatory
domain and a humanized single chain variable fragment targeting BCMA. In pre-clinical
studies, this academic CAR-T has demonstrated potent in vitro and in vivo activity.
Aims: We report the safety and efficacy results of the CARTBCMA-HCB-01 multicenter
clinical trial for patients with relapsed/refractory multiple myeloma (RRMM) (NCT04309981)
who received ARI0002h in 5 Spanish centers.
Methods: Patients (pts) aged 18-75 years old with RRMM were eligible for this study
if they had measurable disease, received ≥2 prior regimens, including a proteasome
inhibitor, an immunomodulatory drug and an anti-CD38 antibody, and were refractory
to the last line of treatment. Bridging therapy was allowed after apheresis. Cyclophosphamide
(300 mg/m2) and fludarabine (30 mg/m2) were used as lymphodepletion regimen. The targeted
dose was 3x106/kg CAR+ cells and was administered in a fractionated manner (10%/30%/60%),
with at least 24h between infusions. A second dose of 3x106 CAR+ cells/kg was planned
at least 4 months after the first dose in pts who achieved any grade of response any
response and had not or serious complications after the first administration. Primary
objectives were overall response rate (ORR; at least partial response -PR-) within
3 months of the first infusion and rate of cytokine release syndrome (CRS) and/or
neurological toxicity in the first 30 days. Response was assessed as per IMWG criteria
and bone marrow minimal residual disease (MRD) was analyzed by next-generation flow
(NGF).
Results: As of February 9th 2022, 35 pts (median age 61 years) with RRMM were included
in the trial. Four pts could not receive ARI-0002h due to MM progression and one died
of infection. Therefore, 30 pts received ARI0002h cells (modified intention-to-treat
population), of which 47% received bridging therapy. Median CAR-T cell production
time was 11 days (range 9-14) with a 100% manufacture success.
Median follow-up after ARI0002h administration for surviving pts was 16 months. The
ORR of 30 evaluable pts was 100%, with a stringent complete remission (sCR) plus very
good partial response (VGPR) rate of 90%. Median time to first response was one month.
Of 26 MRD-evaluable pts at day +100, 92% were MRD-negative in bone marrow by NGF.
53% of patients were alive and without progression at 16 months. Median overall survival
(OS) was not reached and the 16-month OS rate was 80% (Figure 1).
AEs reported in >70% of pts were CRS (87%; grade [gr] 3/4 0%; gr 1 73%), neutropenia
(97%; gr 3/4 100%), anemia (85%; gr 3/4 43%), and thrombocytopenia (79%; gr 3/4 70%).
Median duration of CRS was 4 days (range 1-12). No CAR-T cell-related neurotoxicity
cases were reported. Tocilizumab and corticosteroids were administered in 76% (mainly
for persistent grade 1 CRS) and 12% of pts, respectively.
ARI0002h cells demonstrated peak expansion on day 14 (range 7 days-6 months). 24 out
of 28 eligible pts (86%) received the second dose (range 1.2-3x106 CAR+ cells/kg).
Median time after first infusion was 4 months and 38% received a second lymphodepletion
regimen. No relevant toxicities after second infusions were reported. 7 pts (29%)
improved their response after reinfusion.
Image:
Summary/Conclusion: ARI0002h is the first European academic CART for RRMM that has
demonstrated deep and durable responses and a favorable safety profile, including
the absence of neurotoxicity and the feasibility of a second booster dose.
S104: RBPS DYSREGULATION CAUSE HYPER-NUCLEOLI AND RIBOSOME GAIN-OF-FUNCTION DRIVING
BONE MARROW FAILURE
P. Aguilar-Garrido1,*, M. Velasco1, M. Hernández Sánchez2, M. Á. Navarro Aguadero1,
P. Malaney3, M. JL Aitken3, X. Zhang3, K. H Young3, R. Duan3, P. Hu3, S. Kornblau3,
A. Fernández1, A. Ortiz1, Á. Otero-Sobrino1, P. J. de Andrés2, D. Megías1, M. Pérez1,
J. Gómez1, G. Mata1, J. Martínez López1, S. Post3, M. Gallardo1
1CNIO; 2UCM, Madrid, Spain; 3MD Anderson, Houston, United States of America
Background: Nucleoli and ribosome cross-talk regulates cell translation capacity.
Its dysregulation and impairment drive ribosome and nucleolus stress (NS), related
to the biological mechanism of cancer. Ribosomopathies are a group of diseases characterized
by ribosome defects leading to complex syndromes that include bone marrow failure.
hnRNP K is an RNA binding protein (RBP) in charge of processing nascent RNAs (nucleoli)
into mature mRNAs (ribosome). Our research found a novel ribosome gain-of-function
ribosomopathy phenotype by hyper-nucleoli generation due RBP hnRNP K dysregulation.
Aims: We aim to elucidate how hnRNP K dysregulation impact on haematopoietic stem
cell biology.
Methods:
Hnrnpk overexpression was established in MEFs using CRISPR/SAM (Konermann S. et al,
Nature). Global protein synthesis was tested using a Click-iT OPP and proteasome function
was evaluated by Proteasome 20S activity NS hallmarks were analyzed by confocal microscopy
evaluating Ncl, NS sensor marker. To evoke NS in our cells, we used Actinomycin D
insult. Cell cycle FACS analysis (DAPI) and senescence assays (β-galactosidase staining)
were performed. Molecular mechanism underlying was elucidated by qRT-PCR and WB (p21,
p16, c-Myc, and mTor). To study the impact of hnRNP K overexpression in vivo, we developed
an inducible tamoxifen mouse model activated 30-60 days after birth (Hnrnpk
Tg-Ubc-creERT2). Survival was evaluated by Kaplan-Meier, and phenotype described by
symptoms, signs, CBC and bone marrow IHC panel (CD34, Gr1, B220, MPO).
Results:
Hnrnpk overexpressing cells led to an increment in protein and gene expression of
Ncl, mTor and c-Myc (Figure 1A-B). Moreover, we found an increase in global protein
synthesis (Figure 2C-D). Nevertheless, the elevation of hnRNP K inversely correlated
with the proteasome function, which dropped significantly (Figure 1E). NS induction
promoted higher hnRNP K expression. Additionally, hnRNP K overexpressing cells showed
NS hallmarks associated with an increase of the number of nucleoli, and total area
of the nucleoli and nucleus (Figure 1F-G). Cell cycle analysis confirmed an increment
of arrested G2/M phase cells (Figure 1H-I), linked to an increment in p21 and p16
levels all leading towards a senescent cell phenotype (Figure 1J-L).
HnrnpkTg-Ubc-creERT2 mice had widespread Hnrnpk overexpression (Figure 1M) and a reduction
in lifespan (Figure 1N), mainly due to dysplastic and bone marrow failure phenotype,
with dramatic reduction of CD34 and b-cells, leukopenia, anaemia and thrombocytopenia.
(Figure 1O-Q).
Image:
Summary/Conclusion: The overexpression of hnRNP K drives to an increase in nucleoli
activity, leading to ribosome biogenesis and higher global translation by the regulation
of molecules involved in both systems: Ncl, c-Myc or mTor. Our work found that hnRNP
K overexpression in vivo drives a bone marrow failure phenotype, promoting the exhaustion
of haematopoietic stem cells by ribosome dysregulation that triggered cell senescence.
Of note, this is the first time reported that a nucleoli/ribosome-gain-of-function
induce bone marrow failure ribosomopathies phenotype.
This work was financially supported by CRIS contra el Cancer Association (NGO) AES
ISCIII (PI18/00295), ISCIII Miguel Servet (CP19/00140), Cancer Research UK [C355/A26819],
FC AECC and AIRC under the Accelerator Award Program and National Cancer Institutes
of Health Award (R01CA207204, SMP) Leukemia and Lymphoma Society (6577-19, SMP).
Oral Sessions
S105: IN VIVO PDX CRISPR/CAS9 SCREENS REVEAL MUTUAL THERAPEUTIC TARGETS TO OVERCOME
HETEROGENEOUS ACQUIRED CHEMO-RESISTANCE
A.-K. Wirth1,*, L. Wange2, S. Vosberg3, A. K. Jayavelu4, W Enard2, T Herold5, I Jeremias1
1Apoptosis in Hematopoietic stem cells, Helmholtz Zentrum München, German Research
Center for Environmental Health (HMGU), Munich; 2Anthropology and Human Genomics,
Faculty of Biology, Ludwig Maximilian University (LMU), Martinsried, Germany; 3Clinical
Division of Oncology, Department of Internal Medicine, Medical University of Graz,
Graz, Austria; 4Department of Proteomics and Signal Transduction, Max Planck Institute
of Biochemistry, Martinsried; 5Department of Medicine III, and Laboratory for Leukemia
Diagnostics, Ludwig Maximilian University (LMU), Munich, Germany
Background: Acquired resistance to conventional polychemotherapy regimens leads to
relapse and poor prognosis, and remains a major unmet clinical need.
Aims: To identify therapeutic options to overcome acquired chemo-resistance.
Methods: We studied acute lymphoblastic leukemia (ALL) as model disease and combined
long-term in vivo treatment in orthotopic patient-derived xenograft (PDX) models with
multi-omics profiling and functional genomic CRISPR/Cas9 screens.
Results: We adapted conventional chemotherapeutic protocols to allow treatment of
mice for up to 18 consecutive weeks, using a combination of the widely used drugs
cyclophosphamide and vincristine. Three luciferase-transgenic PDX models were monitored
by repetitive in vivo imaging. Polychemotherapy strongly reduced PDX ALL cells within
the first weeks, proving initial sensitivity of the sample. Under continuous treatment,
tumor load persisted in mice at the level of minimal residual disease for several
weeks, until tumors resumed growth despite treatment, indicating acquired resistance.
In an exemplary PDX model, eight resistant derivatives were generated in replicate
mice and characterized individually. Genomic profiling revealed profound genomic heterogeneity
between distinct derivatives; individual resistant derivatives acquired different
copy number alterations in regions associated with resistance and distinct point mutations,
e.g. in TP53. In contrast to genomic heterogeneity, transcriptome and proteome profiling
identified a group of genes differentially expressed between sensitive and resistant
cells, but similar across all derivatives.
To gain insights into underlying mechanisms and to identify therapeutic targets to
overcome acquired resistance, a customized CRISPR/Cas9 in vivo dropout screen was
performed in individual resistant PDX derivatives, to test the relevance of around
200 candidate genes under treatment. Among others, sgRNAs targeting BCL2, BRIP1 or
COPS2 dropped out in the in vivo screen specifically during treatment; single knockout
experiments confirmed that knockout of either BCL2, BRIP1 or COPS2 re-sensitized PDX
ALL cells towards chemotherapy. Of direct translational relevance, treatment of mice
with the BCL2 inhibitor ABT-199 sensitized resistant PDX cells towards polychemotherapy
in vivo. Interestingly, BCL2 inhibition restored treatment response in resistant derivatives
independently from the highly diverse underlying genetic alterations, e.g., in clones
with and without mutation in TP53.
Summary/Conclusion: Taken together, we established a highly clinically relevant PDX
in vivo model of acquired resistance to conventional chemotherapy. Using this model,
we demonstrate that heterogeneous genomic alterations evolved in parallel in replicate
mice, which could be overcome by a single therapeutic approach to re-sensitize tumors
towards conventional chemotherapy.
S106: UBTF-ATXN7L3 GENE FUSION DUE TO 17Q21.31 DELETION DEFINES NOVEL HIGH-RISK ALL
SUBTYPE AMENABLE TO MRD-BASED TREATMENT INTENSIFICATION
L. Bastian1,*, A. Hartmann1, T. Beder1, S. Hänzelmann1, J. Kässens1, M. Bultmann1,
M. P. Höppner2, S. Franzenburg2, M. Wittig2, A. Franke2, I. Nagel3, M. Spielmann3,
N. Reimer4, H. Busch4, S. Schwartz5, B. Steffen6, A. Viardot7, K. Döhner7, M. Kondakci8,
G. Wulf9, K. Wendelin10, A. Renzelmann11, A. Kiani12, H. Trautmann1, M. Neumann1,
N. Gökbuget6, M. Brüggemann1, C. Baldus1
1Department of Medicine II, Hematology and Oncology, University Medical Center Schleswig-Holstein,
Kiel; 2Institute for Clinical Molecular Biology, Kiel University, Kiel; 3Institute
of Human Genetics, University Medical Center Schleswig-Holstein, Kiel and Lübeck,
Kiel and Lübeck; 4Medical Systems Biology Group and Institute for Cardiogenetics,
University of Lübeck, Lübeck; 5Department of Hematology, Oncology and Tumor Immunology
(Campus Benjamin Franklin), Charité - Universitätsmedizin Berlin, Berlin; 6Department
of Medicine II, Hematology/Oncology, Goethe University Hospital, Frankfurt / Main;
7Department of Internal Medicine III, University Hospital Ulm, Ulm; 8Department of
Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf;
9Department of Hematology and Oncology, University Hospital Göttingen, Göttingen;
10Medical Department V, Hospital Nürnberg, Paracelsus Medizinische Privatuniversität,
Nürnberg; 11Medical Department Oncology and Hematology, University Medical Center
Oldenburg, Oldenburg; 12Department of Medicine IV, Hematology/Oncology, Klinikum Bayreuth,
Bayreuth, Germany
Background: Response to induction chemotherapy assessed by quantification of minimal
residual disease (MRD) is the strongest independent prognosticator in B precursor
acute lymphoblastic leukemia (BCP-ALL). Molecular underpinnings of MRD poor response
are insufficiently understood.
Aims: We aimed to identify novel high-risk subtypes in adult BCP-ALL as cell-intrinsic
determinants of MRD poor response.
Methods: Adult BCP-ALL patients (n=565) were treated according to pediatric inspired
protocols of the German Acute Lymphoblastic Leukemia Study group (GMALL) and profiled
for integrative analyses by RNA-Seq (n=565), SNP-arrays (n=115), whole exome sequencing
(WES; n=84) and whole genome sequencing (WGS; n=3).
Results: Concordance between transcriptomic and genomic profiles was used to allocate
samples to one of 15 established molecular driver subgroups (Figure A). Unsupervised
clustering of gene expression from the remaining samples revealed a distinct cluster
(n=12/565, 2.1%) with an in-frame gene fusion between upstream binding transcription
factor (UBTF) and ataxin-7-like protein 3 (ATXN7L3) as exclusive event in these patients.
Both fusion partners are in direct neighborship at 17q21.31. WGS revealed a 10.08 kb
genomic deletion which truncated UBTF at exon 17/21 and comprised most of the intergenic
region between both genes (Figure B). UBTF-ATXN7L3 rearranged cases frequently harbored
1q gains (n=5/7). Further genomic profiling showed a remarkable paucity of additional
cooperating events compared to other molecular subtypes, supporting a prominent driver
function of the newly identified fusion. UBTF and ATXN7L3 are global epigenetic regulators
involved in transcriptional control. Both genes were highly expressed across the entire
cohort. The gene fusion was associated with a marked increase of Caudal Homeobox 2
(CDX2) expression. Analysis of functional modules related CDX2 to upregulated HOXA9
and MEIS1, described essential co-regulators of KMT2A-driven leukemogenesis. NTRK3
expression was also strongly upregulated, suggesting a possible rationale for specific
inhibitors.
UBTF-AXTN7L3 rearranged patients were older, more frequently female and presented
with normal leukocyte counts, low bone marrow infiltration and pro-B immunophenotypes
or common ALL with reduced CD10 expression. Response to induction chemotherapy in
evaluable patients (n=11) was poor with only 3 patients achieving MRD negativity after
consolidation I compared to n=271/402 (67%; p=0.019) in the remaining cohort. Four
patients suffered either cytologic (n=2) or molecular (n=2) relapse. Immunotherapeutic
treatment intensification using blinatumomab (n=5) or inotuzumab ozogamizin (n=1)
and / or allogenic stem cell transplantation (n=7) in MRD poor responders or relapsed
cases resulted in an overall survival probability of 80% (+/- 12%) vs. 73% (+/- 2%;
p=0.07) in the remaining cohort. Heterogeneous MRD responses were observed for other
molecular subtypes (poor: ZNF384 - 48.2% MRD neg., p=0.056; Ph-like - 54.0% MRD neg.,
p=0.003; KMT2A - 55.8% MRD neg., p=0.127 / good: High Hyperdiploid - 90.9% MRD neg.,
p=0.01; TCF3-PBX1 - 94.1% MRD neg., p=0.016) indicating how molecular drivers affect
chemo-sensitivity in adult BCP-ALL.
Image:
Summary/Conclusion: Molecular driver alterations determine sensitivity to induction
chemotherapy in adult BCP-ALL. UBTF-ATXN7L3 ALL represents a novel subtype with poor
induction chemotherapy response which could be successfully salvaged by MRD-based
treatment intensification using immunotherapeutic strategies.
S107: SOD2 PROMOTES ACUTE LEUKEMIA ADAPTATION TO AMINO ACID STARVATION THROUGH THE
N-DEGRON PATHWAY
N. K. Ibrahim1,*, S. Schreek1, B. Cinar1, L. Loxha1, B. Fehlhaber1, J.-P. Bourquin2,
B. Bornhauser2, C. Eckert3, G. Cario4, M. Forster5, M. Stanulla1, A. Gutierrez6, L.
Hinze1
1Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover,
Germany; 2Department of Pediatric Hematology and Oncology, University Children’s Hospital
Zurich, Zurich, Switzerland; 3Department of Pediatric Hematology and Oncology, Charité
Universitätsmedizin, Berlin; 4Department of Pediatrics I, Christian-Albrecht University
Kiel and University Medical Center Schleswig-Holstein; 5Institute of Clinical Molecular
Biology, Kiel, Germany; 6Division of Pediatric Hematology and Oncology, Boston Children’s
Hospital, Boston, United States of America
Background: The ability of cells to tolerate amino acid starvation is fundamental
for survival under cellular stress conditions. Some cancer cells are vulnerable to
asparagine depletion, which is exploited therapeutically using asparaginase. However,
the mechanisms of adaptation to amino acid starvation in leukemia cells remain incompletely
understood.
Aims: We recently performed a genome-wide CRISPR/Cas9 loss-of-function screen in the
resistant T-ALL cell line CCRF-CEM to identify molecular pathways that promote asparaginase
resistance. We found that Wnt-dependent stabilization of proteins (Wnt/STOP) induces
a profound therapeutic vulnerability to asparaginase in acute leukemias and colorectal
cancers (Hinze et. al., 2019; Hinze et al., 2020). Another unrelated gene on the top
of the screen included SOD2, a mitochondrial superoxide dismutase. Intriguingly, to
date, SOD2 activity has not been linked to a cellular amino acid starvation response,
whose biologic basis we thus sought to further investigate.
Methods: To evaluate the significance of SOD2 in mediating an asparaginase response,
we employed genetic epistasis experiments as well as phenotypic assays including short
hairpin RNA (shRNA) mediated knockdowns, quantitative PCRs, Western blots, amino acid
starvation, and viability assays.
Results: Knockdown of SOD2 (shSOD2) resulted in a profound sensitivity to asparaginase
in several T-ALL and B-ALL cell lines (p<0.0001), and an increase in apoptosis, as
assessed by caspase 3/7 activity (p<0.001). The sensitization was rescued by either
overexpressing SOD2 cDNA (p<0.0001), or by adding the functional SOD2 mimetic MnTBAP
(p<0.01). Of note, shSOD2 mediated sensitization was selective to asparaginase, as
it could not be observed for other commonly used chemotherapeutic agents including
vincristine, doxorubicin, dexamethasone, and 6-mercaptopurine (p=ns). Due to the selectivity
to asparagine depletion, we then investigated whether SOD2 inhibition mediates a broader
amino acid starvation response. Indeed, culturing SOD2-inhibited T-ALL cells in the
absence of essential amino acids (EAA) or non-EAA, induced a significant decrease
in cell viability (p<0.05). Sensitization appeared to be specific to the SOD2 isoform,
and distinct from known SOD2-associated pathways including reactive oxygen species,
cell cycle changes, alterations of mTOR signaling, or glutamine anaplerosis. To better
understand the molecular underpinnings of SOD2 in regulating an amino acid starvation
response, we leveraged the Bioplex Interactome database (Huttlin et al., 2020), and
identified UBR2, an E3 ubiquitin ligase in the N-degron pathway, as a unique binding
partner of SOD2. Ubiquitin E3 ligases target their substrates for ubiquitination,
leading to proteasome-mediated degradation (Yang et al., 2010). Indeed, SOD2 and UBR2
were co-immunoprecipitated, suggesting the formation of a complex that can drive proteasome-dependent
protein catabolism. In line, inhibition of SOD2 significantly decreased ubiquitin
levels, suggesting that SOD2 positively regulates catabolic protein degradation through
the N-degron pathway to promote cancer cell fitness in amino acid starved conditions.
Summary/Conclusion: The interaction of SOD2 and the N-degron pathway represents a
previously unknown molecular adaptation of cancer cells in response to amino acid
starvation. These results serve as a strong proponent for an in-depth characterization
of the N-degron pathway in mediating leukemia cell fitness upon amino acid starvation
and thus provide a basis for therapeutic intervention in refractory leukemias.
S108: PEDIATRIC T- ALL RELAPSE: CONSTITUTIONAL CANCER PREDISPOSITION AND HYPERMUTATATOR
PHENOTYPES
P. Richter-Pechanska1 2,*, J. Kunz1 2, T. Rausch2 3, B. Erarslan-Uysal1 2, B. Bornhauser4,
V. Frismantas4, Y. Assenov5, M. Zimmermann6, M. Happich1, C. von Knebel-Doeberitz1,
N. von Neuhoff7, R. Koehler8, M. Stanulla6, M. Schrappe9, G. Cario9, G. Escherich10,
R. Kischner-Schwabe11, C. Eckert11, S. Avigad12, S. Pfister5 13, M. Muckenthaler1
2, J.-P. Bourquin4, J. Korbel2 3, A. Kulozik1 2 5
1University Hospital Heidelberg; 2MMPU; 3EMBL, Heidelberg, Germany; 4University Children’s
Hospital, Zürich, Switzerland; 5DKFZ, Heidelberg; 6Hannover Medical School, Hannover;
7University Hospital, University of Duisburg-Essen, Essen; 8University Heidelberg,
Heidelberg; 9University Hospital Schleswig-Holstein, Kiel; 10University Medical Center
Hamburg-Eppendorf, Hamburg; 11Charite, Berlin, Germany; 12Schneider Children’s Medical
Center of Israel, Petah Tikva, Israel; 13KiTZ, Heidelberg, Germany
Background: Relapse is the main cause of death from pediatric acute precursor T-cell
leukemia (T-ALL), but the underlying mechanisms of disease evolution from initial
disease to relapse remain incompletely understood and show remarkable interpatient
heterogeneity.
Aims: As cross-sectional studies failed to identify unifying determinants of relapse,
we adopted a longitudinal strategy and performed multi-omic analyses in 13 matched
pairs of initial diagnosis and relapse samples and their matched PDXs. We extended
this set by WES and methylome analyses in an additional cohort of 25 matched DNA samples
from patient cells collected at initial diagnosis, remission, and relapse.
Methods: Thirty-eight patients were recruited from the ALL‐BFM 2000/2009, CoALL97/03/09,
and ALL‐REZ BFM 2002 trials or from Schneider Children’s Medical Center of Israel
at time points of initial diagnosis, remission, and relapse. Material from 13 matched
pairs of PDXs (RNA, cells) was used for multi-omic analyses, including DNA-Seq (WES),
RNA-Seq, ATAC-Seq and methylation analysis with EPIC arrays.
Results: Based on the profile of SNVs and InDels we distinguished 18 (47%) type-1
(derived from the major ancestral clone) and 20 (53%) type-2 relapses (derived from
a minor ancestral clone). We observed stronger remodeling on the way to type 2 than
to type 1 relapses reflected by more evident changes in methylation, chromatin accessibility
and gene expression. At the time of relapse, 3/20 type 2 patients exhibited a hypermutator
phenotype, probably caused by gains of mutations in TP53, BLM and BUB1B combined with
PMS2. Moreover, type 2 T-ALLs were predominantly TAL1-driven (4/8) in contrary to
type 1 (0/5). T-ALLs that later progressed to type-2 relapses exhibited a complex
subclonal architecture, unexpectedly, already at the time of initial diagnosis. The
fraction of subclonal mutations of those T-ALLs that later developed into a type-2
relapse was significantly higher already at the time of initial diagnosis than in
those T-ALLs that later developed into a type-1 relapse (p=0.0387; Fisher’s exact),
a difference that became even more pronounced at the time of relapse (p<0.0001). On
the other hand, relapse type 1 T-ALLs exhibited overexpression of IL7R, its ligand
HGF, and repressors of cytokine signaling (SOCS1, SOCS2, SOCS3) which regulates the
IL7R pathway via negative feedback loop. Deconvolution analysis of ATAC-Seq profiles
showed that T-ALLs later developing into type-1 relapses resembled a predominant immature
thymic T-cell population, whereas T-ALLs developing into type-2 relapses resembled
a mixture of normal T-cell precursors. Moreover, an analysis of remission samples
revealed a significant enrichment of mutations in constitutional cancer predisposition
genes (CPG) in type 2 patients, thus indicating fundamental differences between these
two groups of patients. In both types of relapse, we observed known and novel drivers
of drug resistance including MDR1 and MVP and NT5C2.
Image:
Summary/Conclusion: In sum, our comprehensive analyses revealed fundamentally different
mechanisms driving either type-1 or type-2 T-ALL relapse and indicate that differential
capacities of disease evolution are already inherent to the molecular setup of the
initial leukemia. Leukemias of patients with type-1 relapses were often characterized
by upregulation of the IL7R pathway, whereas type-2 relapses were characterized by
(i) an enrichment of TAL-1 fusion, (ii) and of constitutional mutations in CPG, (iii)
divergent genetic and epigenetic remodeling, and (iv) an enrichment of somatic hypermutator
phenotypes.
S109: ONCOGENIC DEUBIQUITINATION CONTROLS TYROSINE KINASE SIGNALING AND THERAPY RESPONSE
IN ACUTE LYMPHOBLASTIC LEUKEMIA
P. Ntziachristos1,*, Q. Jin2, B. Gutierrez3
1Ghent University, Ghent, Belgium; 2MD Anderson, Houston; 3Northwestern University,
Chicago, United States of America
Background: Dysregulation of kinase signaling pathways via mutations favors tumor
cell survival and resistance to therapy and it is common in cancer. Our data unveil
how dysregulated deubiquitination controls signaling pathways, leading to cancer cell
survival and drug non-response, and suggest novel therapeutic combinations towards
targeting T-cell acute lymphoblastic leukemia (T-ALL). Here, we reveal a novel mechanism
of post-translational regulation of kinase signaling and nuclear receptor activity
via deubiquitination in acute leukemia.
Aims: This study aims at 1) characterizing the function of an oncogenic complex composed
by two deubiquitinating enzymes in in vitro and in vivo leukemia systems and 2) testing
the association of deubiquitinase activity with resistance to therapy in acute lymphoblastic
leukemia.
Methods: We use genetic mouse and human:mouse xenograft models of T-cell leukemia,
biochemical studies (quantitative global proteomics, phosphoproteomics and ubiquitination
analysis) and high-throughput molecular biology (chromatin conformation capture (HiC),
chromatin accessibility (ATAC-Seq) and gene expression (RNA-Seq)) analyses.
Results: We observed that the ubiquitin specific protease 11 (USP11) is highly expressed
in lymphoblastic leukemia and associates with poor prognosis in this disease. USP11
ablation inhibits leukemia growth in vitro and in vivo, sparing normal hematopoiesis
and thymus development, suggesting that USP11 could be a therapeutic target in leukemia.
USP11 forms a complex with USP7 to deubiquitinate the oncogenic lymphocyte cell-specific
protein-tyrosine kinase (LCK). Deubiquitination of LCK controls its activity, thereby
altering T cell receptor signaling. Impairment of LCK activity leads to increased
expression of the glucocorticoid receptor transcript, culminating into transcriptional
activation of pro-apoptotic target genes, and sensitizes cells to glucocorticoids
in T cell leukemia patient samples. The transcriptional activation of pro-apoptotic
target genes, such as BCL2L11, is orchestrated by the deubiquitinase activity and
mediated via an increase in enhancer-promoter interaction intensity. Pharmacological
inhibition of USP7 or genetic knockout of USP7 in combination treatment of glucocorticoid
displayed improved anti-T-ALL efficacy in vivo.
Image:
Summary/Conclusion: Our data unveil how dysregulated deubiquitination controls signaling
pathways, leading to cancer cell survival and drug non-response, and suggest novel
therapeutic combinations towards targeting T-cell leukemia.
S110: A NOVEL AND SUCCESSFUL CD7 GENE KNOCKOUT CAR-T CELL THERAPY FOR RELAPSED OR
REFRACTORY T-CELL HEMATOLOGIC MALIGNANCIES
J. Yang1, J. Li1, X. Zhang1, L. Qiu1, P. Lu1,*
1Hebei Yanda Lu Daopei Hospital, Langfang, China
Background: T cell malignancies represent a group of hematologic cancers with high
relapse and mortality rates. The shared expression of target antigens between chimeric
antigen receptor (CAR) T cells and malignant T cells has limited the development of
CAR-T due to unintended CAR-T fratricide. Here, we develop a fratricide-resistant
anti-CD7 CAR-T modified by CD7 ablation through CRISPR/CAS9 gene editing (KO7CAR).
Aims: In a phase I clinical trial, we explored the efficacy and safety of KO7CAR T-cells
for relapsed or refractory (R/R) T-cell malignancies (NCT04916860 & NCT04938115).
Methods: Peripheral blood mononuclear cells were collected from patients (n=13) or
the transplant donor (n=2) by leukapheresis. CD7-ablated CAR T cells (KO7CAR) were
derived by electroporation of bulk T cells with CD7-targeting Cas9-gRNA RNP 24 hours
before 7CAR transduction. This KO7CAR is a second-generation CAR-T with the co-stimulatory
domain of 4-1BB and CD3ζ targeting CD7. Intravenous fludarabine (30mg/m2/d) and cyclophosphamide
(300mg/m2/d) were given to all patients on day -5 to day -3 prior to KO7CAR-T cells
infusion.
Results: From Oct. 2020 to Oct. 2021, 15 patients with T-cell acute lymphoblastic
leukemia (n=10), T-cell lymphoblastic lymphoma (n=3), and mixed phenotype acute leukemia
(n=2) were enrolled and received KO7 CAR-T cells (Table 1). The median age was 28
(8-46) years old. Four patients had prior hematopoietic stem cell transplantation
(HSCT). At enrollment, 10 patients had bone marrow (BM) blasts >5% by morphology,
and 10 patients had the extramedullary disease (EMD, diffuse involvement, n=8, and
bulky mediastinal masses, n=2). Both patient- and donor-derived KO7CAR-T cells were
successfully generated with a transduction efficiency of 59.0% (22.5%-97.4%). A single
dose of KO7CAR-T cells was infused to patients at low dose (1.5~5x105 cells/kg, n=8),
medium dose (1x106 cells/kg, n=6) or high dose (2x106 cells/kg, n=1).
On day 28, 15/15 (100%) patients achieved minimal residual disease (MRD) negative
complete remission (CR). Among the 10 patients with EMD, 7 achieved EMD CR on day
30, 2 achieved partial response (PR), and 1 who relapsed post 2nd transplant had no
response on day 35 then withdrew. Up to data cutoff Feb.10, 2022, the median follow-up
time was of 309 days (35~407 days). About 2 months post KO7CAR, 12 patients bridged
into allogeneic HSCT, and all remained progression-free after a median time of 253
(30~388) days after HSCT except for 1 who relapsed on day 147 then died from intracerebral
hemorrhage on day 249. The other 2 patients without subsequent HSCT (all had a prior
transplant) died from infection on day 78 and GVHD on day 103, respectively, post
KO7CAR.
Mild cytokine release syndrome (CRS, ≤grade II) occurred in 10/15 (66.7%) patients,
and 5/15(33.3%) patients had grade III CRS. One patient had grade I neurotoxicity,
and 2 had grade III/IV neurotoxicity. All was controlled after the administration
of corticosteroids and/or tocilizumab.
Following infusion, the median peak of circulating KO7CAR-T cells was 1.77×105 (0.279~14.3×105)
copies/μg genomic DNA which occurred around day 20 (day10~ day25) and 63.47% (23.1%~94.18%)
occurring on day15 (day 9~day25) by q-PCR and flow cytometry respectively.
Image:
Summary/Conclusion: This study demonstrated KO7CAR-T therapy had a high efficacy for
CD7+ T-cell malignancies even for those who relapsed post-transplant. Safety was manageable,
however, more data on additional patients and longer observation time are needed to
evaluate the efficacy of KO7 CAR-T products further.
S111: REPEATED INFUSIONS OF ESCALATING DOSES OF EXPANDED AND ACTIVATED AUTOLOGOUS
NATURAL KILLER CELLS IN MINIMAL RESIDUAL DISEASE-POSITIVE PH+ ACUTE LYMPHOBLASTIC
LEUKEMIA PATIENTS. A GIMEMA PHASE 1 TRIAL
G. F. Torelli1,*, S. Chiaretti1, N. Peragine1, W. Barberi1, L. Santodonato2, G. D’Agostino2,
E. Abruzzese3, M. I. Del Principe4, A. Mancino5, M. Matarazzo1, M. S. Bafti1, M Mancini1,
M. Messina5, L. Castiello2, A. Guarini1, R. Foà1
1Hematology, Department of Translational and Precision Medicine, Sapienza University;
2FaBioCell Cell Factory, Istituto Superiore di Sanità; 3Hematology, Sant’Eugenio Hospital,
ASL Roma2, Tor Vergata University; 4Hematology, Department of Biomedicine and Prevention,
Tor Vergata University; 5Fondazione GIMEMA Onlus, Rome, Italy
Background: Due to age and co-morbidities, many Philadelphia-positive acute lymphoblastic
leukemia (Ph+ ALL) patients are ineligible to undergo high-dose chemotherapy or allogeneic
transplant as consolidation treatment. Our group reported the promising results of
the chemo-free scheme D-ALBA based on dasatinib/blinatumomab in induction/consolidation,
underlying the potential role of immunotherapy in this setting (Foà et al, NEJM 2020;383:1616-23).
Considerable interest has been raised by natural killer (NK) cells. We developed a
GMP protocol for NK cell ex vivo expansion in the presence of IL-2 and IL-15, and
report the results of a phase 1 protocol of adoptive immunotherapy with activated
and expanded autologous NK cells for Ph+ ALL patients in complete hematologic remission
(CHR) but with persistent/recurrent minimal residual disease (MRD) ≥60 years or ineligible
for other post-CHR treatment modalities.
Aims: The primary endpoint was to determine the maximum tolerated dose of NK cells
and the recommended dose for subsequent studies. Secondary endpoints were the assessment
of safety and tolerability of the treatment, the immunologic modifications induced
by the procedure and the clinical response to treatment.
Methods: The planned 6 patients were enrolled: 5 in 1st CHR and 1 in 2nd CHR. Patients
underwent repeated infusions (maximum 5) of escalating doses of NK cells, ranging
from 1x106 to 5x107/kg of body weight (BW). No conditioning therapies were administered
before the infusion; patients were allowed to continue tyrosine kinase inhibitors.
Patients underwent a comprehensive MRD monitoring by Q-RT-PCR with a one-year follow-up.
Immunophenotypic analysis on the NK cell product was performed before and after the
expansion. Intracellular cytokine production and PBMC cytotoxic activity against K562
cells, allogeneic and autologous blasts were evaluated after expansion and at time
0 and 7 days from each NK cell infusion.
Results: NK cells presented a 12.3-fold ex vivo expansion. Expanded cells showed an
increased expression of activating receptors and measurable cytotoxicity against primary
allogeneic and autologous blasts. One patient received a maximum NK cell dose of 5x106
cells/kg, 2 patients 1x107 cells/kg and 3 5x107 cells/kg/BW. No patient experienced
infusion-related toxicities. Two adverse events were recorded (grade 1 and 2), both
judged not treatment-related, that resolved after TKI suspension. The higher cell
dose infusion resulted in a significantly increased expression of natural cytotoxicity
receptors, a greater cytokine production by NK, T and NKT cells, and in an increased
capacity of PBMC to lyse K562 cells. These modifications appear persistent over time.
At a 1-year follow-up from the last infusion, 5/6 patients are alive in CHR (Table
1). The MRD levels reduced over time and 4/6 patients reached a complete molecular
response (CMR) or a positive-not-quantifiable (PNQ) status during the study period.
At a median follow-up of 30.8 months from the last infusion, the 5 patients who received
the NK treatment in 1st CHR are still in CMR or PNQ, though 1 patient required additional
treatment. The patient in 2nd CHR at the time of the infusions showed a rise in MRD
and died of disease progression.
Image:
Summary/Conclusion: This phase 1 study demonstrates that autologous NK cells can be
efficiently expanded ex vivo from MRD-positive Ph+ ALL patients in CHR. The infusion
of these expanded cells is safe and induces a marked in vivo host immune response,
suggesting that this approach represents a tolerable and feasible model worthy of
being investigated in larger clinical studies.
S112: TISAGENLECLEUCEL IN PEDIATRIC AND YOUNG ADULT PATIENTS (PTS) WITH RELAPSED/REFRACTORY
(R/R) B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA (B-ALL): FINAL ANALYSES FROM THE ELIANA
STUDY
S. Rives1,*, S. L. Maude2, H. Hiramatsu3, A. Baruchel4, P. Bader5, H. Bittencourt6,
J. Buechner7, T. Laetsch2, B. De Moerloose8, M. Qayed9, H. E. Stefanski10, K. L. Davis11,
P. L. Martin12, E. Nemecek13, C. Peters14, G. Yanik15, A. Balduzzi16, N. Boissel17,
S. L. Khaw18, J. Krueger19, J. Levine20, S. Davies21, G. D. Myers22, A. Yeo23, D.
O’Donovan24, R. Ramos23, M. Pulsipher25, S. Grupp2
1Department of Pediatric Hematology – Oncology, Hospital Sant Joan de Déu Barcelona,
and Institut de Recerca Sant Joan de Déu, Barcelona, Spain; 2Division of Oncology,
Center for Childhood Cancer Research and Cancer Immunotherapy Program, Children’s
Hospital of Philadelphia and Department of Pediatrics, Abramson Cancer Center, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of
America; 3Department of Pediatrics, Graduate School of Medicine, Kyoto University,
Kyoto, Japan; 4University Hospital Robert Debré (APHP) and Université de Paris, Paris,
France; 5Division of Stem Cell Transplantation and Immunology, Hospital for Children
and Adolescents, University Hospital Frankfurt, Frankfurt, Germany; 6Department of
Pediatrics, Faculty of Medicine, University of Montreal, and the Hematology Oncology
Division and Charles-Bruneau Cancer Center, Centre Hospitalier Universitaire Sainte-Justine
Research Centre, Montreal, QC, Canada; 7Department of Pediatric Hematology and Oncology,
Oslo University Hospital, Oslo, Norway; 8Department of Pediatric Hematology-Oncology
and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium; 9Aflac Cancer
and Blood Disorders Center, Emory University, Atlanta, GA; 10National Bone Marrow
Donor Program, Be the Match, Division of Pediatric Blood and Marrow Transplant, University
of Minnesota, Minneapolis, MN; 11Division of Hematology, Oncology and Stem Cell Transplant,
Department of Pediatrics, Stanford University School of Medicine, Stanford, CA; 12Pediatric
Transplant and Cellular Therapy, Duke University Medical Center, Durham, NC; 13Oregon
Health and Science University, Portland, OR, United States of America; 14Stem Cell
Transplantation Unit, St. Anna Children’s Hospital, Vienna, Austria; 15Department
of Pediatrics, University of Michigan Medical Center, Ann Arbor, MI, United States
of America; 16Clinica Pediatrica Università degli Studi di Milano Bicocca, Fondazione
MBBM, Ospedale San Gerardo, Monza, Italy; 17Saint-Louis Hospital (APHP) and Université
de Paris, Paris, France; 18Children’s Cancer Centre, Royal Children’s Hospital and
Murdoch Children’s Research Institute, Parkville, VIC, Australia; 19Division of Haematology/Oncology/Bone
Marrow Transplantation, Department of Paediatrics, The Hospital for Sick Children,
Toronto, ON, Canada; 20Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai,
New York, NY; 21Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; 22Children’s
Mercy Hospital and Clinics, Kansas City, MO; 23Novartis Pharmaceuticals Corporation,
East Hanover, NJ, United States of America; 24Novartis Pharmaceuticals Corporation,
Dublin, Ireland; 25Division of Pediatric Hematology and Oncology, Intermountain Primary
Children’s Hospital, Huntsman Cancer Institute at the University of Utah, Salt Lake
City, UT, United States of America
Background: Pediatric and young adult pts with R/R B-ALL experience a treatment journey
characterized by diminishing likelihood of cure and increasing morbidity. Tisagenlecleucel
is an autologous CD19-directed chimeric antigen receptor (CAR) T-cell therapy approved
for use in pediatric and young adults with B-ALL and adults with B-cell lymphomas.
Tisagenlecleucel provided high rates of remission (>80%) in children and young adults
with R/R B-ALL in ELIANA, with 62% of responders remaining relapse-free at 24 mo (Grupp
et al, Blood, 2018).
Aims: Here, we report the final efficacy and safety analyses in pts followed up to
5.9 years post-tisagenlecleucel infusion.
Methods: ELIANA (NCT02435849) was a pivotal, Phase II, open-label, multicenter, global
study of tisagenlecleucel in pediatric and young adult pts with R/R B-ALL. Pts received
a single infusion of tisagenlecleucel at 0.2-5.0×106 CAR+ viable T cells/kg body weight
for pts ≤50 kg and 0.1-2.5×108 CAR+ viable T cells for pts >50 kg. Endpoints included
overall remission rate (ORR) within 3 mo, relapse-free survival (RFS), duration of
remission (DOR), overall survival (OS), persistence of B-cell aplasia, and short-
and long-term safety events.
Results:
Results: As of September 24, 2021, 97 pts were enrolled and 79 pts (81%) received
tisagenlecleucel. Median time from infusion to data cutoff was 5.5 y; 64 pts had ≥5
y of follow-up. At study entry, the median age was 11 y (range, 3-24). Pts were heavily
pretreated with a median of 3 prior lines of therapy (range, 1-8) and 61% had a history
of prior stem cell transplant (SCT). ORR (complete remission [CR] or CR with incomplete
hematologic recovery within 3 mo after infusion) was 82% (95% CI, 72-90). Among pts
in remission (CR/CRi), the 5y RFS rate was 49% (95% CI, 34-62), and the median RFS
was not reached (Figure, 46.8 mo when censoring for SCT; n=15). The median time to
B-cell recovery was 38.6 mo (95% CI, 23-not reached) and the probability of B-cell
aplasia at 6 mo and 12 mo was 83% (95% CI, 71-91) and 71% (95% CI, 57-82), respectively.
Pts with B-cell recovery (<6 mo, n=10; 6-12 mo, n=4; >12 mo, n=7) experienced a 2y
cumulative incidence of relapse of 25.2% (with SCT treated as a competing risk). Among
all pts, the 5y EFS and OS rates were 42% (95%CI, 29-54) and 55% (95% CI, 43-66),
respectively. There were no significant differences in any efficacy endpoint between
pediatric (<18 y; n=65) and young adult (≥18 y; n=14) pts. No new or unexpected AEs
were reported during long-term follow-up. Among pts in remission, the most commonly
reported grade ≥3 AEs occurring >1 y post-infusion were infection (20%) and cytopenias
(6%). Ten (14%) pts in remission experienced long-term cytopenias persisting for >1
y; however, none of these pts experienced cytopenias persisting for >5 y (median 2
y; range, 1.1-5y). Eighty-two percent of pts received IVIG any time post-infusion.
Image:
Summary/Conclusion: This >5 y follow-up study demonstrates continued durable efficacy
of tisagenlecleucel without late adverse effects in heavily pretreated pediatric and
young adult pts with R/R B-ALL. Tisagenlecleucel continues to be a potentially curative
treatment option for pediatric and young adult patients with R/R B-ALL.
S113: NATIONAL PEGASPARGASE-MODIFIED RISK-ORIENTED PROGRAM FOR PHILADELPHIA-NEGATIVE
ADULT ACUTE LYMPHOBLASTIC LEUKEMIA/LYMPHOBLASTIC LYMPHOMA (PH− ALL/LL). GIMEMA LAL
1913 FINAL RESULTS.
R. Bassan1,*, S. Chiaretti2, I. Della Starza3, O. Spinelli4, A. Santoro5, L. Elia3,
M. S. De Propris3, A. M. Scattolin1, F. Paoloni6, M. Messina7, E. Audisio8, L. Marbello9,
E. Borlenghi10, P. Zappasodi11, C. Vetro12, G. Martinelli13, D. Mattei14, N. Fracchiolla15,
M. Bocchia16, P. De Fabritiis17, M. Bonifacio18, A. Candoni19, V. Cassibba20, P. Di
Bartolomeo21, G. Latte22, S. Trappolini23, A. Guarini24, A. Vitale3, P. Fazi6, M.
Vignetti6, A. Rambaldi4, R. Foà3
1Hematology, Ospedale dell’Angelo, Venice; 2Translational and Precision Medicine,
Sapienza Univesrity of Rome; 3Translational and Precision Medicine, Sapienza University
of Rome, Rome; 4UOC Ematologia, ASST-Papa Giovanni XXIII, Bergamo; 5Divisione di Ematologia
con UTMO, Ospedali Riuniti Villa Sofia-Cervello, Palermo; 6GIMEMA Data Center; 7GIIMEMA
Data Center, Fondazione GIMEMA – Franco Mandelli Onlus, Rome; 8Ematologia, Città della
Salute, Torino; 9Hematology, Niguarda Ca’ Granda Hospital, Milan; 10Hematology, Spedali
Civili, Brescia, Italy, Spedali Civili, Brescia; 11Hematology, Foundation IRCCS Policlinico
San Matteo, Pavia; 12General Surgery and Medical-Surgical Specialties, University
of Catania, Catania; 13Institute of Hematology, L. and A. Seràgnoli, Bologna; 14Hematology,
Ospedale S. Croce, Cuneo, Italy, Cuneo; 15UOC Oncoematologia, Fondazione IRCCS Ca’
Granda Ospedale Maggiore Policlinico di Milano, Milan; 16Hematology Unit, Azienda
Ospedaliera Universitaria Senese, Siena; 17Hematology Division, S. Eugenio Hospital,
Rome; 18Ospedale Policlinico “G.B. Rossi”, University of Verona, Verona; 19Clinica
Ematologica, Azienda Sanitaria Universitaria Integrata di Udine, Udine; 20Divisione
di Ematologia, Ospedale Civile, Bolzano; 21Oncology Hematology, Ospedale Civile, Pescara;
22Hematology, S. Franceso Hospital, Nuoro; 23Clinica di Ematologia, Azienda Ospedaliero
- Universitaria Ospedali Riuniti Umberto I, Ancona; 24Molecular Medicine, Sapienza
University of Rome, Rome, Italy
Background: Pediatric-inspired chemotherapy is standard of care for younger adults
with Ph− ALL/LL. An essential component of these regimens is pegaspargase, here incorporated
into a national treatment program for patients 18-65 years.
Aims: To assess in the GIMEMA Phase 2 LAL 1913 study the feasibility and efficacy
of a pegaspargase-containing induction and consolidation regimen sustaining a risk-oriented
strategy for adult Ph− ALL/LL (ClinicalTrials.gov ID NCT02067143).
Methods: Our prior, reference 8-block chemotherapy protocol (Blood Cancer J 2020;10:119)
was modified to include pegaspargase 2000 IU/m2 at courses 1 (d10), 2 (d8), 5 (d3,
with HD-MTX) and 6 (d8), with dose reductions in patients >55 years (pegaspargase
1000 IU/m2). Serum drug activity was not assessed in this study. Responders were risk-stratified
for allogeneic stem cell transplantation (SCT) or maintenance according to a mixed
risk model based on WBC count, immunophenotype, genetics and post-remission molecular
minimal residual disease (MRD): patients with high-risk (HR) features or MRD ≥ 10-4
at weeks 10-16 or positive at week 22 were eligible to SCT; standard-risk (SR) patients
were eligible to maintenance.
Results: Two hundred and three patients entered the study (median age 39.8 years;
139 B- and 64 T-phenotype). The complete remission (CR) rate was 91% (100% in T-ALL/LL),
with a 3-year cumulative relapse incidence and non-relapse mortality of 24.2% and
12.6%, respectively; 60 patients underwent a SCT. Overall (OS), event-free (EFS) and
disease-free (DFS) survival were 66.7% (95% CI, 60.1-74.1%), 57.7% (95% CI, 51.0-65.3%)
and 63.3% (95% CI, 56.3-71.1%) at 3 years. HR class (n=95) and LL diagnosis (n=20)
did not affect prognosis. T-cell phenotype (CR 100%, P=0.001; EFS 67.1%, P=0.038),
age 18-40 years (EFS 72.6%, P<0.0001) and MRD <10-4 after courses 1 (55%: DFS 77.9%,
P=0.023) and 3 (79%: DFS 75.2%, P=0.048) were prognostically favorable. One hundred
and eighty-seven patients had pegaspargase at course 1 (92.1%, 11 delayed, 3 reduced),
154 at course 2 (84.6%; 11 delayed, 12 reduced), 110 at course 5 (83.9%; 2 delayed,
11 reduced) and 73 at course 6 (68.8%; 3 delayed, 7 reduced). Dose reductions and
delays were related to high-risk profile (liver dysfunction/steatosis, obesity etc.)
or treatment toxicity. Toxicity of grade 2 or more was mainly observed at course 1
(hepatic 12.8%, coagulation/thrombosis 3.2% [enoxaparin prophylaxis recommended with
platelets >30-50], pancreatic 1.6%), contributing to an induction death in 3 patients
(1.4%), but was rare afterwards.
Image:
Summary/Conclusion: This pegaspargase-based ALL regimen was safely applicable to the
majority of study patients, resulting in 3-year OS, EFS and DFS rates >50% in a patient
population aged 18-65. The results were more favorable in patients up to the age of
55, especially in those aged 18-40 years, and in those who achieved maximum MRD response
regardless of age (Figure). Subsequently, a pegaspargase dosing algorithm based on
patient age, body mass index, hepatosteatosis and selected toxicities at first or
prior drug exposure was developed to minimize toxicity, and was used in a successor
GIMEMA trial of sequential chemotherapy-blinatumomab for CD19+ adult B-ALL (EHA Congress
2021, abstract S114).
S114: PONATINIB AND BLINATUMOMAB FOR PATIENTS WITH PHILADELPHIA CHROMOSOME-POSITIVE
ACUTE LYMPHOBLASTIC LEUKEMIA: UPDATED RESULTS FROM A PHASE II STUDY
N. Short1,*, H. Kantarjian1, M. Konopleva1, N. Jain1, F. Ravandi1, X. Huang2, W. Macaron1,
W. Wierda1, G. Borthakur1, T. Kadia1, K. Sasaki1, G. Issa1, G. Montalban-Bravo1, Y.
Alvarado1, G. Garcia-Manero1, C. Dinardo1, J. Thankachan1, R. Delumpa1, E. Mayor1,
W. Deen1, A. Milton1, J. Rivera1, L. Waller1, C. Loiselle1, R. Garris1, E. Jabbour1
1Department of Leukemia; 2Department of Biostatistics, The University of Texas MD
Anderson Cancer Center, Houston, United States of America
Background: Ponatinib and blinatumomab are both highly effective therapies for Philadelphia
chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). The combination of these
two agents may offer an effective chemotherapy-free strategy in these patients (pts).
Aims: We evaluated the efficacy and safety of ponatinib and blinatumomab in pts with
newly diagnosed (ND), relapsed/refractory (R/R) Ph+ ALL or CML in lymphoid blast phase
(CML-LBP). For pts with ND Ph+ ALL, the primary endpoint was the complete molecular
response (CMR) rate. For pts with R/R Ph+ ALL, the primary endpoint was the CR/CRi
rate. Secondary endpoints included safety, event-free survival (EFS) and overall survival
(OS).
Methods: In this phase II study, adults with ND Ph+ ALL, R/R Ph+ ALL, or CML-LBP were
eligible. Pts were required to have a performance status of ≤2, total bilirubin ≤2x
the upper limit of normal (ULN), and ALT and AST ≤3x the ULN. Pts with uncontrolled
cardiovascular disease or clinically significant central nervous system (CNS) comorbidities
(except for CNS leukemia) were excluded. Pts received up to 5 cycles of blinatumomab
as a continuous infusion at standard doses. Ponatinib 30mg daily was given during
cycle 1 and was decreased to 15mg daily once CMR was achieved. After 5 cycles of blinatumomab,
ponatinib was continued for at least 5 years. Twelve doses of prophylactic IT chemotherapy
with alternating cytarabine and methotrexate were administered.
Results: Between 2/2018 to 1/2022, 55 pts were treated (35 with ND Ph+ ALL, 14 with
R/R Ph+ ALL and 6 with CML-LBP). Baseline characteristics are shown in Table 1.
Among the 35 pts with ND Ph+ ALL, 12 were in CR at enrollment (including 2 pts in
CMR). 22 of the 23 evaluable pts (96%) achieved CR/CRi. One pt died on day 18 from
intracranial hemorrhage in the setting of chemotherapy administered prior to enrollment.
After one cycle, 21/33 pts (64%) achieved CMR, and 28/33 pts (85%) achieved CMR at
any time. 11 of 15 tested pts (73%) also became MRD-negative by an NGS assay with
sensitivity of 1x10-6.
CR/CRi was achieved in 12/13 (92%) evaluable pts with R/R Ph+ ALL. CMR was achieved
in 10 pts (71%) after cycle 1 and in 11 pts (79%) overall. 5 of 6 pts with CML-LBP
achieved CR/CRi, and 1 pt achieved PR as best response. 2 pts (40%) achieved CMR.
In the ND Ph+ ALL cohort, 1 of 34 pts who received at least 1 full cycle died in CR;
the other 33 are in ongoing hematologic remission. Only one pt underwent stem cell
transplant (SCT) in first remission for persistently detectable BCR/ABL1 transcripts.
Among 13 responding pts in the R/R Ph+ ALL cohort, 6 proceeded to SCT, 4 did not undergo
SCT and subsequently relapsed, 1 died in CR, and 2 are in ongoing remission without
SCT. In the CML-LBP cohort, 3 of the 5 responding pts subsequently relapsed.
The median follow-up is 11 months (range, 1-46+). For ND Ph+ ALL, the 2-year EFS and
OS are both 93% (Figure 2). There were no relapses or leukemia-related deaths in this
cohort. In the R/R Ph+ ALL cohort, the 2-year EFS rate was 42% and the 2-year OS rate
was 61%. In the CML-LBP cohort, the 2-year EFS was 33% and the 2-year OS was 60%.
The treatment was well-tolerated, and most toxicities were grade 1-2 and consistent
with the known toxicities of the two agents. Two pts discontinued ponatinib due to
toxicity (1 due to stroke and 1 due to DVT). One pt discontinued blinatumomab due
to persistent grade 2 tremor.
Image:
Summary/Conclusion: The chemotherapy-free regimen of simultaneous ponatinib and blinatumomab
is safe and effective in pts with Ph+ ALL. For pts with ND Ph+ ALL, SCT does not appear
to be needed in first remission.
S115: MUTANT NPM1 BINDS CHROMATIN AND COOPERATES WITH MLL1 TO REGULATE ONCOGENIC TRANSCRIPTION
H. Uckelmann1,*, S. Armstrong1, E. Haarer1, E. Wong1, C. Hatton1, F. Perner1, C. Marinaccio1,
C.-W. Chen2
1Pediatric Oncology, Dana-Farber Cancer Institute, Boston; 2Department of Systems
Biology, Beckman Research Institute, City of Hope, United States of America
Background: The dysregulation of stem cell and self-renewal associated genes is a
common phenomenon during leukemia development. In acute myeloid leukemia (AML) around
50 % of cases express high levels of HOXA cluster genes and MEIS1. Most of these AML
cases harbor an NPM1 mutation (NPM1c), which encodes for an oncogene that is mislocalized
from the nucleolus to the cytoplasm. Hence, most studies of NPM1c have focused on
a potential cytoplasmic role. However, it remains unclear how NPM1c expression in
hematopoietic cells leads to its characteristic gene expression pattern. Furthermore,
NPM1c AMLs are highly sensitive to the disruption of the MLL1 histone methyltransferase
complex. Small molecule inhibitors that block the interaction between MLL1 and its
adaptor protein Menin have been shown to impair binding of MLL1 to a subset of its
target genes and to inhibit leukemia cell proliferation and self renewal. Several
MLL1-Menin inhibitors are currently in Phase I/II clinical trials and show promising
activity in patients with NPM1c AML. The effectiveness of these molecules in NPM1c
AML prompts the question whether NPM1c and the wildtype MLL complex cooperate directly
on chromatin to drive leukemic self-renewal.
Aims: In this study we investigated the potential role of NPM1c in regulating oncogenic
transcription on chromatin and the interplay between NPM1c and the histone methyltransferase
complex KMT2A (MLL1).
Methods: We used an endogenously degrader tagged NPM1c leukemia cell line that allows
rapid small molecule induced degradation to show that NPM1c occupies specific chromatin
targets in AML. To characterize the effects of NPM1c degradation on the chromatin
landscape and transcriptional output at genomic loci that are bound by NPM1c we used
ChIPseq, PROseq and nascent RNAseq methods.
Results: Our results show that endogenous NPM1c directly binds to chromatin at specific
target genes, such as HOXA9 and MEIS1, which are highly expressed in NPM1c patient
samples. The loss of NPM1c from its targets leads to specific alterations in active
chromatin marks and RNA Polymerase II (Pol II) chromatin occupancy which are accompanied
by rapid changes in gene expression as well as Pol II transcriptional activity. The
recruitment of NPM1c to chromatin is dependent on the nuclear exporter CRM1 as well
as one of the acidic domains of NPM1c. We further show that NPM1c is lost from specific
loci after treatment with small molecules that disrupt the MLL1-Menin complex interaction
thus functionally linking targeted epigenetic therapy and NPM1c function.
Summary/Conclusion: Overall, we demonstrate that NPM1c directly regulates a network
of leukemia self-renewal associated genes through direct chromatin interaction. We
further found that NPM1c acts in collaboration with the MLL1 complex and define the
mechanism by which MLL1-Menin small molecule inhibitors produce clinical responses
in patients with NPM1-mutated AML.
S116: CELLULAR AND MOLECULAR MECHANISMS OF EVI1-EXPRESSING MLL-REARRANGED ACUTE MYELOID
LEUKEMIA
Hugues-Etienne Châtel-Soulet1, Sabine Juge1, Ana Luisa Pereira2, Frederik Otzen Bagger3,
Alexandar Tzankov4, Mineo Kurokawa5, Athimed El Taher6, Jonathan Seguin1, César Nombela
Arrieta2, Juerg Schwaller1
1Biomedicine, University Children’s Hospital, Basel, Switzerland; 2Medical Oncology
& Hematology, University Hospital, Zürich, Switzerland; 3Center for Genomic Medicine,
University Hospital, Kopenhagen, Denmark; 4Institute for Pathology, University Hospital,
Basel, Switzerland; 5Hematology & Oncology,The University of Tokyo, Tokyo, Japan;
6Biomedicine, University of Basel, Basel, Switzerland
Background: Expression of a doxycycline (DOX)-inducible acute myeloid leukaemia (AML)-associated
iMLL-AF9 fusion transgene in long-term haematopoietic stem cells (LT-HSC) can lead
to an invasive and chemo-resistant disease expressing the transcription factor EVI1.
High EVI1 expression has been suggested as marker of poor outcome in AML patients
even without rearrangements of the EVI1 locus at 3q26.
Aims: We addressed the association between EVI1 expression, the cellular origin and
poor disease outcome in AML driven by the iMLL-AF9 fusion gene.
Methods: The role of EVI1 expression was studied in iMLL-AF9 transgenic mice carrying
an Evi1-IRES GFP reporter in vitro using flow cytometry, colony formation and RT-qPCR
assays, ex vivo with high-resolution bone marrow (BM) imaging, and in vivo, by transplantation
of enriched naïve Evi1+ iMLL-AF9 hematopoietic stem and progenitor cells (HSPC) into
irradiated recipients on DOX. Haematopoiesis of symptomatic mice was analysed by flow
cytometry and histology. For mechanistic studies, single cell and bulk RNA sequencing
was performed on enriched HSPC or BM samples from diseased mice.
Results: Analysis of BM cells from Evi1-IRES-GFP reporter mice revealed that not only
the mostly quiescent LT-HSC but also fractions of the more proliferating multipotent
progenitors (MPP1-3) express abundant Evi1 (“Evi1high”). Induction of the iMLL-AF9
fusion did not result in significant changes in numbers of Evi1+ cells nor levels
of Evi1 mRNA expression in the LT-HSC and MPP1 compartments. However, in colony assays,
Evi1high iMLL-AF9 cells retained a more immature phenotype and produced more colonies
with an invasive morphology than Evi1low cells (n=11, p<0.05). While Evi1 expression
did not influence disease induction upon transplantation of LT-HSC, recipients of
Evi1+ MPP1 cells developed AML earlier than Evi1- MPP1 (n=11, 79 vs. 269d, p<0.05).
Disease induced by Evi1+ cells presented with more extensive leukemic organ infiltration
than Evi1- AML. Evi1 expression also correlated with in vitro Ara-C resistance. We
also examined whether some exogenous factors may increase AML susceptibility by expanding
the Evi1+ HSPC. Although a single injection of recombinant mouse thrombopoietin (TPO)
only increased the number of LT-HSC and not of MPP1, the Evi1high cell fraction was
enlarged in both compartments (LT-HSC: 23 vs. 50%; MPP1: 22 vs. 47%; n=29, p<0.0001)
supported by high-resolution imaging. Interestingly, increased TPO induced HSPC cycling
was confined to the Evi1high cell population (n=3, p<0.05). Transplantation of TPO-treated
iMLL-AF9 LT-HSC or MPP1 resulted in a significantly faster induction of Evi1+ AML
than controls (n=19, MPP1: 35 vs. 79d, p<0.001; LT-HSC: 41 vs. 90d, p<0.001). To better
understand mechanisms of aggravated AML after TPO mediated expansion of Evi1+ HSCP
we performed multiplexed single cell RNA sequencing of highly enriched HSPC cells
in iMLL-AF9 and control mice. While we observed no changes of cellular cluster organisation
2 days after TPO injection, we found some differentially expressed genes in TPO-stimulated
cycling iMLL-AF9 HSPC, including potential stemness regulators.
Summary/Conclusion: Our results suggest that expansion of Evi1-expressing HSPC by
exogenous factors can result in a more aggressive MLL-AF9-driven AML. Ongoing data
exploration and validation may characterize aberrantly expressed genes in TPO-stimulated
Evi1+ iMLL-AF9-expressing HSPC as potential therapeutic targets to impair stemness
of AML cells.
S117: EVI1 DRIVES LEUKEMOGENESIS THROUGH ABERRANT ERG ACTIVATION
J. Schmoellerl1,*, I. Barbosa1, M. Minnich1, F. Andersch1, L. Smeenk2, M. Havermans2,
T. Eder3, T. Neumann1, J. Jude1, M. Fellner1, A. Ebert1, M. Steininger1, R. Delwel2,
F. Grebien3, J. Zuber1
1Research Institute of Molecular Pathology (IMP), Vienna, Austria; 2Department of
Hematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands; 3Institute for Medical
Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
Background: Chromosomal rearrangements leading to overexpression of EVI1 (MECOM) on
chromosome 3q26 define a distinct subtype of acute myeloid leukemia (AML) that is
associated with chemotherapy resistance and a 2-year survival of <10%. While genetic
events driving aberrant expression of EVI1 are increasingly understood, the molecular
functions of EVI1 that drive leukemogenesis are unclear, which has so far precluded
the development of targeted therapeutics.
Aims: We aimed to elucidate transcriptional programs that are maintained by aberrant
EVI1 expression and to systematically identify vulnerabilities of EVI1-driven AML.
Methods: We developed a panel of mouse models that recapitulate phenotypic and transcriptional
hallmarks of patients suffering from EVI1-driven AML, allow tetracycline-controllable
EVI1 expression and the functional interrogation of genetic targets using CRISPR/Cas9.
We mapped transcriptional programs upon acute EVI1 repression in vivo and in vitro,
profiled global EVI1 chromatin occupancy in human AML cell lines and primary patient-derived
AML cells and performed comparative genome-wide CRISPR/Cas9-based loss-of-function
screens in murine and human EVI1-driven AML.
Results: Integration of these datasets revealed a conserved core of genes that is
transcriptionally regulated by EVI1 in murine and human AML, among which we identified
the ETS transcription factor ERG as the only dependency that is highly selective for
EVI1-driven AML. Suppression of ERG specifically triggered cellular differentiation
and apoptosis of EVI1-driven leukemia cells while other AML cell lines were unaffected.
Strikingly, ectopic expression of ERG was sufficient to functionally rescue loss of
EVI1 in EVI1-driven AML cells, suggesting that the major oncogenic function of EVI1
in AML is the aberrant activation of ERG.
Summary/Conclusion: Interfering with the EVI1/ERG regulatory axis may provide entry
points for the development of rational targeted therapies that are urgently needed
for this group of AML patients.
S118: IDENTIFICATION OF DIRECT TRANSCRIPTIONAL TARGET GENES OF NUP98-KDM5A REVEALS
REGULATORY NETWORKS IN ACUTE MYELOID LEUKEMIA
S. Troester1,*, J. Schmoellerl2, T. Eder1, G. Manhart1, G. Winter3, J. Zuber2, F.
Grebien1
1Institute for Medical Biochemistry, University for Veterinary Medicine Vienna; 2Research
Institute of Molecular Pathology (IMP); 3Research Center for Molecular Medicine of
the Austrian Academy of Sciences (CeMM), Vienna, Austria
Background: Oncogenic fusion proteins involving the Nucleoporin 98 (NUP98) gene are
recurrently found in acute myeloid leukemia (AML) with a particular prevalence in
pediatric patients. A chromosomal rearrangement resulting in the fusion of NUP98 to
the gene encoding the lysine-specific demethylase 5A (KDM5A) is the most frequent
NUP98-fusion in infant leukemia and is associated with particularly poor prognosis.
The urgent need for the development of tailored treatments requires a better understanding
of the effects of NUP98-KDM5A on the deregulation of gene expression programs. Although
it has been shown that oncogenic NUP98-fusion proteins act as transcriptional regulators,
it is unclear if and how NUP98-KDM5A directly regulates gene expression to drive leukemia.
Aims: In this study, we aimed to identify immediate critical effectors of the NUP98-KDM5A
fusion protein and to characterize the transcriptional programs through which they
regulate the development and maintenance of NUP98-KDM5A-driven AML.
Methods: We conducted a genome-scale CRISPR/Cas9 loss-of-function screen in a NUP98-KDM5A-driven
murine AML cell line to unravel functional genetic dependencies that could be exploited
to target leukemia cells. In parallel, we developed a new model for degradation tag
(dTAG)-mediated ligand-induced degradation of the NUP98-KDM5A protein to gain a detailed
understanding of direct transcriptional effects of NUP98-fusion-dependent gene regulation.
We used this model to measure immediate changes in transcription upon acute NUP98-KDM5A
degradation by nascent RNA-seq (SLAM-seq). An inducible shRNA system was used to assess
the requirements of direct NUP98-KDM5A target genes for leukemia cell growth and to
measure global gene expression changes by RNA-seq.
Results: Analysis of the CRISPR/Cas9 screen identified 4105 genes that are required
for the proliferation and survival of NUP98-KDM5A-driven AML cells. Complete loss
of the dTAG-NUP98-KDM5A fusion protein was achieved within one hour after ligand addition,
resulting in cell cycle arrest, terminal differentiation and apoptosis of leukemia
cells. Global analysis of nascent mRNA expression by SLAM-seq revealed 45 immediate
NUP98-KDM5A target genes, as their transcription was significantly downregulated upon
fusion protein degradation. Among these genes, 12 were classified as essential factors
for NUP98-KDM5A cell growth from the CRISPR/Cas9 screen. This list included known
target genes of NUP98-fusion proteins, such as members of the Hoxa gene cluster, but
also other transcription factors, enzymes and RNA binding proteins. shRNA-mediated
knockdown of the 12 candidate genes confirmed their essentiality for NUP98-KDM5A leukemia
cell proliferation. Through RNA-seq studies, we found that the knockdown of a small
subset among the 12 candidate genes was able to recapitulate global patterns of gene
deregulation that are induced by NUP98-KDM5A knockdown.
Summary/Conclusion: Using a combination of CRISPR/Cas9 screening and ligand-induced
degradation, we identified direct transcriptional target genes of NUP98-KDM5A that
are functionally essential in AML. Multi-layered investigations of the interplay between
the members of this small network of genes using a variety of models including primary
patient samples will allow us to further dissect their role in the regulation of aberrant
gene expression in NUP98-KDM5A-expressing leukemia cells and might identify novel
therapeutic targets.
S119: CEBPA AND TET2 MUTATIONS COOPERATE TO INDUCE AGGRESSIVE AML VIA GATA-2 DOWNREGULATION
E. Heyes1,*, A. S. Wilhelmson2 3 4, A. Wenzel2 3 4, M. B. Schuster2 3 4, M. Ali3 4,
T. D’Altri2 3 4, T. Eder1, G. Manhart1, E. Rzepa1, L. Schmidt1, M. Meggendorfer5,
T. Haferlach5, G. Volpe6, C. Nerlov7, J. Frampton6, K. Jae Won3 4, F. Grebien1, B.
Porse2 3 4
1Institute of Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna,
Austria; 2The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences; 3Biotech
Research and Innovation Center (BRIC); 4Novo Nordisk Foundation Center for Stem Cell
Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Copenhagen,
Denmark; 5MLL Munich Leukemia Laboratory, München, Germany; 6Institute of Cancer and
Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham,
Birmingham; 7MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine,
University of Oxford, Oxford, United Kingdom
Background: The transcription factor CCAAT-enhancer-binding protein alpha (C/EBPα)
is a master regulator of granulopoiesis and is mutated in 10-15 % of Acute Myeloid
Leukemia (AML) patients. N-terminal frameshifts represent the predominant type of
lesions and ablate the expression of the full-length protein p42, leading to overexpression
of the shorter isoform p30. Mutations in the C-terminal basic-region leucine zipper
(bZip) can disrupt the DNA-binding ability of C/EBPα. AML patients harbor mono- or
biallelic CEBPA mutations (CEBPAmo or CEBPAbi). The most commonly co-occurring mutations
are loss-of-function mutations in the methylcytosine dioxygenase TET2, resulting in
adverse overall survival. We hypothesized that combinatorial effects of CEBPA mutations
together with TET2 loss specifically rewire transcriptional and epigenetic circuitries
in AML cells, thereby strongly influencing disease outcome.
Aims: We aimed to elucidate the molecular mechanisms behind cooperative effects of
CEBPA and TET2 mutations through state-of-the-art transcriptomic and epigenomic analyses
of relevant in vitro and in vivo models as well as data from AML patients.
Methods: We used the CRISPR/Cas9 technology to introduce Tet2 mutations in murine
cell lines expressing only p30 (Cebpa
p30/p30
) or mimicking biallelic CEBPA mutations (Cebpa
p30/C-mut.
) to study the functional cooperation of these mutations in vitro. A Cebpa
-/p30
Tet2
-/- mouse model was used to study effects of Tet2 loss in CEBPA mutated AML. We performed
RNA-, C/EBPα-ChIP-, ATAC-, Bisulfite-, CUT&RUN and CRISPR/Cas9-mediated enhancer screens
to generate a comprehensive dataset for in-depth comparative analysis and correlation
with relevant patient data from the beatAML collection.
Results: Integration of transcriptomic and epigenomic data from in vitro and in vivo
models of CEBPA-TET2 co-mutated AML, in combination with gene expression analyses
in AML patients, identified the transcription factor GATA2 as a conserved target of
the CEBPA-TET2 axis. p30 and TET2 were strongly bound to the -77 kb enhancer of the
Gata2 gene, and CRISPR/Cas9-induced enhancer deletions diminished Gata2 expression.
Furthermore, TET2 loss reduced chromatin accessibility and increased DNA methylation
of the Gata2 promoter, resulting in decreased Gata2 mRNA levels. RNAi-mediated silencing
revealed a dose-dependent effect of Gata2 expression on leukemia cell fitness in vivo.
Reduction of Gata2 levels by 25-50 % provided a strong competitive advantage to Cebpa
p30/p30
cells while near-complete downregulation of Gata2 expression (>75 %) dramatically
reduced cellular fitness. Finally, treatment with the demethylating agent 5-azacytidine
restored Gata2 expression in an AML model with Cebpa and Tet2 mutations and caused
a significant survival benefit.
Summary/Conclusion: The datasets generated from these models enable deeper insights
into the epigenetic and transcriptomic changes that depend on CEBPA and TET2 mutations
in a physiologically relevant mutational context. Our results reveal that mutational
disruption of CEBPA and TET2 results in down-regulated GATA2 expression, causing aggressive
AML. We propose a mechanism in which C/EBPα p30 mediates recruitment of TET2 to regulatory
regions in the GATA2 gene to maintain its expression. Conversely, loss of TET2 leads
to reduced GATA2 levels, which is restored by 5-azacytidine treatment. Thus, interference
with the C/EBPα-TET2 axis may provide entry points for the development of rational
targeted therapies in AML patients with these mutations.
S120: ACUTE MYELOID LEUKEMIA REPRESENTS A FERROPTOSIS-SENSITIVE CANCER ENTITY RAISING
THE POSSIBILITY FOR NOVEL TARGETING STRATEGIES
A. Narr1 2 3,*, H. Alborzinia1 2, E. Donato1 2, F. Vogel4, T. Boch1, A.-M. Leppä1
2, A. Waclawiczek1 2, S. Renders1 2, A. Schulze4, A. Trumpp1 2
1Division of Stem Cells & Cancer, German Cancer Research Center (DKFZ); 2Heidelberg
Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH); 3University
Heidelberg; 4Division of Tumor Metabolismus und Microenvironment, German Cancer Research
Center (DKFZ), Heidelberg, Germany
Background: Despite advances in the treatment of Acute Myeloid Leukemia (AML), the
5-year patient survival rate remains poor with 15-20%. The majority of patients develop
recurrence partly due to resistance mechanisms to classical cell death programs. Exploiting
new forms of cell death therefore may allow novel therapeutic options to limit survival
or occurrence of resistant clones. Ferroptosis has been identified as non-apoptotic
and iron-dependent form of cell death, characterized by an excessive ROS-induced peroxidation
of cell membranes. Recently it has been implicated with a higher sensitivity in cancer
stem cells and drug-resistant cancer cells, potentially making ferroptosis-inducing
(FIN) therapies a promising option.
Aims: We aim to elucidate whether AML represents a ferroptosis-sensitive cancer entity
and is susceptive for potential FIN-therapies.
Methods: IC50 values of ferroptosis inducers were determined and used in combination
with the TCGA-LAML and a pan-cancer CRISPR screen dataset (CERES) to describe ferroptosis
sensitivity of AML. A drug screen with known ROS-inducers was performed in 9 AML cell
lines to identify ferroptosis induction. To mechanistically characterize ferroptotic
cell death by the identified drug on the genetic and metabolic level, we performed
SLAM-RNA-, RNA-Sequencing, Mass Spectrometry and Metabolite Tracing (Cystine/Ser/Gln)
in HL-60 cells in vitro and in vivo. Genetic (CRISPR-Cas9-KO) and chemical inhibition
were used to characterize the role of identified hits and pathways, aiming to design
new synergistic combination therapies.
Results: We find that AML belongs to the most dependent cancer entities for various
ferroptosis-associated genes and represents a ferroptosis-sensitive cancer entity.
Furthermore, a 12-gene-ferroptosis-signature allows us to predict the Overall Survival
of AML patients and shall help to identify a potential cohort for FIN-therapies. By
screening known ROS-inducing drugs for ferroptosis induction, we identify one drug
capable of inducing lipid peroxidation and subsequently ferroptosis in multiple AML
and other tumor cell lines. Notably, in detail characterization of the drug’s mechanism
on the genetic and metabolic level reveal the activity of the iron-metabolism gene
HMOX-1 as essential for ferroptosis induction. Mechanistically, we further find that
this drug directly inhibits GPX-4, one of the major ferroptosis-suppressive regulators,
and facilitates its degradation. Furthermore, we demonstrate that drug treatment results
in an upregulation of different metabolic pathways involved in glutathione (GSH) synthesis;
namely (1) SLC7A11-mediated Cystine-uptake, (2) transsulfuration pathway as well as
(3) glutaminolysis. Genetical and chemical inhibition of these pathways together with
drug treatment in in vitro as well as in vivo studies shows strong synergistic effects
marking them as new targetable vulnerabilities in AML. Using these insights of the
drug’s mechanism of action, we design a synergistic combination therapy and demonstrate
its tumor-eradicating capacity in AML cell lines (in vitro & in vivo) and in primary
de novo as well as relapse AML patient samples (ex vivo).
Summary/Conclusion: These findings classify AML as ferroptosis sensitive and reveal
the capacity of this drug to induce ferroptosis. Detailed characterization of the
drug’s mechanism on the genetic and metabolic level allows us to design an effective
combination therapy exploiting newly identified vulnerabilities in AML. These results
provide a rational basis for the additional pre-clinical and subsequent clinical evaluation
of FIN-therapies in AML patients.
S121: CD123-CD33 COMPOUND CAR-T CELLS WITH NOVEL ANTIGEN BINDING DOMAINS PROVIDE A
NEW HOPE FOR THE TREATMENT OF ACUTE MYELOID LEUKEMIA
Z. Wang1,*, Y. LU1, S. Qiu1, M. Wang1, D. Xiong1, J. Wang1
1State Key Laboratory of Experimental Hematology, National Clinical Research Center
for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe
Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital,
TianJin, China
Background: CAR-T therapy needs to be optimized to treat myeloid malignancies. Antigen-escape
mediated relapse after CAR-T treatment is the vital mechanism responsible for the
nondurable response. Targeting CD33 and CD123 simultaneously could cover almost all
patients with AML. The off-target cytotoxicity of CD123 CAR-T and CD33 CAR-T, especially
on HSPCs, has always been concerned. Several researchers have explored this issue
and the conclusions are inconsistent in different studies.
Aims: To improve the outcome of relapse and/or refractory acute myeloid leukemia and
explore the influence of CD123-CD33 CAR-T on hematopoiesis.
Methods: We develpoed a panel of anti-CD123 monoclonal antibodies (mAbs) using hybridoma
technology, named 6E11, 8D7, 12H7 and 13C3 and incorporated each of the four scFvs
into 4-1BB/CD3ζ signaling domain to generate 4 second generation CARs. T cells were
infected with lentiviral supernatants to generate CAR-T cells. Comparative analysis
were made from the expression property of CAR protein, in vitro and in vivo anti-leukemia
efficacy to screen out the best candidate CD123 CAR. Next, we generated a CD123-CD33
dual targeting CAR in a compound way. We established an escape-simulation model to
explore whether the compound CAR-T could reduce the risk of relapse associated with
antigen loss. Flowcytometry analysis, CFU test and single cell RNA sequecing methods
were jointly used to explore the influence of our CD123-CD33 CAR-T on hematopoiesis.
Results: CD123 CAR constructs with four different novel scFvs differed in CAR protein
expression ratio. A changing trend of CAR proteins from dropping to rising after encountering
target cells was observed in 3 CAR-T cell group except in 8D7 CAR-T. 13C3 CAR was
the most suitable construct from the perspective of basic and dynamic expression.
123CAR-T cells with different scFvs mediated variable anti-leukemia effect in vitro
and in vitro.
123CAR and 33CAR proteins expressed efficiently on the compound CAR-T cell surface
without codon optimization. The compound CAR-T showed superiority in the treatment
of antigen-escape mouse model.
FCM analysis showed no significant difference of residual CD34+ cells proportion between
NCT cell group and compound CAR-T group. scRNA-seq analysis showed that the most undifferentiated
HSC population barely changed among each group. GSEA revealed that the HSC population
in CAR-T groups had no difference in the enrichment of quiescence, cell cycle, G2M
checkpoint related genes compared with that of NCT group. In addition, no downregulation/enrichment
of TGF-beta signaling pathway/ PI3K-AKT-mTOR signaling pathway was observed in compound
CAR-T group compared with NCT group.The CFU assay demonstrated that there were still
quite numbers of hematopoietic colonies formed in compound CAR-T treatment groups.
Image:
Summary/Conclusion: Taken together, in this study we developed a panel of second generation
CD123 CARs with novel scFvs on the basis of 4 novel anti-CD123 mAbs. We proved that
scFv sequence has a significantly impact on CAR expression both originally and dynamically
and further affects CAR-T function, addressing the importance of scFv development
in CAR-T therapy refining. The compound CAR-T in our study minimize the risk of antigen-loss
mediated escape. Notably, wildtype DNA sequence of cCAR without codon optimization
brings no potential risk such as immunogenic complications. scRNA-seq and CFU assay
convincingly demonstrated that the CD123-CD33 compound CAR-T had limited toxicity
on hematopoiesis. Thus, the CD123-CD33 compound CAR-T with novel scFvs provides new
hope for R/R AML patients.
S122: ROCK INHIBITORS TARGET SRSF2 LEUKEMIA BY DISRUPTING CELL MITOSIS AND NUCLEAR
MORPHOLOGY
M. Su1 2,*, T. Fleisher3, I. Grosheva4, M. Horev4, M. Olszewska5, B. Haim6, A. Plotnikov6,
S. Carvalho6, Y. Moskovitz3, M Minden7, N. Chapal-Ilani3, E. Papapetrou8, N. Dezorella9,
T. Cheng10, N. Kaushansky3, B Geiger4, L. Shlush11 12 13
1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences
& Peking Union Medical College, Tianjin, China; 2Molecular and Cellular Biology, Weizmann
Institute of Science, Rehovot; 3Molecular and Cellular Biology, Weizmann Institute
of Science; 4Immunology, Weizmann Institute of Science, Rehovot, Israel; 5Icahn School
of Medicine at Mount Sinai, New York, United States of America; 6Wohl Institute for
Drug Discovery, Weizmann Institute of Science, Rehovot, Israel; 7University Health
Network (UHN), Toronto, Canada; 8Icahn School of Medicine at Mount Sinai, New York,
United States of America; 9Electron Microscopy Unit, Weizmann Institute of Science,
Rehovot, Israel; 10Institute of Hematology & Blood Diseases Hospital, Chinese Academy
of Medical Sciences & Peking Union Medical College, Tianjin, China, Tianjin, China;
11Molecular and Cellular Biology, Weizmann Institute of Science, Rehovot; 12Division
of Hematology, Rambam Healthcare Campus, Haifa; 13Molecular hematology clinic, Maccabi
Healthcare, Tel Aviv, Israel
Background: Spliceosome machinery mutations are common early mutations in myeloid
malignancies, however effective targeted therapies against them are still lacking
in clinical settings.
Aims: Exploring safe and efficient methods to target hematopoietic cells carrying
SRSF2 mutations might be a powerful tool not just for leukemia prevention and treatment,
but also to understand the mechanisms behind SRSF2 mutations.
Methods: We generated five SRSF2 Mut (P95H) isogenic cell lines with CRISPR/CAS9 system
and performed high throughput drug screen with 3988 compounds in a single dose. After
we narrowed down our targets, dose response assay was performed on four different
ROCK inhibitors (ROCKi). The leading ROCKi compound was validated in vivo with SRSF2
Mut AML xenograft models. Next, we aimed at understanding why ROCKi target SRSF2 Mut
cells and used proteomics, gene expression and imaging of the cytoskeletal system
to study the effect of ROCKi on Mut and WT cells.
Results: In the current study, we used an in vitro high-throughput drug screen among
four different isogenic cell lines and identified ROCK inhibitors (ROCKi) as selective
inhibitors of SRSF2 Mut in MOLM14 and AML2 cells. To study the efficacy of RKI-1447
on human samples, we conducted six AML patient-derived xenograft (PDX) experiments,
with SRSF2 Mut samples. In four out of the six primary AML samples, RKI-1447 significantly
reduced engraftment compared to the untreated group and inhibited the engraftment
of both leukemic blasts and pre leukemic-HSPCs (preL-HSPCS). RKI-1447 was not toxic
to mice nor human cells. ROCKi induced mitotic catastrophe (G2M arrest and multipolar
spindles) through their apparent effects on microtubules and nuclear organization,
and leading to cell death. Confocal imaging and transmission electron microscopy (TEM)
data revealed that SRSF2 mutations induce deep nuclear indentation and segmentation,
which is reminiscent of the nuclear shape of Pelger–Huët anomaly (PHA). The nuclear
volume and area were significantly higher in SRSF2 Mut compared with WT MOLM14 cells
regardless of ROCKi. To investigate why SRSF2 Mut cause the structural changes, we
looked into the cytoplasmic intrusions that segment the nuclei, and the structures
that connect the lobes of the nuclei by TEM. This examination revealed enrichment
of microtubule bundles inside the nuclear indentations. More importantly, these microtubules
were located at the tip of the cytoplasmic intrusions suggesting that they take an
active role in the nuclear segmentation process. After exposure to RKI-1447, the cytoskeletal
and nuclear morphological abnormalities of SRSF2 Mut are augmented to a level which
are incompatible with cell survival.
Image:
Summary/Conclusion: We believe it is the first report describing in high resolution
the changes in the nucleus after SRSF2 mutations are introduced. The mechanisms we
identified are most probably relevant to other SMMs and to the dysplastic phenotype
observed in MDS and AML. Accordingly, our findings have wide implications as ROCKi
might be even more useful than describe here. Altogether, we provide data from many
directions that ROCKi should be tested in clinical trials.
S123: DECODING TRANSCRIPTOMIC AND EPIGENETIC CONSEQUENCES OF STRUCTURAL VARIANTS IN
CK-AML AT SINGLE-CELL RESOLUTION
A.-M. Leppä1,*, K. Grimes2, H. Jeong2, T. Boch1, D Karpova1, A. Jauch3, F. Grünschläger1,
A. Dolnik4, L. Bullinger4, A. Krämer5, A. D. Sanders2, J. O. Korbel2, A. Trumpp1
1Division of Stem Cells and Cancer, German Cancer Research Center; 2Genome Biology
Unit, European Molecular Biology Laboratory; 3Institute of Human Genetics, University
of Heidelberg, Heidelberg; 4Hämatologie, Onkologie und Tumorimmunologie, Charité Universitätsmedizin
Berlin, Berlin; 5Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg,
Germany
Background: Acute myeloid leukemia (AML) with complex karyotype (CK) is a heterogenous
AML subgroup with dismal response to standard treatment. CK-AML has remained poorly
characterized at the molecular level, with mainstream techniques struggling to unravel
the complexity of the structural variants (SVs) and link them to phenotypic characteristics.
To improve our understanding of molecular dynamics in CK-AML, we established an integrated
single cell multi-omics approach that combines the analysis of SVs and nucleosome
occupancy (NO) profiling (scNOVA) with concurrent immunophenotypic and transcriptomic
profiling (CITE-seq).
Aims: To characterize the genetic and non-genetic intra-patient heterogeneity at single
cell resolution in CK-AML.
Methods: For scNOVA analysis, blasts from four CK-AML patients were cultured in the
presence of BrdU for the duration of one cell division, followed by single-cell template
strand-sequencing (Strand-seq). For CITE-seq analysis, cells were stained with 38
antibody-derived tags for combined analysis of single cell transcriptome and immunophenotype.
For functional studies, xenografts of the same CK-AML patients were generated by injection
of AML cells into the femoral BM cavity of sublethally irradiated NOD.Prkdc
scid
.Il2rg
null
(NSG) mice.
Results: To characterize cellular hierarchies together with epigenetic, transcriptomic,
and surface protein features of CK-AML, we applied an integrated single-cell analytical
framework, termed scNOVA-CITE, on over 15,000 leukemic cells from four diagnostic
CK-AML patient samples. We detected complex clonal evolution patterns driven by simple
and complex SVs in all patients. Three out of four patients harbored SVs leading to
loss of one TP53 wild-type allele, often accompanied by loss-of-function mutations
in the remaining allele. One patient showed parallel evolution of TP53-deleted and
TP53 wild-type subclones in the same sample, with the latter clone harboring a lower
abundance of SVs compared to the TP53-deleted cells. Moreover, based on NO we inferred
lowest activity of TP53 pathway in the cells with TP53 aberrations, emphasizing the
role of TP53 in intra-patient cytogenetic evolution as well as inter-patient heterogeneity.
Thereby, we unraveled novel single-cell SV landscapes in CK-AML and explored their
epigenetic impact.
To assess in depth the functional outcomes of the genetic complexity, we also characterized
the transcriptome and cell surface proteome using scNOVA-CITE. Two patients showed
high genetic intra-patient heterogeneity and harbored multiple subclones at diagnosis
with unique transcriptomic features. When transplanted into NSG mice, we detected
predominantly monoclonal engraftment and strongly reduced genomic instability in the
respective patient-derived xenografts. The engrafting clones could be traced back
to a small subclone in the original patient samples. Molecular analysis of the subclones
in the diagnostic samples by scNOVA-CITE revealed converging characteristics leading
to stem-like phenotypes. Taken together, these data indicate that cytogenetic evolution
resulting in stemness phenotype in CK-AML is advantageous for leukemic stem cell expansion
in mice, unfolding novel ways to study regulation of stemness and to identify potential
therapeutic targets.
Summary/Conclusion: By developing scNOVA-CITE, we established an integrative single-cell
multi-omics framework allowing to characterize genetic, epigenetic, transcriptomic
and cell surface proteomic properties of CK-AML patient cells. We provide unique insights
into the genetic complexity of CK-AML, and evidence of the resulting functional outcomes.
S124: BONE MORPHOGENETIC PROTEIN 2 TRIGGERS OFF AN IMMUNOSUPPRESSIVE EFFECT OF ΓΔ
T CELLS IN ACUTE MYELOID LEUKEMIA
S. Liang1 2,*, T. Dong1, K. Yue1, H. Gao1, N. Wu1, R. Liu1, J. Liu1, X.-J. Huang1
2
1Peking University People’s Hospital, Peking University Institute of Hematology, National
Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic
Stem Cell Transplantation; 2Peking-Tsinghua Center for Life Sciences, Academy for
Advanced Interdisciplinary Studies, Peking university, Beijing, China
Background: Description of immune landscapes that indulge or restrain survival and
proliferation of malignant cells is critical to the improvement of therapeutic strategies.
Acute myeloid leukemia (AML) remains a severe life-threatening malignancy and often
confronts treatment dilemma in clinic. Although γδ T cells exhibit independent and
potent cytotoxicity against leukemic cells in vitro and in the mouse models, efficacy
of γδ T cell-based immunotherapy on AML patients has seemed unsatisfying so far. How
the anti-AML capacity of γδ T cells is suppressed in vivo is unknown.
Aims: In this study, we aim to dissect the abnormal changes and functions of intrinsic
γδ T cells in the context of AML and dig out the correlated factors in the AML tumor
environment.
Methods: 1. Immunophenotyping analyses of γδ T cells in bone marrows from newly diagnosed
AML patients (n = 62) were detected using flow cytometry, compared with healthy donors
(n=51). The levels of TGF-β family members in the supernatants of bone marrows were
detected by ELISA kits. 2. Peripheral blood mononuclear cells (PBMCs) isolated from
healthy donors were stimulated with AML blasts and morphogenetic protein 2 (BMP2)
to validate clinical phenotypes. Functional experiments were performed to depict the
role of a BMP2-induced subset of γδ T cells in the anti-AML activity of effector γδ
T cells. 3. To confirm the findings described above, we established human CD34+ cells-implanted
mice models followed by injection with luciferase and GFP co-expressing AML cells.
Results: The proportions of total γδ T cells and Vδ1+ fraction were not significantly
different between AML patients and donors. In contrast, the percentage of Vδ2+ T cells
was markedly decreased in AML patients. Meanwhile, a key activating receptor of γδ
T cells, NKG2D, was dramatically decreased and regulatory receptors CD25 and PD-1
were increased on Vδ2+ T cells in AML patients. The expansion capacity of Vδ2+ T cells
was profoundly impaired in AML patients. The functional impairments of γδ T cells
were accompanied with an abnormally increased levels of BMP2, rather than TGF-β, BMP4
and ActivinA, in AML patients. Furthermore, we demonstrated that BMP2 directly induced
an aberrant γδ T cells subset expressing Vδ2+CD25+CD127low (abbreviated as BMP2-Vδ2)
in PBMCs from healthy donors. Consistently, BMP2-Vδ2 cells were significantly increased
in the bone marrows of AML patients compared with donors. The frequencies of BMP2-Vδ2
cells correlated to soluble BMP2 levels and the disease status. Distinct from effector
Vδ2 cells, BMP2-Vδ2 not only lost the ability of killing AML cells, but also was able
to reduce the anti-AML activity of effector Vδ2 cells after co-cultures. In AML-loaded
humanized mice, treatment with BMP2 induced the occurrence of BMP2-Vδ2 cells and reduced
the survival rate of mice. Injection of BMP2-Vδ2 cells significantly attenuated the
activity of effector Vδ2 cells on the elimination of AML cells in mice. Together,
these results demonstrated the immunoregulatory effect of BMP2 on the phenotype and
function of γδ T cells in AML microenvironment.
Summary/Conclusion: We first reported that dysregulated BMP2 in AML triggered an immunosuppressive
subset of γδ T cells, leading to the impairment of anti-AML function of effector γδ
T cells. Our findings provide a novel insight into the mechanisms of immunosuppression
in the context of leukemia and suggest potential targets for the treatment of AML
and other hematopoietic malignancies.
S125: 10-DAY DECITABINE VS. CONVENTIONAL CHEMOTHERAPY (“3 + 7”) FOLLOWED BY ALLOGRAFTING
(HSCT) IN AML PATIENTS ≥60 YEARS: A RANDOMIZED PHASE III STUDY OF THE EORTC LEUKEMIA
GROUP, GIMEMA, CELG, AND GMDS-SG
M. Lübbert1,*, P. Wijermans2, M. Kicinski3, S. Chantepie4, W. van der Velden5, R.
Noppeney6, L. Griskevicius7, A. Neubauer8, M. Crysandt9, R. Vrhovac10, M. Luppi11,
S. Fuhrmann12, E. Audisio13, A. Candoni14, O. Legrand15, R. Foà16, G. Gaidano17, D.
van Lammeren-Venema2, E. F. Posthuma18, M. Hoogendoorn19, A. Giraut3, M. Stevens-Kroef20,
J. H. Jansen21, E. Ammatuna22, J.-P. Vilque4, R. Wäsch1, H. Becker1, N. Blijlevens5,
U. Dührsen6, F. Baron23, S. Suciu3, S. Amadori24, A. Venditti24, G. Huls22
1Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine,
University Medical Center Freiburg, University of Freiburg, Freiburg, Germany; 2Department
of Hematology, Haga Teaching Hospital, The Hague, Netherlands; 3EORTC Headquarters,
Brussels, Belgium; 4Institut d’Hématologie de Basse Normandie, Centre Hospitalo-Universitaire
de Caen, Caen, France; 5Department of Hematology, Radboud University Medical Centre,
Nijmegen, Netherlands; 6Klinik für Hämatologie, Universitätsklinikum Essen, Essen,
Germany; 7Department of Hematology, Oncology and Transfusion Medicine Center, Vilnius
University Hospital Santaros Klinikos, Vilnius University, Vilnius, Lithuania; 8Department
of Internal Medicine, Hematology, Oncology and Immunology, Philipps University Marburg
and University Hospital Gießen and Marburg, Campus Marburg, Marburg; 9Department of
Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty,
Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen,
Germany; 10Department of Haematology, University Hospital Centre Zagreb, Zagreb, Croatia;
11Dipartimento di Scienze Mediche e Chirurgiche Materno-Infantili e dell’Adulto, University
of Modena and Reggio Emilia, Azienda Ospedaliera Universitaria, Modena, Italy; 12Department
of Hematology and Oncology, HELIOS Hospital Berlin-Buch, Berlin, Germany; 13SC Ematologia
Città della Salute e della Scienza Torino, Torino; 14Clinica Ematologica Azienda Sanitaria
Universitaria Integrata di Udine, Udine, Italy; 15Service d’Hématologie Clinique et
de Thérapie cellulaire, Hôpital Saint Antoine, APHP, Paris, France; 16Ematologia,
Dipartimento di Medicina Traslazionale e di Precisione, “Sapienza” Università di Roma,
Rome; 17Division of Hematology, Department of Translational Medicine, Università del
Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara,
Italy; 18Department of Internal Medicine, Reinier de Graaf Hospital, Delft; 19Department
of Hematology, Medical Center Leeuwarden, Leeuwarden; 20Radboud University Medical
Center, Nijmegen, Netherlands; 21Dept laboratory Medicine, Lab Hematology, Radboud
University Medical Center, Nijmegen; 22University Medical Center Groningen, Groningen,
Netherlands; 23University of Liège, Liège, Belgium; 24Department of Biomedicine and
Prevention, University of Rome Tor Vergata, Rome, Italy
Background: Older, fit AML patients (pts) treated with induction chemotherapy (IC)
have poor long-term survival unless HSCT is performed. DNA-hypomethylating agents
have become the backbone of AML therapy in pts unfit for IC. Promising outcomes have
been reported for the 10-day decitabine (DEC) schedule, suggesting it may be a better
treatment prior to HSCT as compared to IC.
Aims: To compare efficacy and safety of 10-day DEC followed by allografting to IC
followed by allografting in older fit AML pts.
Methods: This was an international open-label randomized phase III trial (NCT02172872).
Key inclusion criteria were newly diagnosed AML, age ≥60 years, eligible for IC, WHO
performance status 0-2. DEC was administered 10 days consecutively in cycle 1 (20 mg/m2),
10 or 5 days in subsequent cycles (depending on bone marrow blast clearance at day
28). IC treatment was daunorubicin 60 mg/m2 x 3 days, cytarabine 200 mg/m2 x 7 days,
followed by 1-3 additional chemotherapy cycles. Pts who had an HLA-matched donor and
at least stable disease were encouraged to undergo HSCT after ≥1 treatment cycle.
Pts from the DEC arm not receiving HSCT could continue DEC treatment. The primary
endpoint was overall survival (OS). Pts were randomized 1:1, stratified by de novo
AML vs. secondary AML, age (60-64 vs 65-70 vs ≥70 yrs), and institution. The statistical
design aimed to detect a hazard ratio (HR) for OS of 0.75 (HR<1 indicates longer survival
for DEC), requiring 441 deaths (one-sided alpha 0.025, 85% power). Due to the slow
accumulation of deaths, the final analysis was performed with a clinical cut-off (CCO)
date June 30, 2021, following the Data Monitoring Committee recommendation.
Results: Between 12/2014 and 8/2019, 606 pts were randomized, 303 in each arm. Median
follow-up was 4.0 yrs. Median age was 68 yrs (range 60-81), 34% of pts were ≥70 yrs
old and 57% were male, 21% and 32% had good and adverse ELN 2017 risk profile, respectively.
A median of 3 DEC cycles (Q1-3: 2-5) and 2 IC cycles (Q1-3: 1-2) were administered.
The CR/CRi rate was 48% with DEC and 61% with IC. HSCT as part of the protocol was
performed in 122 pts (40%, 30 of them not in CR/CRi) from the DEC and 118 (39%, 11
of them not in CR/CRi) from the IC arm, and in 52% in both arms at any time. By the
CCO, 423 deaths occurred. The OS was not significantly different between DEC and IC
groups (HR=1.04, 95% confidence interval [CI]: 0.86-1.26; 2-sided p=0.68). The median
OS was 15 months (95% CI: 13-18) in the DEC and 18 months (95% CI: 14-22) in the IC
group. The OS rates (%) after 1, 2, 3 and 4 years for the DEC and IC groups were 58
vs 59, 38 vs 40, 30 vs 33, and 26 vs 30, respectively. In age subgroups, the estimated
HR for OS for DEC vs IC was 1.34 (99% CI: 0.79-2.28) for pts aged 60-64, 1.14 (99%
CI: 0.77-1.69) for pts aged 65-69, and 0.84 (99% CI: 0.55-1.26) for pts aged ≥70 yrs
(p-value for trend: 0.058). Notable differences in the incidence of grade 3-5 adverse
events (%) reported (before HSCT) were: febrile neutropenia (37% for DEC vs 57% for
IC), decrease in platelets (24% for DEC vs 32 % for IC), oral mucositis (2% for DEC
vs 10% for IC), diarrhea (1% for DEC vs 8% for IC), decrease in neutrophils (19% for
DEC vs 13% for IC). The 30-day mortality rate was 3.6% for DEC and 6.4% for IC. The
incidence of grade 5 treatment-related adverse events after HSCT was comparable in
both treatment arms (25% for DEC and 22% for IC).
Summary/Conclusion: Treatment with DEC resulted in a similar OS and HSCT rate but
a better safety profile compared to IC in older AML pts ≥60 yrs, eligible for IC.
S126: GEMTUZUMAB-BASED INDUCTION CHEMOTHERAPY COMBINED WITH MIDOSTAURIN FOR FLT3 MUTATED
AML. RESULTS FROM THE NCRI AML19 “MIDOTARG” PILOT TRIAL
N. Russell1,*, C. Wilhelm-Benartzi2, J. Othman3, R. Dillon3, N. Potter3, J. Jovanovic3,
A. Gilkes4, L. M. Batten2, J. Canham2, E. L. Hinson2, P. Kottaridis5, J. Cavenagh6,
C. Arnold7, M. Dennis8, S. Knapper4
1Haematology, Guy’s and St Thomas’ NHS Foundation Trust, London; 2Centre for Trials
Research, Cardiff University, Cardiff; 3Department of Medical and Molecular Genetics,
Kings College London, London; 4School of Medicine, Cardiff University, Cardiff; 5Haematology,
University College London Hospitals NHS Foundation Trust; 6Department of Haematology,
St Bartholomew’s Hospital, London; 7Haematology, Belfast City Hospital, Belfast; 8Haematology,
The Christie NHS Foundation Trust, Manchester, United Kingdom
Background: Following the RATIFY study Midostaurin in combination with “7 + 3” like
chemotherapy has become the standard of care for patients with newly diagnosed FLT3
mutated AML. The ALFA 0701 trial also suggested a benefit for Gemtuzumab Ozogamicin
(GO) in FLT3 mutated AML however the combination of GO-based induction with Midostaurin
has not been formally assessed.
Aims: To assess the safety of midostaurin in combination with Gemtuzumab and intensive
induction therapy and to assess impact of the combination on MRD kinetics
Methods: The NCRI AML19 trial randomised patients to receive DA 3 + 10 (Daunorubicin
60mg/m2 on days 1,3,5 plus AraC 100mg/m2 bd on days 1-10) plus a single dose of GO
3mg/m2 on day 1 (DAGO1) or 2 doses (3mg/m2, maximum 5mg) on days 1 and 4 (DAGO2) plus
50mg bd of midostaurin (m) for 14 days following completion of chemotherapy. Eligibility
included age 18-60 years with a FLT3-ITD or TKD mutation. Enhanced pharmacovigilance
(PV) was performed for four weeks following the first induction course. Midostaurin
was also given following second induction (DA 3 + 8 without GO) and 2 courses of HDAC
consolidation and as maintenance for 12 cycles in non-transplanted patients. From
November 2020 to November 2021, 77 patients were enrolled into the Midotarg pilot
receiving DAGO1m (n=39) or DAGO2m (n=38). 59 had a FLT3 ITD and 22 a FLT3-TKD (and
4 had both). 59 patients have completed course 1 and are evaluable. RT-qPCR MRD monitoring
for patients with an NPM1 mutation (n=48) was performed following each cycle of chemotherapy.
A descriptive comparison is presented here of toxicity and MRD kinetics with FLT3
mutated patients receiving DAGO1 or DAGO2 without Midostaurin in the same trial.
Results: Treatment compliance in course 1 was 88% for DAGO1m and 100% for DAGO2m.
One patient did not receive any Midostaurin because of colitis during induction. Dose
interruption for QTc prolongation occurred in 1 patient. A total of 17 SAEs (CTC grade
3 or greater) were reported (GO1, n=11, GO2 n= 6). Day 60 mortality was 0%. No cases
of VOD were reported. Blood count recovery was not delayed. Time to neutrophil recovery
to 1 x 109/L was 31 and 32 days with DAGO1m and DAGO2m compared to 31 and 32 days
in DAGO1 and DAGO2 .. Time to platelet recovery to 100 x 109/L was 28 and 29 days
with DAGO1m and DAGO2m respectively compared to 30 days in DAGO1 and DAGO2.
Complete remission (CR plus CRi) was achieved in 51/59 (88%) evaluable patients who
have completed induction with DAGOm. In 44 evaluable patients in remission with NPM1
mutation, 34 (74%) were MRD negative in the peripheral blood after course 2. This
compares with 63% in 54 evaluable patients with DAGO only. Bone marrow NPM1 transcript
levels after courses 1 to 4 were compared with FLT3 mutated patients receiving DAGO1
and DAGO2 without Midostaurin (Figure 1) with a higher proportion of patients becoming
MRD negative from course 2 onwards and 81% being MRD negative after course 4.
Image:
Summary/Conclusion: The addition of midostaurin to DAGO using a single or fractionated
dose of GO was well tolerated with no evidence of increased toxicity, high response
rate and with encouraging clearance of NPM1 mutant transcripts. A randomised study
of DAm versus DAGOm in FLT3 mutated AML is planned
S127: QUIZARTINIB WITH DECITABINE AND VENETOCLAX (TRIPLET) IS ACTIVE IN PATIENTS WITH
FLT3-ITD MUTATED ACUTE MYELOID LEUKEMIA - A PHASE I/II STUDY
M. Yilmaz1,*, M. Muftuoglu1, H. Kantarjian1, C. DiNardo1, T. Kadia1, G. Borthakur1,
N. Pemmaraju1, N. Short1, Y. Alvarado1, A. Maiti1, L. Masarova1, G. Montalban Bravo1,
S. Loghavi2, K. Patel2, S. Kornblau1, E. Jabbour1, G. Garcia-Manero1, M. Andreeff1,
N. Daver1
1Leukemia; 2Hematopathology, MD Anderson Cancer Center, Houston, United States of
America
Background: Quizartinib (QUIZ), a potent 2nd generation FLT3 inhibitor (FLT3i) demonstrated
synergy with venetoclax (VEN) in AML cell lines and PDX models (Mali Haematologica
2020).
Aims: To evaluate the safety and efficacy of Decitabine (DAC) + VEN + QUIZ triplet
in patients (pts) with newly diagnosed (ineligible for intensive induction chemotherapy)
or relapsed/refractory (R/R; up to 5 prior chemotherapies) FLT3 ITD mutated AML.
Methods: All pts received 10 days of DAC (20 mg/m2) in Cycle 1. Pts underwent day
14 bone marrow (BM) biopsy, and VEN (400 mg/day starting from day1) was put on hold
in pts with BM blasts ≤ 5% or aplasia. Those with day14 BM blast >5% continued VEN
for 21 days during cycle 1. In subsequent cycles, DAC was reduced to 5 days. QUIZ
(30 or 40 mg/day) was administered daily continuously.
Results: Overall 28 pts were enrolled and evaluable at the time of this report. Of
23 pts with R/R AML (median 3 prior Rx, 78% with ≥1 prior FLT3i including prior gilteritinib
in 70%, and 39% had a prior alloSCT), 78% achieved CRc (3 CR, 15 CRi) with 6/16 and
5/18 responders achieving FLT3-PCR and multicolor flow cytometry negativity, respectively.
The CRc rates were 75% and 72% in pts who received prior gilteritinib and prior HMA+VEN,
respectively. 60-day mortality rate was 5%. Of 5 pts with newly diagnosed AML (median
age 69), all achieved CRc (2 CR, 3 CRi) with 4/5 and 2/4 responders achieving FLT3-PCR
and MFC negativity, respectively. 60-day mortality in frontline was 0.
RAS/MAPK mutations appear to drive both primary and secondary resistance. Pts with
underlying RAS/MAPK mutations had the lowest response rates, at 40% compared to 94%
in those without. None of the six pts who achieved a durable remission (> 6months)
had RAS/MAPK mutations at baseline (Figure A). 4 of 16 (25%) of pts who relapsed (<
6 months of remission) or were refractory to this triplet regimen had baseline RAS/MAPK
mutations. We had pre- and post-treatment NGS data from 8 pts who had a response but
then relapsed (Figure B). Emergent RAS/MAPK mutations were noted in 37% of relapses
(3/8), while emergent FLT3 F691L gatekeeper mutations was noted in 25% of relapses
(2/8). Interestingly, there were no emergent FLT3 TKD mutations.
No pts developed a dose limiting toxicity (DLT) with 30 mg/day quizartinib, however
with the 40mg/day quizartinib 2 pts developed hematologic DLT (grade 4 neutropenia
with a <5% cellular bone marrow lasting ≥42 days). Hence, quizartinib 30 mg/day dose
was determined to be the recommended phase 2 dose for the triplet. Most common Grade
3/4 non-hematologic toxicities included lung infections (42%) and neutropenic fever
(30%). No QTcF prolongations >480 msec were noted.
With a median follow-up (f/u) of 13 months, the median OS was 7.6 months in R/R cohort.
Median OS in prior Gilt exposed pts was 6.3 months and ≥1 prior FLT3i exposed pts
was 6.3 months. 8/18 R/R pts (including 5/8 prior Gilt exposed pts) underwent ASCT
with a median OS of 19 vs 8 months in pts who underwent ASCT versus not (p=0.26).
Of the 5 frontline responding pts median OS was 14.5, 2 were alive in CR, 1 died in
CR1 post-ASCT, 2 died due to progressive disease at the last f/u.
Image:
Summary/Conclusion: DAC + VEN + QUIZ is active in R/R FLT3-ITD mutated AML pts, with
CRc rates of 78% and the median OS of 7.6 months. The high response rate was maintained
in prior Gilteritinib exposed pts. Interestingly, RAS/MAPK mutations but not emergent
TKD mutations were associated with primary and secondary resistance to the triplet.
Accrual continues and updated clinical, NGS and mass cytometry (CyTOF) data will be
presented.
S128: A RANDOMISED COMPARISON OF CPX-351 AND FLAG-IDA IN HIGH RISK ACUTE MYELOID LEUKAEMIA.
RESULTS FROM THE NCRI AML19 TRIAL
N. Russell1,*, C. Wilhelm-Benartzi2, S. Knapper3, L. Batten2, J Canham2, E. Hinson2,
U. Malthe Overgaard4, J. Othman5, R. Dillon5, P. Mehta6, P. Kottaridis7, J. Cavenagh8,
C. Hemmaway9, C. Arnold10, M. Dennis11
1Guy’s and St Thomas’ NHS Foundation Trust, Department of Haematology, London; 2Centre
for Trials Research; 3School of Medicine, Cardiff University, Cardiff, United Kingdom;
4Copenhagen University Hospital, Copenhagen, Denmark; 5Department of Medical and Molecular
Genetics, Kings College London, London; 6University Hospitals of Bristol and Weston
NHS Trust, Bristol; 7University College London Hospitals NHS Foundation Trust; 8Department
of Haematology, St Bartholomew’s Hospital, London, United Kingdom; 9Aukland Hospital,
Aukland, New Zealand; 10Belfast City Hospital, Belfast; 11The Christie NHS Foundation
Trust. On behalf of the NCRI AML Working Group, Manchester, United Kingdom
Background: Liposomal daunorubicin and cytarabine (CPX-351) has shown a survival advantage
for older patients (>60years) with secondary AML compared to 3 + 7 chemotherapy however
in younger patients there is a lack of randomised evidence for benefit. We have previously
reported improved survival with FLAG-Ida treatment as treatment intensification for
younger patients identified with high risk (HR) AML following induction therapy and
for patients with secondary AML (Burnett AK et al. Leukemia. 2018 Dec;32(12):2693-2697)
and considered this regimen an appropriate comparator for trials in younger patients.
Aims: We compared CPX-351 with FLAG-Ida in a randomised fashion in patients who were
either HR at trial entry based on cytogenetics or identified as HR following induction
or at relapse.
Methods: The AML19 trial (ISRCTN78449203) randomised CPX-351 vs FLAG-Ida in 635 patients
mainly <60 years with HR AML or MDS (>10% blasts) (median age 53.6 yrs). Three groups
of HR patients were randomised 2:1 in favour of CPX with the aim of proceeding to
allogeneic transplant. Group 1 (n=195) had known adverse risk cytogenetics (Grimwade
et al, Blood 2010,116, 354) and were randomised at diagnosis between 4 courses of
CPX-351 and 2 courses of FLAG-Ida followed by MACE/MidAC consolidation. Group 2 (n=263)
were HR by validated risk score, had FLT3-ITD without an NPM1 mutation, had refractory
disease and were randomised after induction course 1. Group 3 (n=177) were randomised
after course 2 if they had persisting MRD at the time of relapse. Here we present
results for Group 1. Group 1 was not powered to claim statistical significance; therefore,
these results are intended to be exploratory and hypothesis generating.
Results: Group 1 included 49.2% with de novo AML 20.3% patients with secondary AML
and 30.5% with HR MDS.
The Overall response rate(CR/CRi) was 64.8% for CPX-351 and 74.4% for FLAG-Ida (univariate
OR:0.57, 95%CI 0.30-1.10, p=0.09). Overall survival (OS) at 3 years was 32% and 24%,
median OS was 13.3 months vs 10.2 months (univariate HR:0.83, 95%CI 0.58-1.18, p=0.3)
for CPX -351 and FLAG-Ida respectively (Figure 1a). Event free survival (EFS) was
not significantly different (HR:0.91 95%CI: 0.50-1.64, p=0.76). Relapse free survival
(RFS) at 3 years was 43% and 28%, median RFS was 22.1 months vs 14 months (univariate
HR:0.66, 95%CI 0.41-1.06, p=0.09) for CPX -351 and FLAG-Ida respectively (Figure 1b).
RFS was significant when adjusting for NPM1 mutation status or FLT3 mutation status
using multivariable cox regression model with RFS being better with CPX-351 compared
to FLAG-Ida (HR:0.58, 95% CI0.36-0.95, p=0.03). Median duration of remission favoured
CPX and was 319.5 days vs 167 days (p=0.046) for CPX vs FLAG-Ida respectively. Haematological
toxicity was greater in course 1 with CPX-351 with platelet recovery to 100x109/L
at 34.5 days versus 29 days (p<0.001) and neutrophil recovery to 1.0x109/L at 32 days
vs 30 days.(p=0.12). Day 30 and day 60 mortality were not significantly different
between arms with 4.8% vs 7.3% (p=0.46) and 12.4% vs 11.0% (p=0.77) for CPX-351 and
FLAG-Ida respectively.Compliance was better with CPX-351 with 90.7% vs 83.0% receiving
the scheduled course 1 dose. More patients receiving CPX-351 were transplanted (50.5%
vs 41.5%) with the median number of courses given prior to transplant 2 in both arms.
Image:
Summary/Conclusion: In patients with adverse cytogenetics CPX-351 did not improve
response, OS or EFS compared to FLAG-Ida but was associated with better duration of
remission and RFS. Further follow-up is needed to determine the clinical significance
of those differences.
S129: TAKEAIM LEUKEMIA- A PHASE 1/2A STUDY OF THE IRAK4 INHIBITOR EMAVUSERTIB (CA-4948)
AS MONOTHERAPY OR IN COMBINATION WITH AZACITIDINE OR VENETOCLAX IN RELAPSED/REFRACTORY
AML OR MDS
G. Garcia-Manero1,*, E. S. Winer2, D. J. DeAngelo2, S. Tarantolo3, D. A. Sallman4,
J. Dugan5, S. Groepper6, A. Giagounidis6, K. Götze7, K. H. Metzeler8, C.-C. Li9, L.
Zhou9, E. Martinez9, M. Lane9, R. von Roemeling9, M. Bohme9, A. S. Kubasch10, A. Verma11,
U. Platzbecker10
1Luekemia, MD Anderson Cancer Center, Houston; 2Dana-Faber Cancer Institute, Boston;
3Nebraska Cancer Specialist, Omaha; 4Moffitt Cancer Center, Tampa; 5Novant Health
Cancer Institute, Forsyth Medical Center, Winston-Salem, United States of America;
6Marien Hospital/ Univ. of Dusseldorf Germany, Dusseldorf; 7Faculty of Medicine Technical
University of Munic, Munich; 8Hematology, Cellular Therapy and Hemostaseology, University
Hospital Leipzig, Leipzig, Germany; 9Curis, Lexington, United States of America; 10University
Hospital Leipzig, Leipzig, Germany; 11Albert Einstein College of Medicine, Montefiore
Medical Center, Bronx, United States of America
Background: Emavusertib (CA-4948) is a novel oral inhibitor of interleukin-1 receptor-associated
kinase 4 (IRAK4) and FLT3. IRAK4 is critical in triggering inflammation, oncogenesis,
and survival of cancer cells. Genetic mutations in the splicing factors SF3B1 and
U2AF1 drive overexpression of a highly active long isoform of IRAK4 and have been
associated with disease progression and poor prognosis of high-risk myelodysplastic
syndrome (HR-MDS) and acute myeloid leukemia (AML).
Aims: Assessment of safety, clinical activity, and Recommended Phase 2 Dose (RP2D)
of emavusertib as monotherapy or in combination with azacitidine (AZA) or venetoclax
(VEN).
Methods: This is an open-label, phase 1/2a dose escalation and cohort expansion trial
(NCT04278768). In phase 1 Dose Escalation, patients with R/R AML or HR-MDS are treated
with emavusertib monotherapy. Phase 1b includes 2 arms of combination therapy: emavusertib
+ AZA and emavusertib + VEN. The primary objectives of this study are to assess the
safety, clinical activity, and identify the RP2D of emavusertib as monotherapy or
in combination with AZA or VEN in R/R AML or HR-MDS. The Phase 2a Dose Expansion includes
patients for emavusertib monotherapy: R/R AML with FLT3 mutation, or AML and HR-MDS
R/R to HMA with U2AF1 or SF3B1 mutations.
Results: As of December 16th, 2021, 49 patients have been treated in the phase 1 portion,
of whom 43 started by September 30th, allowing 2 on-study disease assessments. The
median number of prior therapies was 2 (range 1-5). Four monotherapy dose levels of
emavusertib were tested (200 to 500 mg orally BID). No dose-limiting toxicities were
observed at 200 mg and 300 mg BID. No Grade 4 or 5 treatment-related AEs (TRAEs) were
reported, and all the TRAEs were manageable. Reversible, manageable Grade 3 rhabdomyolysis
occurred in 1/26 (4%) patients at 300 mg BID, 2/17 (12%) at 400 mg BID, and 1/3 (33%)
at 500 mg BID. RP2D was determined as 300 mg BID. Of 43 patients starting before Sept
30th, 2021, 14 had SF3B1, U2AF1 or FLT3 mutations and demonstrated more promising
efficacy. In the 5 evaluable AML patients with spliceosome mutations, 40% reached
CR/CRh (1 CR, 1 CRh), both with study duration >6 months. In the 7 spliceosome-mutated
HR-MDS patients, 57% reached marrow CR, including 1 with RBC transfusion independence
and 1 proceeding to HSCT. One of the three FLT3-mutated AML reached CR, and 2 became
FLT3-negative. Among the 29 patients without SF3B1/U2AF1/FLT3 mutations, 1 reached
CR and 2 PR. Phase 1b and Phase 2a are ongoing. RNA-seq on selected samples showed
decrease in relative expression of IRAK4-long isoforms with response to emavusertib.
Summary/Conclusion: Emavusertib is well tolerated and effective in heavily pretreated
AML and HR-MDS patients, especially in those with U2AF1/SF3B1/FLT3 mutations. No dose-limiting
myelosuppression was reported, suggesting emavusertib may be a candidate for combination
therapy. Accrual of Phases 1b and 2a is ongoing.
S130: NPM1 MUTATED AML: IMPACT OF CO-MUTATIONAL PATTERNS - RESULTS OF THE EUROPEAN
HARMONY ALLIANCE
A. Hernández-Sánchez1 2,*, Á. Villaverde-Ramiro2, J. Martínez Elicegui2, T. González2
3, A. Benner4, E. Sträng5, G. Castellani6, C. A. Heckman7 8, J. Versluis9, M. Abáigar2
3, M. Sobas10, R. Azibeiro1 2, L. Tur11, P. J. Valk9, K. H. Metzeler12, R. Ayala13,
D. Dall’Olio6, J. Tettero14, J. Martínez-López13, H. Dombret15, M. Pratcorona16, F.
Damm5, K. I. Mills17, J. Mayer18, C. Thiede19, M. T. Voso20, G. F. Sanz21 22, F. Calado23,
K. Döhner24, V. I. Gaidzik25, M. Heuser26, T. Haferlach27, A. T. Turki28 29, D. Reinhardt28,
R. Villoria Medina11, M. van Speybroeck30, R. Schulze-Rath31, M. Barbus32, J. E. Butler33,
J. M. Hernández Rivas1 2 3, B. J. Huntly34, G. J. Ossenkoppele14 35 36, H. Döhner24,
L. Bullinger5
1Hospital Universitario de Salamanca; 2Instituto de Investigación Biomédica de Salamanca
(IBSAL); 3Centro de Investigación del Cáncer (CIC), Salamanca, Spain; 4German Cancer
Research Center (DKFZ), Heidelberg; 5Charité Universitätsmedizin Berlin, Berlin, Germany;
6University of Bologna, Bologna, Italy; 7University of Helsinki; 8Institute for Molecular
Medicine Finland, Helsinki, Finland; 9Erasmus University Medical Center Cancer Institute,
Rotterdam, Netherlands; 10Wroclaw Medical University, Wroclaw, Poland; 11GMV Innovating
Solutions, Valencia, Spain; 12University of Munich, Munich, Germany; 13Hospital Universitario;
12de Octubre, Madrid, Spain; 14Amsterdam University Medical Center, Amsterdam, Netherlands;
15EA3518 Leukemia Translational Laboratory, Paris, France; 16Hospital de la Santa
Creu i Sant Pau, Barcelona, Spain; 17Queens University Belfast, Belfast, United Kingdom;
18University Hospital Brnoand Masaryk University, Brno, Czechia; 19University of Technics
Dresden Medical Dept., Dresden, Germany; 20University of Rome “Cattolica S. Cuore”,
Rome, Italy; 21Hospital Universitario y Politécnico La Fe; 22Instituto de Salud Carlos
III (CIBERONC), Valencia, Spain; 23Novartis, Oncology Region Europe, Basel, Switzerland;
24University Hospital of Ulm; 25Ulm University Hospital, Ulm; 26Hannover Medical School,
Hannover; 27MLL Munich Leukemia Laboratory, Munich; 28Essen University Hospital; 29West-German
Cancer Center, Essen, Germany; 30Janssen Pharmaceutica N.V., Beerse, Belgium; 31Bayer
Pharma AG, Berlin; 32AbbVie Germany GmbH & Co. KG, Wiesbaden; 33Bayer AG, Berlin,
Germany; 34Wellcome - MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom;
35VU University Medical Center; 36Cancer Center Amsterdam, Amsterdam, Netherlands
Background: Acute myeloid leukemia (AML) is a heterogeneous disease in terms of clinical
features, outcomes and genetics. While mutations of NPM1 are usually considered as
a favorable prognostic marker, the vast majority of the patients carry several co-mutations
that might influence the prognosis. Therefore, a better understanding of the NPM1
mut AML mutational landscape is warranted. The large cohort of AML patients collected
within the European HARMONY Alliance provides an excellent basis for this purpose.
Aims: To identify clinically significant co-mutational patterns in NPM1
mut AML in order to establish a revised risk stratification model.
Methods: From the HARMONY Alliance AML database, a total of 1001 NPM1
mut intensively treated patients were selected. Clinically significant co-mutations
were evaluated using graphical patterns created with the Gephi tool and confirmed
by detailed survival analysis using Kaplan-Meier and Cox regression models. Finally,
a novel multi-state risk stratification model for NPM1
mut AML was established.
Results: The study population of 1001 NPM1
mut AML patients included 57% females and median age was 53 years. Regarding ELN2017
classification, 68% of patients were classified into the favorable, 29% intermediate
and 3% adverse risk groups. The most frequent co-mutations were DNMT3A (54%), followed
by FLT3-ITD (38%). In total, 24% of patients presented with a high allelic mutant-to-wildtype
ratio ≥0.5 (FLT3-ITDhigh) while 14% had low allelic ratio <0.5 (FLT3-ITDlow). Other
frequent co-mutations were NRAS (21%), TET2 (20%) and PTPN11 (15%).
The triple mutation pattern of NPM1
mut + FLT3-ITDhigh + DNMT3A
mut identified a subgroup with adverse prognosis (2-year OS of 25%), similar to NPM1
mut + TP53
mut. The combination of FLT3-ITDlow + DNMT3A
mut or FLT3-ITDhigh + DNMT3A
wt was associated with intermediate prognosis (2-year OS of 45% and 53% respectively).
Notably, mutations of NRAS, KRAS, PTPN11 or RAD21 were identified to be associated
with better OS. However, in the context of NPM1
mut + DNMT3A
mut these mutations did not affect the prognosis when a FLT3-ITD was present. This
information is summarized in a 3-category risk classification model (Figure 1).
The revised NPM1
mut favorable group presented with a 2-year OS of 73%, while for intermediate and
adverse groups the OS was 54% and 27% respectively (p<0.001). Regarding relapse free
survival (RFS), the median was not reached in the favorable group, while it was 23
months for intermediate and 6 months for adverse group (p<0.001). It should be noted
that 171 patients in the NPM1
mut intermediate group would be considered as favorable according to the ELN2017 criteria,
as well as 162 patients in the NPM1
mut adverse group were previously classified as intermediate risk. Therefore, our
model was able to reclassify 33% of NPM1
mut AML patients in comparison to ELN2017 criteria.
Multivariate analysis of OS in NPM1
mut AML identified the following independent prognostic factors: NPM1
mut model (taking favorable group as reference, HR 1.6 for intermediate and HR 2.7
for adverse group, p<0.001); secondary or therapy-related AML (HR 1.8, p<0.001), WBC
at diagnosis >100x103/μL (HR 1.5, p<0.001) and age >60 years (HR 1.4, p<0.001).
Image:
Summary/Conclusion: Analysis of large NPM1
mut AML cohorts allows the discovery of co-mutation patterns associated with prognostic
outcome. In accordance, we propose a new genetic stratification model for NPM1
mut AML that identifies 3 groups with different OS and RFS. This model improves ELN2017
criteria as it is able to correctly reclassify 33% of NPM1
mut AML patients.
S131: CLINICAL IMPLICATIONS OF SECONDARY-AML TYPE MUTATIONS IN PATIENTS WITH DE NOVO
ACUTE MYELOID LEUKEMIA
K. Sun1,*, C.-H. Tsai1, M.-Y. Lo1, M.-H. Tseng1, Y.-Y. Kuo2, M.-C. Liu3, C.-C. Lin1,
C.-L. Cheng1, S.-J. Wu1, C.-Y. Chen1, B.-S. Ko1 4, M. Yao1, W.-C. Chou1, H.-A. Hou1,
H.-F. Tien1 1
1Hematology; 2Oncology; 3Pathology, National Taiwan University Hospital; 4Hematology,
National Taiwan University Cancer center, Taipei, Taiwan
Background: More and more gene mutations have been identified in patients with de
novo acute myeloid leukemia (AML). Among them, a set of gene mutations, including
SRSF2, ZRSR2, SF3B1, U2AF1, ASXL1, EZH2, STAG2, and BCOR mutation, are categorized
as secondary AML (sAML)-type mutations, for their distinct distribution in secondary
AML, compared to primary AML (Lindsley et al, Blood 2015), but the reports regarding
the prognostic impact have been scanty, especially in younger patients.
Aims: In this study, we aimed to explore the clinical significance and prognostic
implication of sAML-type mutations in non-M3 AML patients.
Methods: We consecutively enrolled 921 de novo non-M3 AML patients; 368 were 60 years
or older (older patients) and 553 were younger. Patients with an antecedent history
of hematologic diseases, or therapy-related AML were excluded. sAML-type mutations
were identified by targeted next-generation sequencing of 54 myeloid malignancies
related gene mutations.
Results: A total of 243 (26.4%) patients harbored sAML-type mutations (ST group),
40.2% in older patients and 17.2% in younger ones. Patients in the ST group were significantly
older, had a lower WBC count, peripheral blast count and lactate dehydrogenase level
at diagnosis. sAML-type mutations were negatively correlated with inv(16), monosomy
17, and complex karyotype, but positively associated with 2017 European LeukemiaNet
(ELN)-defined unfavorable-risk genetic category.
Among the patients receiving standard chemotherapy (n=686, 74.5%), the ST group had
a significantly lower CR rate (62.9% vs. 81.5%, P< 0.01), especially among younger
patients (70.8% vs. 86.7%, P<0.01), but only a trend in older patients (49.0% vs.
59.6, P=0.17). With a median follow-up of 4.7 years, patients in the ST group had
a shorter overall survival (OS, median, 2.1 years vs. not reached, P< 0.01) and disease-free
survival (DFS, median, 0.4 years vs. 0.9 years, P< 0.01) than the non-ST group. Subgroup
analyses showed that sAML-type mutations conferred a significantly poorer DFS (median,
0 years vs. 0.5, P=0.03) and a trend of shorter OS (0.8 years vs. 1.1 years, P=0.08)
in older patients, and a significantly worse OS (not reached vs. not reached, P=0.03)
and a trend of shorter DFS (0.7 years vs. 1.0 years, P=0.16) in younger patients.
The numbers of sAML-type mutations had prognostic impacts on both OS (median, 5.8
years vs. 1.5 vs. 1.3 for patients with 0, 1, and ≥2 mutations, respectively, P<0.01)
and DFS (median, 0.9 years vs. 0.6 vs. 0 for those with 0, 1, and ≥2 mutations, respectively,
P< 0.01). Intriguingly, these findings were valid among both the younger and older
patients. Furthermore, the ELN-defined intermediate-risk patients with sAML-type mutations
had similar poor OS and DFS to the ELN unfavorable-risk patients in total cohort (Figure
1), as well as in the older and younger patients. Among the patients with sAML-type
mutations, allogeneic hematopoietic stem cell transplantation did improve their outcome
(median OS 3.0 vs. 0.8 years, P< 0.01).
Image:
Summary/Conclusion: AML patients with sAML-type mutations had distinct clinical features
and poorer outcomes. Incorporating sAML-type mutations can further refine the 2017
ELN risk stratification. It is suggested that AML patients with sAML-type mutations
receive more intensive treatment, such as allo-HSCT, and/or novel therapies.
S132: TOLERABILITY AND EFFICACY OF THE FIRST-IN-CLASS ANTI-CD47 ANTIBODY MAGROLIMAB
COMBINED WITH AZACITIDINE IN FRONTLINE PATIENTS WITH TP53-MUTATED ACUTE MYELOID LEUKEMIA:
PHASE 1B RESULTS
N. G. Daver1,*, P. Vyas2, S. Kambhampati3, M. M. Al Malki4, R. Larson5, A. Asch6,
G. Mannis7, W. Chai-Ho8, T. Tanaka9, T. Bradley10, D. Jeyakumar11, E. Wang12, G. Xing13,
M. Chao13, G. Ramsingh13, C. Renard13, I. Lal13, J. Zeidner14, D. Sallman15
1The University of Texas MD Anderson Cancer Center, Houston, United States of America;
2University of Oxford, Oxford, United Kingdom; 3Healthcare Midwest, Kansas City; 4City
of Hope National Medical Center, Duarte; 5University of Chicago, Chicago; 6University
of Oklahoma, Oklahoma City; 7Stanford University, Stanford; 8University of California
Los Angeles, Los Angeles; 9University of California San Diego, San Diego; 10University
of Miami, Miami; 11University of California Irvin, Irvine; 12Roswell Park Comprehensive
Cancer Center, Buffalo; 13Gilead Sciences, Inc., Foster City; 14Lineberger Comprehensive
Cancer Center, University of North Carolina, Chapel Hill; 15Moffitt Cancer Center,
Tampa, United States of America
Background: Patients (pts) with TP53-mutated acute myeloid leukemia (AML) have a poor
prognosis, with limited responses to currently available therapies and low survival
outcomes, representing a significant unmet medical need. Magrolimab is a monoclonal
antibody blocking CD47, a “don’t eat me” signal overexpressed on cells in cancers
such as AML. This blockade induces phagocytosis of tumor cells and is synergistic
with azacitidine (AZA) via upregulation of “eat me” signals.
Aims: To report tolerability and efficacy data from a phase 1b trial of magrolimab
+ AZA in frontline pts with TP53-mutated AML unsuitable for intensive chemotherapy
(NCT03248479).
Methods: Frontline pts with AML not suitable for intensive chemotherapy received magrolimab
IV starting with a priming dose (1 mg/kg) followed by ramp-up to 30 mg/kg QW or Q2W
as the maintenance dose. AZA 75 mg/m2 was given IV or SC on days 1-7 of each 28-day
cycle. Primary endpoints were safety/tolerability and complete remission (CR) rate
by European LeukemiaNet (ELN) 2017 criteria.
Results: 72 pts with TP53-mutated AML were treated (Table). Common all-grade treatment-emergent
adverse events (TEAEs) were constipation (52.8%), diarrhea (47.2%), febrile neutropenia
(45.8%), nausea (43.1%), fatigue (37.5%), decreased appetite (37.5%), thrombocytopenia
(31.9%), peripheral edema (30.6%), and cough (30.6%). Most common grade ≥3 TEAEs were
febrile neutropenia (37.5%), anemia (29.2%; grade 3, 26.4%; grade 4, 2.8%), thrombocytopenia
(29.2%), pneumonia (26.4%), and neutropenia (20.8%). Grade 3 hemolysis was reported
in 1 pt (1.4%); no grade 4 hemolysis was reported. Objective response rate by intent
to treat was 48.6% (CR, 33.3%; CR with incomplete hematologic recovery [CRi]/CR with
partial hematologic recovery [CRh], 8.3%; morphologic leukemia-free state [MLFS],
1.4%; partial remission, 5.6%). Stable disease and progressive disease (PD) were reported
in 16.7% and 5.6% of pts, respectively; 30- and 60-day mortality rates were 8.3% and
18.1%, respectively. Response assessments were unavailable in an additional 4.2% of
pts who discontinued due to AEs and 6.9% due to other reasons, prior to the cycle
3 day 1 assessment. Median time to CR/CRi was 2.2 months (range, 1.7-7.2 months) and
to CR was 3.0 months (range, 1.8-9.6 months); 14 of 31 (45.2%) evaluable pts with
CR/CRi/CRh/MLFS achieved negative MRD by flow cytometry (investigator reported). Of
24 pts with CR, 8 had a longitudinal TP53 variant allele frequency (VAF) assessment
and 5 of 8 (63%) had VAF decreased to ≤5%. Treatment was stopped due to stem cell
transplant (9 [12.5%]), PD (26 [36.1%]), death (8 [11.1%]), AE (13 [18.1%]), and other
(14 [19.4%]). Median durations of CR and CR/CRi were 7.7 months (95% CI, 4.7-10.9
months) and 8.7 months (95% CI, 5.3-10.9 months), respectively. Median overall survival
(OS) in 72 pts was 10.8 months (95% CI, 6.8-12.8 months) (figure), with median follow-up
of 8.3 months.
Table.
Baseline characteristics
N=72
Age (range), years
73 (31-89)
ECOG, n (%)
0-1
61 (84.7)
2
11 (15.3)
ELN cytogenetic risk, n (%)
Favorable
1 (1.4)
Intermediate
2 (2.8)
Adverse
57 (79.2)
Unknown
12 (16.7)
AML with MDS-related changes, n (%)
34 (47.2)
Therapy-related AML, n (%)
15 (20.8)
ECOG, Eastern Cooperative Oncology Group; MDS, myelodysplastic syndrome.
Image:
Summary/Conclusion: In high-risk frontline pts with TP53-mutated AML unsuitable for
intensive chemotherapy, magrolimab + AZA showed durable responses and encouraging
OS in a single-arm study. A Phase 3 trial of this combination vs standard of care
in TP53-mutated AML (ENHANCE-2; NCT04778397) is ongoing.
S133: OFF-THE-SHELF CD33 CAR-NK CELL THERAPY FOR RELAPSE/REFRACTORY AML: FIRST-IN-HUMAN,
PHASE I TRIAL
R. Huang1,*, Q. Wen1, X. Wang1, H. Yan1, Y. Ma1, M. Wang1, X. Han1, L. Gao1, L. Gao1,
C. Zhang1, X. Zhang1
1Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing,
China
Background: The primary results of CAR-T therapy for patients with R/R AML has shown
limited efficacy and severe side-effect. One of the main challenges is that current
targets for myeloid malignancies are either widely expressed on healthy hemopoietic
stem cells such as CD33 or specific for a group of tumor cells presented as Lewis
Y antigen, which could cause lasting bone marrow depression induced by “on target
off tumor” side-effect or target negative relapse. Therefore, to receive a balance,
we designed a CD33 CAR to recognize AML cells and using NK cells to replace T cells
as the carrier to eliminate tumor cells. The CD33 CAR NK cells have combined the wide-expression
advantage of CD33 target and the safety of NK cells.
Aims: To evaluate the safety and primary efficacy of CD33 CAR NK cells
Methods: 5 qualified subjects with R/R AML aged between 18 and 65 years-old were enrolled
and received round(s) of infusion of anti-CD33 CAR NK cells (6×108, 1.2×109 or 1.8×109
cells per round after the precondition with Fludarabine (30mg/m2) and Cytoxan 300-500mg/m2
for 3 days to 5 days, determined by tumor burden at baseline. We investigated the
response rate at D28 and treatment related side-effect after the CAR NK cell infusion
and the long-term efficacy.
Results: As of data cut (February 26, 2021), 5 pts have finished CAR NK cells infusion.
The median age was 43 (18-65) years-old and the median tumor burden before infusion
is 31% (21%-77.5%). 4 of 5 patients have received MRD negetive CR at day 28 assessment.
In dose group one, three patients have received 3 rounds of CAR NK cells (6×108, 1.2×109
and 1.8×109 cells) with the interval of 7 days after last round, and only patient
1 developed grade 1 CRS represented as fever after the infusion of 1.8×109 CAR NK
cells and alleviated within 24h after symptomatic treatment. Patient 1 and patient
2 received MRD negetive CR at day 14 and 21, but patient 2 relapsed at day 43 after
first round infusion. In dosage group 2, patient 4 and patient 5 both received one
dose of 1.8 ×109 cells CD33 CAR NK cells after precondition, patient 5 developed grade
2 CRS presenting as lasting fever for 6 days after infusion and alleviated after 5mg
Dexamethasone I.V., both patient 4 and patient 5 recieved MRD negetive CR at day 28
after infusion. At the time of this abstract being uploaded, three patients remain
MRD negetive CR.
Image:
Summary/Conclusion: Our primary data of the phase I trial have proved the primary
efficacy and safety of CD33 CAR NK cells for patients with R/R AML. The efficacy needs
expanded samples and longer follow up.
S134: INTRA-PATIENT FUNCTIONAL HETEROGENEITY OF AML DETERMINES FIRST-LINE TREATMENT
RESPONSE
Y. Severin1,*, Y. Festl1, M. Roiss2, T. Benoit3, T. Heinemann1, R. Wegmann1, A.-K.
Kienzler3, M. Bissig3, M. Scharl3, M. Manz3, A. M. Müller3, B. Snijder1
1Institute of Molecular Systems Biology, ETH Zurich; 2Medical clinic of Oncology and
Haematology, University Hospital Zurich; 3Medical clinic of Oncology and Haematology,
University Hospital Zurich, Zürich, Switzerland
Background: Acute myeloid leukemia (AML) is a heterogeneous disease with limited treatment
options and poor long-term survival. AML is characterized by a fast clonal expansion
of premature myeloid cells with highly variable combinations of chromosomal aberrations
and somatic mutations. Patient-to-patient variability is further complicated by the
fact that AML partially recapitulates myeloid maturation with a rare leukemic stem
cell population at the top of the hierarchy. Although eradication of bulk tumor cells
is necessary, matched targeted therapies against the AML stem cell compartment are
likely essential for a long lasting cure of patients. Despite this large tumor heterogeneity
in de-novo AML patients, most patients receive the same standard-of-care treatment
resulting in large differences of treatment success and overall survival. Functional
ex vivo drug-response profiling offers a possible route to tailor individual treatments
for AML patients. However, the intra-patient functional heterogeneity of divergent
AML cell subpopulations, and their contribution to clinical outcome, have not yet
been systematically studied.
Aims: This study has two aims: 1) To analyze the intra-patient functional heterogeneity
of AML blasts, and their contribution to the response to first-line treatment. 2)
To improve the clinical response predictions from functional ex vivo drug response
profiling by explicit analysis of functional tumor heterogeneity.
Methods: In this prospective, non-interventional study, 180 AML patient biopsies were
collected with written informed consent (from 44 patients with newly diagnosed AML
undergoing intensive induction chemotherapy). Patient matched bone marrow and blood
samples were obtained for each donor at three different time points across the course
of the treatment. For each sample, we perform ex vivo drug-response profiling analyzed
by multiplexed immunofluorescence, automated microscopy, and deep learning-based morphological
profiling to determine drug sensitivities on a single-cell level. The drug library
includes 100 distinct drugs and drug combinations, including standard of care induction
chemotherapy, and single-cell drug sensitivity data is analyzed as a function of cellular
clone and blast maturation state. The drug response data is integrated with patient-matched
serum cytokine profiling, RNA-sequencing, and clinical data, allowing to identify
the cellular subpopulations whose functional response is predictive of treatment response.
Results: Our workflow excelled at predicting patient responses to first line treatment,
with an overall predictive power of 85% accuracy and a diagnostic odds ratio of 47.
Molecular and morphology-based phenotyping of cancer subpopulations strengthened clinical
associations and revealed blast maturity as well as immune activation to be strong
predictors of treatment success and the risk of chemotherapy-related infectious complications.
Furthermore, the screening of 100 unique drugs and combinations enabled individualized
ex vivo treatment predictions, suggesting more potent treatment options than those
currently deployed.
Image:
Summary/Conclusion: Maturation-associated intra-tumor drug response heterogeneity
is a strong predictor of clinical response to first-line intensive induction chemotherapy
in AML. Our multiplexed ex vivo drug screening framework is highly flexible, allowing
us to discover, molecularly describe, and target clinically relevant AML subpopulations
on a personal basis, leading to improved personalized treatment recommendations.
S135: CRISPR/CAS9 EDITING REVEALS DEPENDENCE OF HUMAN RICHTER SYNDROME AND MURINE
CLL CELLS ON SIGNALS FROM B CELL RECEPTOR, CXCR4 RECEPTOR AND MACROPHAGES BUT NOT
FROM TOLL-LIKE RECEPTORS IN VIVO
C. Martines1,*, S. Chakraborty1, V. Guastafierro1, M. Vujovikj2, T. Vaisitti3, S.
Deaglio3, L. Laurenti4, A. Dimovski2, D. Efremov1
1Molecular Hematology, International Centre for Genetic Engineering and Biotechnology,
Trieste, Italy; 2Research Center for Genetic Engineering and Biotechnology, Macedonian
Academy of Sciences and Arts, Skopje, North Macedonia, Republic of; 3Functional Genomics
Unit, Department of Medical Sciences, University of Turin, Torino; 4Department of
Hematology, Catholic University Hospital “A. Gemelli”, Rome, Italy
Background: Numerous signals from the microenvironment have been identified that can
increase the survival or induce the proliferation of chronic lymphocytic leukemia
(CLL) cells in vitro. These signals typically represent various secreted or cell surface
ligands that are expressed by different cell types present in the lymph node tumor
microenvironment, such as T cells, macrophages and stromal cells, or molecules that
would be expected to be released by apoptotic cells, such as apoptosis associated
autoantigens or CpG-unmethylated mitochondrial DNA. However, the extent to which these
different signals contribute to the growth and survival of the leukemic cells in vivo
has still not been fully established.
Aims: To determine the relevance of signals from Toll-like receptors (TLRs), the B
cell receptor (BCR) and the chemokine receptor CXCR4 for the growth of murine Eμ-TCL1
CLL cells and human Richter Syndrome (RS) patient-derived xenograft (PDX) cells in
vivo.
Methods: To understand the impact of pharmacological inhibition of the TLR pathway,
immunocompetent C57BL/6 or immunodeficient NSG mice were transplanted with murine
Eμ-TCL1 CLL or human RS-PDX cells, respectively, and were treated with an inhibitor
of the kinase IRAK4 or vehicle control. To determine the effects of genetic disruption
of the TLR, BCR and CXCR4 pathways, the IRAK4, IgM heavy chain (IGHM) and CXCR4 genes
were disrupted by CRISPR/Cas9 editing in the murine Eμ-TCL1 CLL cells or the human
RS-PDX lines RS9737, RS1316 and IP867/17 prior to transplantation. The effects of
genetic disruption of these pathways on the growth of the malignant cells in vivo
were investigated by analyzing the proportion of mutant and wild type alleles in different
anatomical compartments of the transplanted mice.
Results: Treatment with the IRAK4 inhibitor significantly prolonged the survival of
C57BL/6 mice transplanted with murine Eμ-TCL1 CLL cells and significantly delayed
the growth of the human RS-PDX lines RS9737 and RS1316 xenografted in immunodeficient
NSG mice. However, genetic disruption of IRAK4 in the human RS-PDX lines RS9737, RS1316
and IP867/17 or of the TLR adaptor MyD88 in the murine Eμ-TCL1 CLL cells did not result
in negative selection of these cells in vivo, suggesting that these tumors do not
receive or do not rely on TLR signals for their growth and that the therapeutic activity
of the IRAK4 inhibitor is not caused by disruption of TLR signaling in the malignant
cells themselves. In contrast, genetic disruption of the IGHM or CXCR4 gene was associated
with significantly reduced growth of these cells compared to their wild type counterparts
and resulted in almost complete disappearance of the mutated cells at later timepoints
following transplantation. Analysis of the effects of the IRAK4 inhibitor on other
cell types from the tumor microenvironment revealed a significant reduction in the
number of macrophages, which in co-culture experiments were shown to strongly protect
human Richter syndrome and murine Eμ-TCL1 leukemia cells from spontaneous apoptosis.
Summary/Conclusion: These data provide evidence that signals from the BCR, CXCR4 and
macrophages support the growth and survival of CLL and RS cells in vivo and argue
against a role for TLR signals in the pathogenesis of CLL. In addition, they suggest
that targeting the TLR pathway may potentially provide a therapeutic benefit in CLL,
but that this benefit would be derived from inhibition of TLR signaling in monocytes
and macrophages rather than inhibition of TLR signaling in the malignant cells themselves.
S136: CONSTITUTIVE VLA-4 ACTIVATION IN CHRONIC LYMPHOCYTIC LEUKEMIA. THE OTHER SIDE
OF BCR AUTONOMOUS SIGNALING.
E. Tissino1,*, R. Bomben1, P. C. Maity2, F. Pozzo1, G. Forestieri3, A. Nicolò2, A.
Härzschel4, T. Bittolo1, F. M. Rossi1, M. Datta2, F. Zaja5, G. Capasso1, G. D’Arena6,
A. Chiarenza7, F. Di Raimondo7, H. Jumaa2, D. Rossi3, G. Del Poeta8, T. N. Hartmann4,
A. Zucchetto1, V. Gattei1
1Clinical and Experimental Onco-Hematology, Centro di Riferimento Oncologico, Aviano,
Italy; 2Institut für Immunologie, Universitätsklinikum Ulm, Ulm, Germany; 3Division
of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland;
4Department of Internal Medicine, Faculty of Medicine, University Medical Center Freiburg,
University of Freiburg, Freiburg, Germany; 5Dipartimento di Scienze Mediche e Chirurgiche
e della Salute, University of Trieste, Trieste; 6IRCCS Centro Di Riferimento Oncologico
Della Basilicata, IRCCS Centro Di Riferimento Oncologico Della Basilicata, Rionero
In Vulture; 7Divisione di Ematologia, Ospedale Ferrarotto, A.O.U. Policlinico-OVE,
Università di Catania, Catania; 8Division Hematology, S.Eugenio Hospital and University
of Tor Vergata, Rome, Italy
Background: In chronic lymphocytic leukemia (CLL), CD49d, the alpha chain of the heterodimer
CD49d/CD29 (VLA-4), is a strong negative prognosticator and key player of CLL microenvironmental
interactions. The adhesive properties of VLA-4 can be rapidly inside-out activated
by signals through the B-cell receptor (BCR), thus favoring the capability of the
integrin to interact with its ligands. In CLL, beside the canonical antigen (Ag)-dependent
mechanism, BCR signaling has been recently demonstrated to occur via an autonomous
Ag-independent manner.
Aims: To investigate the role of autonomous BCR signaling on constitutive VLA-4 activation
state in CLL.
Methods: VLA-4 activation/affinity was determined by flow cytometry (FC) using the
conformation-sensitive anti-CD29 mAb HUTS21 and/or by “real-time” (FC) measuring the
binding of the VLA-4 ligand LDV-FITC as reported (Tissino et al, J Exp Med, 2018)
in: i) 1,984 consecutive CLL all with IGHV gene mutations/BCR features available;
ii) sequential samples (0, 14, 30, 60 90 days) from CLL patients (n=26) treated in
vivo with ibrutinib (IB) in real-world and from a clinical trial (NCT02827617). HUTS21
staining was also performed in the presence of: plasma depletion/replacement, soluble
(s) VCAM-1, fibronectin (FN) and blocking anti-CD49d (HP1/2) mAbs. ELISA assays were
used to quantify sVCAM-1 in plasma samples (n=122). BCR signaling were investigated
by Ca++ influx assay in a 4-OH tamoxifen (4-OHT)-inducible murine TKO cell model (Dühren-von
Minden M et al, Nature, 2012).
Results: Out of 1,984 CLL, 1,070 (54%) expressed CD49d (cutoff 30%) and, among them,
250/1,070 (23%) were HUTS21+ (cutoff 20%), indicating an activated VLA-4 conformation.
HUTS21 staining was: i) impaired by depletion of plasma from whole blood samples,
and reconstituted by specific plasma components (sVCAM-1, FN); ii) impaired by pre-incubation
with anti-CD49d HP1/2 blocking mAbs before addition of plasma, sVCAM-1 and FN.
sVCAM-1 was higher in CD49d+ vs CD49d- CLL (p<0.0001); among CD49d+ cases, sVCAM-1
was lower in HUTS21+ (i.e. VLA-4 activated) cases (p=0.0096), suggesting ligand sequestration
by activated surface VLA-4.
CLL with mutated IGHV expressed higher levels of activated VLA-4 compared to unmutated
IGHV CLL (p=0.001). Higher levels of activated VLA-4 were found in CLL using the IGHV3
and IGHV4 families, compared to cases using the IGHV1 family (p=0.043 and p=0.004).
Finally, analysis of BCR stereotypy highlighted higher VLA-4 activation levels in
CLL from subset#2 compared to CLL from subset#1 (p=0.02).
To validate these data, murine TKO cells, expressing high VLA-4 levels, were transfected
with different BCRs derived from 4 CLL with high level of constitutively activated
VLA-4 (TKO-high) and 4 CLL with low level of constitutively activated VLA-4 (TKO-low).
Compared to TKO-low cells, TKO-high cells showed a higher autonomous Ca++ influx (p=0.03),
and consistently higher VLA-4 affinity (p=0.01). IB treatment impaired both BCR autonomous
signaling and VLA-4 affinity. Notably, anti-IgM stimulation induced high Ca++ influx
and high VLA-4 affinity state in both TKO-high and TKO-low, irrespective of IB treatment.
According to TKO data, a decreased constitutive VLA-4 activation was observed in CLL
cells collected at pre-treatment and at day 14, 30, 60 and 90 from patients on IB,
confirming an IB-dependent impairment of VLA-4 activation via BCR signal.
Summary/Conclusion: The presence of a constitutively activated form of VLA-4 is observed
in a fraction of CD49d+ CLL, due to a continuous VLA-4 inside-out stimulation derived
from autonomous BCR signaling.
S137: SMALL EXTRACELLULAR VESICLES IN THE LEUKEMIA MICROENVIRONMENT SUSTAIN CLL PROGRESSION
BY HAMPERING T CELL-MEDIATED ANTI-TUMOR IMMUNITY
E. Gargiulo1,*, E. Viry1, P. E. Morande1 2, A. Largeot1, S. Gonder1 3, F. Xian1 4,
N. Ioannou5, M. Benzarti1 3, F. Kleine Borgmann1 3 6, M. Mittelbronn1 3 7, G. Dittmar1
3, P. V. Nazarov1, J. Meiser1, B. Stamatopoulos8, A. G. Ramsay5, E. Moussay1, J. Paggetti1
1Luxembourg Institute of Health, Luxembourg, Luxembourg; 2Instituto de Medicina Experimental,
Buenos Aires, Argentina; 3University of Luxembourg, Luxembourg, Luxembourg; 4University
of Vienna, Vienna, Austria; 5King’s College London, London, United Kingdom; 6Centre
Hospitalier de Luxembourg, Luxembourg; 7Laboratoire national de santé, Dudelange,
Luxembourg; 8Université Libre de Bruxelles, Brussels, Belgium
Background: Small extracellular vesicles (sEV) are nano-sized particles released by
every cell and found in all biofluids. Given their composition and abundance, sEV
are commonly involved in cell-to-cell communication through the transfer of genetic
material and proteins. Furthermore, sEV possess direct functions carried out by sEV-ligands
capable to affect the biological functions of targeted cells. In cancer, tumor-derived
sEV are involved in the re-education of microenvironment (ME) cells promoting tumor
proliferation, immune escape and metastasis. We previously demonstrated that leukemia-derived
sEV are involved in the re-education of surrounding cells and increased immune escape.
Indeed, chronic lymphocytic leukemia (CLL)-derived sEV induce stromal cell conversion
into cancer-associated fibroblasts (Paggetti et al., Blood, 2015), and modulate PD-L1
expression in monocytes (Haderk et al. Science Immunology, 2017).
Aims: The goal of the present work was to characterize leukemia ME-derived sEV (LME-sEV)
and to evaluate their role in the disease development and progression in vivo.
Methods: To obtain a biological representation of sEV in CLL microenvironment, we
isolated LME-sEV directly from spleens of leukemic mice, obtaining a complex mix of
sEV released by both CLL and ME cells alike. Small EV characterization was performed
using a wide range of techniques, including qPCR, mass spectrometry and single sEV
flow cytometry (FC). The effect on target cells was evaluated both ex vivo and in
vivo using high-throughput techniques, FC, qPCR and cytotoxic assay. Small EV impact
on CLL development in vivo was evaluated by generating a novel preclinical mouse model
in which sEV release is genetically impaired due to Rab27a/b knock-out. Finally, we
analyzed the expression of sEV-related genes in a cohort of 144 CLL patients using
qPCR followed by regression analysis.
Results: LME-sEV showed a distinct proteome (A) and RNA contents compared to healthy
counterparts (HCME-sEV), including miRNA enriched in the plasma of CLL patients. Furthermore,
FC-based immune checkpoint (ICP) screening showed the presence of multiple ICP ligands
anchored on CLL-derived sEV (CD20+ subset of LME-sEV) (B), while high expression of
the corresponding ICP receptors was found on T cells from matching LME. We also found
that LME-sEV are internalized by different T cell subsets, thus we performed in vivo
and ex vivo functional studies to assess sEV impact on T cells. High-throughput analysis
of cells isolated from spleens of control mice treated with LME-sEV revealed considerable
physiological changes mainly in CD8+ T cells. Indeed, CD8+ T cells showed alterations
in their transcriptome, proteome and metabolome leading to cell exhaustion, decreased
functions and survival. In line with this, absence of sEV dramatically delayed CLL
progression in vivo. This effect was due to CLL inability to escape immune surveillance
in absence of sEV and this was rescued by LME-sEV treatment (C). Finally, we identified
a consistent sEV gene signatures in CLL patients correlating with treatment-free survival,
overall survival, and with unfavorable clinical parameters routinely used in CLL diagnosis
and prognosis (D).
Image:
Summary/Conclusion: By using different preclinical murine models and strategies, our
results demonstrated for the first time that sEV in CLL ME play a key pro-tumoral
role in leukemia development by negatively affecting the anti-tumor immune response.
Furthermore, high expression of sEV-related genes correlated with poor survival and
clinical parameters in CLL patients, suggesting sEV profiling as prognostic tool in
CLL.
S138: MICROENVIRONMENT-REGULATED TRANSCRIPTIONS FACTORS AHR AND HIF-1Α EXPRESSION
IN TREGS PROMOTE CLL PROGRESSION BY IMPAIRING CD8+ T-CELL MEDIATED ANTI-TUMOR IMMUNITY
G. Pagano1,*, M. Wierz1, I. Fernandez Botana1, S. Gonder1, E. Gargiulo1, E. Moussay1,
J. Paggetti1
1Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
Background: Chronic Lymphocytic Leukemia (CLL) progression is highly dependent on
complex interactions between tumor cells and the tumor microenvironment (TME). Indeed,
CLL cells can modify stromal cells and immune cells to promote the survival of the
leukemic clone and to escape from the immune system surveillance. Within the TME,
regulatory T cells (Tregs) represent a subtype of CD4+ T cells with immunosuppressive
abilities, causing the evasion of cancer cells from the immune system. We previously
characterized extensively the immune microenvironment of pre-clinical CLL mouse models
using mass cytometry, and we described a significant increase in the Tregs subsets
with an enhanced immunosuppressive and activated phenotype compared to non-leukemic
animals (Wierz et al., Blood, 2018). Interestingly, TIGIT+ Tregs are more immunosuppressive
than their TIGIT- Treg counterparts and express higher levels of several transcription
factors, including Ahr and Hif1α (Joller et al., Immunity, 2014), both involved in
the cellular response to microenvironment-mediated stimuli.
Aims: The aim of the present study is to investigate the role of AHR and HIF-1α in
the suppressive ability of Tregs during CLL development.
Methods: We generated conditional knock out mice (cKO) lacking Ahr or Hif1a genes
exclusively in Tregs (Foxp3
YFP-Cre
Ahr
fx/fx
and Foxp3
YFP-Cre
Hif1a
fx/fx
mice). We then performed adoptive transfer (AT) of CLL cells obtained from diseased
Eµ-TCL1 mice into cKO and control mice. In order to decipher the mechanism by which
AHR and HIF-1α pathways in Tregs affect CLL progression, we analyzed the splenic TME
of recipient mice and evaluated immune checkpoint expression and cytokine production.
Finally, we evaluated the suppression ability of the regulatory T cells with ex vivo
suppression assays.
Results: Generated cKO mice showed no sign of abnormalities or autoimmune phenotypes,
and immune cells phenotyping revealed no major differences. However, we showed that
CLL growth in cKO mice was drastically delayed compared to the control mice (A). Interestingly,
this decrease was mitigated when CD8+ T cells were depleted. The analysis of the splenic
TME in recipient cKO mice revealed an increase in IL-17 and TNF-α production, two
major T-cell cytokines, in CD4+ T cells as compared to their WT counterparts. We also
measured the expression of immune checkpoints and activation markers in the different
T cell subpopulations and observed that Tregs lacking AHR or HIF-1α show decreased
levels of the immune checkpoints CTLA-4 and TIGIT, two key proteins for the suppressive
functions of Tregs.
Ex vivo suppression assays demonstrated an increased proliferation of CD8+ T cells
in the presence cKO Tregs (B-C). This result confirmed the decreased suppressive ability
of Tregs in absence of the two transcription factors, explaining the observed delay
in CLL progression in cKO mice.
Image:
Summary/Conclusion: Altogether, these results indicate that the TME-regulated transcription
factors AHR and HIF-1α in Tregs are crucial for CLL development by promoting escape
to anti-tumor immune response, and therefore represent potential therapeutic targets
during CLL progression.
S139: BCOR DELETION SUSTAINS NOTCH1 SIGNALLING ACTIVATION TO ACCELERATE CHRONIC LYMPHOCYTIC
LEUKEMIA (CLL) PROGRESSION TOWARD RICHTER TRANSFORMATION IN MICE
C. Rompietti1,*, D. Sorcini1, F. De Falco1, E. Dorillo1, F. M. Adamo1, E. C. Silva
Barcelos1, A. Stella1, A. Scialdone1, A. Esposito1, R. Arcaleni1, B. Bigerna2, G.
Martino3, L. Moretti2, M. G. Mameli2, C. Geraci1, L. Sandoletti1, A. Cipiciani1, E.
Rosati4, B. Falini1, P. Sportoletti1
1Medicine and Surgery, University of Perugia (Center for Hemato-Oncology Research);
2Hospital of Perugia, Perugia; 3Pathology Unit, Azienda Ospedaliera Santa Maria di
Terni, University of Perugia, Terni; 4Medicine and Surgery, University of Perugia,
Perugia, Italy
Background: BCL6 co-repressor (BCOR) is a transcription factor involved in various
biological processes including lymphoid development. BCOR disruptive mutations were
found in up to 2% of CLL and frequently associated with aberrations of NOTCH1, one
of the most common genetic alterations with poor prognosis in CLL and Richter transformation
(RT). Recent evidence also indicates that BCOR is involved in NOTCH signalling suppression
during embryogenesis. These data indicate the need for further investigation on the
role of BCOR mutation and its interplay with NOTCH1 in CLL pathogenesis.
Aims: We aim to elucidate the impact of Bcor deficiency in CLL and RT, focusing on
the role of active NOTCH1 signalling.
Methods: We used a conditional knockout mouse of Bcor (Sportoletti et al, Leukemia
2021) crossed with CD19-Cre mice, to specifically delete Bcor in B-cells, and with
the Eμ-TCL1 mouse model of CLL. Mice were characterized for disease phenotype (using
flow-cytometry and histological analyses), overall survival and drug response. NOTCH1
activity was assessed by western blot (WB) for the NOTCH1-intracellular domain (NOTCH1-IC)
and HES1 expression.
Results: B-cell restricted loss of Bcor in Eµ-TCL1 mice significantly expanded leukemic
CD5+CD19+ cells in the peripheral blood (PB; p<0.05), spleen (p<0.01), bone marrow
(BM; p<0.001), compared to Eµ-TCL1 control mice. No leukemic cells were found in BcorCD19Cre+
mice. Cellular changes resulted in a poorer survival of double mutant mice compared
to single-mutant and wild-type (WT) controls (median survival of 334 days vs unreached
in BcorCD19Cre+;Eμ-TCL1 vs the other groups, respectively). Adoptive transfer of spleen
cells (CD5+CD19+ leukemic burden >50%) resulted in a more rapidly lethal disease in
recipient of BcorCD19Cre+;Eμ-TCL1 compared to Eμ-TCL1 cells (median survival of 37.5
vs 74 days, respectively, p<0.05; Figure1A). At necropsy, double mutant mice presented
massive splenomegaly, whose histopathological examination revealed a diffuse infiltration
by high-mitotic blastoid cells, significantly increased in size, defining a high-grade
lymphoid malignancy distinct from the leukemia of Eμ-TCL1 counterparts (Fig.1B). In
order to gain mechanistic insight relating to the phenotypic observations, we measured
NOTCH1 activity. BcorCD19Cre+;Eμ-TCL1 splenic CD5+CD19+ cells showed significantly
increased levels of the active NOTCH1-IC and the up-regulation of its direct target
HES1, compared to leukemic cells from Eμ-TCL1 mice and CD19+ cells from non-leukemic
BcorCD19Cre+ and WT mice used as control (Fig.1C). In vivo treatment with the NOTCH1
inhibitor Bepridil resulted in a significant growth delay of CD5+CD19+ cells in the
PB of mice transplanted with BcorCD19Cre+;Eμ-TCL1 leukemic cells compared to vehicle.
At sacrifice, Bepridil caused a significant reduction of spleen dimensions and CD5+CD19+
cellularity, associated with reduced levels of active NOTCH1-IC, c-MYC and HES1 compared
to vehicle (28%, 54% and 38% reduction, respectively). NOTCH1 inhibition significantly
improved the survival of diseased mice (median survival of 39 vs 33 days in Bepridil
vs vehicle, respectively; N=4, p<0.05).
Image:
Summary/Conclusion: We showed for the first time the tumour suppressor activity of
Bcor in a CLL mouse model, ultimately leading to transformation towards a high-grade
lymphoma, mimicking human RT. Mechanistically, we implied NOTCH1 signalling activation
in Bcor loss mediated tumorigenesis with potential for targeted treatment for high-risk
CLL and RT patients.
S140: SINGLE-CELL MULTIOMICS ANALYSES REVEAL COMPLEX INTRA-PATIENT HETEROGENEITY IN
RELAPSED CLL FOLLOWING VENETOCLAX THERAPY
R. Thijssen1 2,*, L. Tian1 2, C. Flensburg1 2, M. A. Anderson1 2 3 4, A. Jarratt1
2, H. Peng1 2, I. Majewski1 2, C. Tam3 4, J. Seymour3 4, P. Blombery3 4, M. Ritchie1
2, D. Huang1 2, A. Roberts1 2 3 4
1The Walter and Eliza Hall Institute of Medical Research; 2Medical Biology, University
of Melbourne; 3Clinical Haematology, Royal Melbourne Hospital and Peter MacCallum
Cancer Centre; 4Medicine, University of Melbourne, Melbourne, Australia
Background: Venetoclax (VEN) is the first approved-in-class BH3-mimetic therapy that
inhibits pro-survival BCL2 to induce apoptosis. It is now a standard of care for patients
with chronic lymphocytic leukaemia (CLL) and acute myeloid leukaemia. Despite high
remission rates, secondary resistance remains problematic with disease progressing
on continuous therapy in most patients. While previous studies by ourselves and others
demonstrated a BCL2 mutation (Blombery et al., 2019 Cancer Disc) or MCL1 amplification
(Guièze et al., 2019 Cancer Cell) confer resistance in patient samples, it is clear
that these only account for a fraction of tumour cells at disease progression and
are not present in all leukaemias. Given that these changes only provide partial explanations,
other mechanisms must operate to subvert the action of VEN.
Aims: To uncover why VEN fails in patients by assessing heterogeneity, distinguishing
cells with known changes (e.g., BCL2 mutation, MCL1 amplification) and to elucidate
what is happening in the cells not harbouring these alterations.
Methods: We applied a single-cell (sc) sequencing method that simultaneously measures
gene expression and full-length transcripts to determine genotype on samples from
patients with progressive CLL with long term follow-up on trials of continuous VEN
therapy (Figure 1).
Results: By applying scRNA-seq to 161,499 tumour cells, we discovered a high degree
of inter- and intra-patient heterogeneity. The majority of CLL cells displayed an
altered transcriptional profile at relapse compared to before VEN treatment. 8/13
VEN-relapse samples demonstrated a complex sub-clonal architecture (Figure 1).
By applying scLong-read sequencing, we detected BCL2 mutations in 4/13 samples at
relapse. We also identified subclones with loss of NOXA or BAX, but overall, alterations
in genes for the pro-apoptotic proteins were uncommon. In contrast, altered expression
of the pro-survival genes was common and high MCL1 expression was seen in most of
the CLL cells in 11/13 relapses. This could only be fully explained by MCL1 amplification
in 1 patient and partially in 2 (Figure 1).
At VEN resistance, we identified near universal activation of NF-ΚB in circulating
tumour cells and this was highly correlated with MCL1 expression. We next demonstrated
that the MCL1 locus was transcriptionally targeted by NF-ΚB accounting for the MCL1
increase. NF-ΚB activation and associated high MCL1 was also observed in cells harbouring
the BCL2 mutations.
Strikingly, NF-ΚB activation (and consequent increased MCL1) dissipated after VEN
cessation and commencement of a BTK inhibitor as the leukaemic cells substantially
recovered their in vitro sensitivity to VEN. Consistent with the hypothesis that VEN
was driving these changes, NF-ΚB activation (and high MCL1) was not detected in a
patient (CLL26) with disease relapse 3 years after ceasing VEN in CR. The disease
in this patient responded promptly to VEN reinduction.
Image:
Summary/Conclusion: Taken together, our findings provide a much clearer understanding
of resistance to VEN and provide impetus for improved VEN-based clinical management
of CLL patients. Given the multiple ways a CLL population in a patient can become
resistant to ongoing VEN, it seems unlikely that simply adding other drugs at relapse
will be durably effective in most patients. The data pinpoint NF-ΚB activation as
a biomarker for in vivo VEN-resistance and provide a specific biological rationale
for ceasing VEN in deep response, as is currently being used in time-limited and explored
in response-directed regimens.
S141: ELICITING ANTI-TUMOR T CELL ACTIVITY IN CHRONIC LYMPHOCYTIC LEUKEMIA WITH BISPECIFIC
ANTIBODY-BASED COMBINATION THERAPY
D. Papazoglou1,*, L. Ysebaert2, N. Ioannou1, B. Apollonio1, P. Patten3, S. Herter4,
M. Bacac4, A. Deutsch5, C. Klein4, A. Vardi6, A. Quillet-Mary2, A. Ramsay1
1Hemato-oncology, King’s College London, London, United Kingdom; 2Centre de Recherches
en Cancérologie de Toulouse, Toulouse, France; 3King’s Health Partners, King’s College
Hospital, London, United Kingdom; 4Roche Innovation Center Zurich, Roche Pharma Research
and Early Development, Zurich, Switzerland; 5Medical University of Graz, Graz, Austria;
6Hematology Department and HCT Unit, G. Papanikolaou Hospital, Thessaloniki, Greece
Background: Identifying effective combination immunotherapy could offer hope to chronic
lymphocytic leukemia (CLL) patients who relapse on previous lines of therapy. Although
prior studies have described an exhausted and pro-tumor T cell state, there may be
potential to stimulate anti-CLL cytolytic T cell activity with novel therapy.
Aims: Investigate the ability of the CD20-targeted T-cell-engaging bispecific antibody
glofitamab (CD20-TCB) to overcome T cell exhaustion and tumor microenvironment (TME)-mediated
immunosuppression in CLL.
Methods: We pre-clinically investigate CD20-TCB efficacy in CLL utilizing immune and
TME model assays that include in vitro functional T cell assays, patient-derived xenografts
and lymph node organotypic models.
Results: Our previous studies of the CLL lymph node (LN) TMEs suggested that low numbers
of infiltrated CD8+ T cells likely contribute to the generation of a “cold” TME that
could represent a barrier to effective therapy. Here, we show that CD20-TCB treatment
induced the activation, proliferation and migration of previously exhausted patient
CD4+ and CD8+T cells and triggered the release of immunostimulatory cytokines related
to immune cell recruitment and cytotoxic function. Additionally, we detected enhanced
cytolytic (perforin+) immune synapse activity in CD20-TCB-treated cultures which correlated
with high levels of T cell-mediated CLL cell death. Importantly, bispecific antibody
treatment also induced high levels of anti-CLL T cell activity in both ibrutinib responding
and refractory patient samples.
Given the emerging evidence that endothelial cells (ECs) that reside within the TME
can possess immunomodulatory activity, we next evaluated CD20-TCB-mediated T cell
responses in the presence of this stromal cell compartment. We established triple
culture assays that allowed patient T and CLL cells to be cultured with CLL-activated
ECs (CLL-ECs). These assays revealed that TCB-activated T cells in the presence of
CLL-ECs showed a reduced ability to kill target CLL cells compared to healthy ECs.
Flow cytometric and multicolor confocal microscopy analysis revealed that CLL-ECs
in vitro and in situ expressed increased levels of ICAM-1 and PD-1 ligands that we
demonstrated contribute to T cell suppression. In order to overcome this TME-mediated
inhibition and enhance anti-CLL CD20-TCB activity, we next investigated combination
immunotherapy. Combination of CD20-TCB with a CD19-4-1BBL fusion protein (tumor-targeted
co-stimulatory drug) resulted in enhanced CLL cell killing in both T:CLL and triple
T:CLL-EC culture assays.
Patient-derived xenograft models further confirmed the ability of bispecific antibody
combination therapy to induce higher levels of anti-CLL T cell activity compared to
monotherapy. Additionally, the pairing of CD20-TCB with CD19-4-1BBL promoted the proliferation
and activation of patient T cells within splenic TMEs. Finally, we established 3D
organotypic cultures of excess diagnostic LN biopsies that preserve the intact TME
cellular composition and spatial organization. LNs of treatment naïve, as well as
ibrutinib and venetoclax relapsed CLL patient samples, were treated ex vivo with CD20-TCB
combination for subsequent 3D image analysis. Importantly, combination immunotherapy
triggered improved anti-tumor T cell killing function and promoted an inflammatory
cytokine-rich “hot” TME.
Summary/Conclusion: Collectively, this preclinical study supports the concept of triggering
cytolytic T cell activity in CLL with bispecific antibody combination immunotherapy
that could help overcome T cell exhaustion and the immunosuppressive TME.
S142: DECIPHERING THE COMPLEXITY OF T CELLS IN BLOOD AND LYMPH NODES OF PATIENTS WITH
CLL BY INTEGRATIVE SINGLE-CELL RNA-SEQ AND MASS CYTOMETRY ANALYSES
L. Llaó Cid1, J. Wong1, M. Wierz2, Y. Paul1, S. Gonder2, I. Fernandez Botana2, T.
Roider3, S. Dietrich3, D. Colomer4, P. Lichter1, M. Zapatka1, E. Moussay2, J. Paggetti2,
M. Seiffert1,*
1Molecular Genetics, B060, German Cancer Research Center, Heidelberg, Germany; 2Department
of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg; 3Department of
Medicine V, University Clinic Heidelberg, Heidelberg, Germany; 4Hematopathology Unit,
Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
Background: Abnormal distribution and impaired function of T cells are key features
of chronic lymphocytic leukemia (CLL), a malignancy of mature B lymphocytes that develops
in secondary lymphoid tissue and blood. These defects have been linked to failure
of immune control and resistance to immunotherapy including immune checkpoint or CAR
T-cell therapy. A better characterization of T cells and their loss of function in
CLL will help to improve such treatment approaches.
Aims: The goal of this study was an in-depth characterization of the T-cell compartment
in blood and tissue samples of patients and mouse models with CLL to gain insights
into the spectrum of phenotypes and transcriptional programs of T cells and the underlying
mechanisms of their development.
Methods: We performed mass cytometry (CyTOF) with a panel of 35 antibodies to characterize
T cells in blood (n=8), bone marrow (n=3), and lymph nodes (n=21) of CLL patients,
as well as reactive lymph nodes of non-tumor patients (n=13). We further generated
single-cell transcriptome and T-cell receptor sequencing data of T cells from lymph
nodes of CLL patients (n=5) and spleen samples of the Eµ-TCL1 mouse model (n=3), and
performed integrative analyses of all data sets.
Results: CyTOF analysis allowed for the identification and quantification of 15 clusters
of CD4+ and 14 clusters of CD8+ T cells in blood, lymph node and bone marrow samples.
T cells in blood and bone marrow were clearly distinct from lymph node derived cells,
and several cell subsets showed a positively correlated abundance. A comparative analysis
revealed an accumulation of several regulatory T-cell subsets, as well as T cells
harboring an exhausted and dysfunctional phenotype in CLL lymph nodes.
Single-cell transcriptome and T-cell receptor sequencing demonstrated the presence
of clonally expanded and gradually exhausted T-cell clusters in human and murine CLL,
and provided insights into the transcriptional programs and regulatory networks of
these cell subtypes. Using CellPhoneDB (Efremova et al., Nature Protocols, 2020) and
CellChat (Jin et al., Nature Communications, 2021) repositories, we identified novel
ligand-receptor-interactions between CLL and T-cell clusters with impact on anti-tumor
immune control which are currently tested in preclinical models for intervention therapy.
Summary/Conclusion: Altogether, our study provides a detailed characterization of
the T-cell compartment in CLL that helps us to understand T-cell exhaustion and suggests
novel targets to improve immunotherapy for patients with CLL and likely also other
malignancies.
MZ, EM, JP and MS share senior authorship.
S143: TRANSCRIPTOMIC CHARACTERIZATION OF MRD RESPONSE AND NON-RESPONSE IN PATIENTS
TREATED WITH FIXED-DURATION VENETOCLAX-OBINUTUZUMAB
O. Al-Sawaf1 2 3,*, H. Y. Jin4, C. Zhang1, Y. Choi4, S. Balasubramanian4, S. Robrecht1,
A. Kotak5, N. Chang5, A.-M. Fink1, E. Tausch6, C. Schneider6, M. Ritgen7, K.-A. Kreuzer1,
B. Chyla8, J. Paulson4, B. Eichhorst1, S. Stilgenbauer6, Y. Jiang4, M. Hallek1, K.
Fischer1
1Department I of Internal Medicine and Center of Integrated Oncology Aachen Cologne
Bonn Duesseldorf, University Hospital of Cologne, Cologne, Germany; 2Cancer Institute,
University College London; 3Francis Crick Institute, London, United Kingdom; 4Genentech
Inc., South San Francisco, CA, United States of America; 5Roche Products Ltd, Welwyn
Garden City, United Kingdom; 6Department III of Internal Medicine, Ulm University,
Ulm; 7Department II of Internal Medicine, University of Schleswig Holstein, Kiel,
Germany; 8AbbVie Inc., North Chicago, IL, United States of America
Background: Minimal residual disease (MRD) is a key surrogate for the depth of remission
of CLL. The highest rates of undetectable MRD have been observed with regimens using
the BH3 mimetic venetoclax. In CLL14, most patients (pts) receiving venetoclax-obinutuzumab
(Ven-Obi) had undetectable MRD (uMRD <10-4) at the end of treatment (EoT). However,
a subgroup of pts remained MRD positive, regardless of TP53 aberrations or unmutated
IGHV status. The biological drivers of response or non-response have so far not been
elucidated.
Aims: This study explores whether differential transcriptomic profiles can discriminate
between pts with vs without deep MRD remissions, particularly after Ven exposure.
This might eventually allow for biology-informed treatment development for pts in
this high-risk disease setting.
Methods: NGS-based MRD measurements (clonoSEQ) from peripheral blood (PB) at follow-up
month 3 were grouped into uMRD <10-6 (MRD6), 10-6 ≤ and <10-4 (MRD4/5), and detectable
MRD ≥10-4 (MRD+). Pre-treatment CD19-enriched PB samples, and samples collected at
relapse from a subset of pts, were subjected to bulk RNAseq on the Illumina NovaSeq
platform. Transcriptomic clustering, linear modeling, differential expression (DE),
and gene set enrichment analyses were run using the R packages Seurat, DESeq2, fGSEA,
and GSVA. Top DE genes were selected based on P-values <0.05 and log2-fold-change
>0.5, comparing MRD+ and MRD6 populations.
Results: Within the intention-to-treat (ITT) population after a median observation
time of 65.4 months, pts with MRD6 had a significantly longer PFS than pts who had
MRD4/MRD5 (A). Pre-treatment RNAseq data were available for 405 of the 432 pts in
the ITT population (202 Clb-Obi, 203 Ven-Obi), and at relapse for 41 pts (14 Clb-Obi
and 27 Ven-Obi). The pre-treatment transcriptomic profile correlated with the IGHV
status, trisomy 12, and del13q (B). No clustering according to MRD status was observed,
suggesting that individual pathways/genes rather than global differences are associated
with MRD response. Individual gene-level analysis of pre-treatment cohorts showed
that MRD+ status was associated with resistance markers such as multi-drug response
(MDR) gene or ABCB1, but MRD6 was associated with pro-apoptotic BCL2L11 (BIM) (C).
Hallmark apoptotic pathways (P53 and Apoptosis), as well as canonical oncogenic pathways
(MYC, mTORC1, TNFɑ/NFκB), were enriched in MRD6 pts in both Ven-Obi and Clb-Obi-treated
pts (D). In contrast, inflammatory pathway gene sets (Inflammatory response, IFNγ
response, IL2/STAT5) were enriched in MRD+ pts in the Ven-Obi arm, suggesting their
role in Ven resistance. Gene sets enriched at progression compared to baseline were
mostly related to cell proliferation and oncogenic signaling (E). Moreover, in a GSVA
analysis, inflammatory pathway gene sets were also markedly upregulated at progression
compared to baseline, providing an orthogonal validation that inflammatory signaling
might be associated with resistance to venetoclax therapy (Fig F). Common leading
edge genes included IL2RB, LCP2, BST2, IRF7, CASP3, CD86, IRF8, and NCOA3.
Image:
Summary/Conclusion: This analysis of a large, prospective CLL RNAseq dataset confirms
that global transcriptomic profiles cluster according to IGHV, trisomy 12 and del13q.
Response and non-response to therapy, as assessed by NGS-based MRD status, was associated
with a distinct transcriptomic profile that was characterized by upregulated oncogenic
pathways and inflammatory signaling, respectively. These data suggest possible biological
vulnerabilities that could be leveraged to overcome resistance to venetoclax.
S144: IMMUNE RESTORATION AND SYNERGISTIC ACTIVITY WITH FIRST-LINE (1L) IBRUTINIB (IBR)
PLUS VENETOCLAX (VEN): TRANSLATIONAL ANALYSES OF CAPTIVATE PATIENTS WITH CLL
I. Solman1,*, R. Singh Mali2, L. Scarfo3, M. Choi4, C. Moreno5 6, A. Grigg7, J. P.
Dean1, E. Szafer-Glusman1
1Pharmacyclics LLC, an AbbVie Company, South San Francisco, CA; 2AbbVie, North Chicago,
IL, United States of America; 3Ospedale San Raffaele, Milan, Italy; 4University of
California San Diego, San Diego, CA, United States of America; 5Hospital de la Santa
Creu I Sant Pau, Autonomous University of Barcelona; 6Josep Carreras Leukaemia Research
Institute, Barcelona, Spain; 7Austin Hospital, Heidelberg, VIC, Australia
Background: Ibr and Ven work synergistically through their distinct and complementary
modes of action: BTK inhibition by Ibr decreases levels of anti-apoptotic MCL-1 and
BCL-XL proteins, but not BCL-2, and therefore increases sensitization to BCL-2 inhibition
by Ven in vivo. Single agent Ibr has been shown to normalize CLL-associated immune
cell alterations in number and function but the impact of the Ibr plus Ven combination
(I+V) on immune cells has not been evaluated.
Aims: To 1) confirm BCL-2 sensitization by single-agent Ibr and 2) monitor changes
in the cellular immune profile of patients (pts) treated with I+V in CAPTIVATE (NCT02910583),
a multicenter phase 2 study evaluating 1L I+V treatment (tx) in CLL and SLL.
Methods: Ibr (420mg po daily) effects on anti-apoptotic proteins were evaluated by
flow cytometry with samples from 4 previously untreated pts with CLL treated for 1
cycle (28 days). Immune restoration was evaluated in 79 previously untreated pts with
CLL enrolled in the CAPTIVATE Minimal Residual Disease (MRD) cohort. Pts received
3 cycles of Ibr, followed by 12 cycles of I+V (Ibr 420 mg/d orally; Ven ramp-up to
400 mg/d orally). Pts who met criteria for confirmed undetectable MRD (Confirmed uMRD)
were then randomized 1:1 to placebo fixed-duration (FD) tx or Ibr at cycle 16; pts
who did not meet Confirmed uMRD criteria (uMRD Not Confirmed) were randomized 1:1
to Ibr or I+V. Cryopreserved peripheral blood mononuclear cells at baseline (pre-dose
cycle 1) and day 1 of cycles 4, 7, 16, 20, 23, and 29 in each of the 4 arms were analyzed
by high-dimensional flow cytometry (34-color panel). Immune cell subset counts of
B cells, T cells, monocytes, dendritic cells (DCs), myeloid-derived suppressor cells,
natural killer cells, and innate lymphoid cells were calculated and compared to those
of 20 age-matched healthy donors. Median changes from baseline are reported.
Results: In pts treated with single-agent Ibr for 1 cycle, MCL-1, BCL-XL and BCL-2
expression decreased by 74%, 95% and 10%, respectively, in lymph node emigrant (CD5+
CXCR4dim) CLL cells, confirming increased BCL-2-dependence and corresponding inhibitor
sensitization. In CAPTIVATE pts treated with I+V, a rapid and significant decrease
in circulating CLL cells was observed within the first 3 cycles of Ven tx initiation
(Fig 1A, cycle 7). From cycle 16 and onwards, Confirmed uMRD pts had CLL cell counts
similar to healthy donors (≤0.8 CLL cell/mL), while in uMRD Not Confirmed pts counts
were 1.5 to 22.9 CLL cell/mL. Subsequently, normal B-cell count restoration occurred
in Confirmed uMRD pts randomized to placebo (FD tx) with a recovery to levels similar
to healthy donors (+332% at cycle 29; Fig 1B). Across all arms, abnormal counts of
T cell subsets, classical monocytes and conventional DCs characteristic of CLL were
normalized to healthy donor levels (‒49%, +101% and +91% respectively) within the
first 6 months of tx and were maintained thereafter regardless of randomized tx. Plasmacytoid
DCs counts recovered by cycle 20 (+598%) in all 4 arms.
Image:
Summary/Conclusion: Sensitization to BCL-2 inhibitor effects by Ibr supports the synergistic
activity of I+V. The FD regimen (Confirmed uMRD, placebo arm) of I+V effectively eradicated
CLL cells to healthy donor levels and enabled sustained regeneration of normal B cell
counts in contrast to ongoing therapy in the remaining tx arms. Combination I+V also
allowed for normalization of other critical immune cells, including T cell subsets,
classical monocytes, and conventional and plasmacytoid DCs. These data demonstrate
promising evidence of immune restoration with FD I+V tx.
S145: THE COMBINATION OF IBRUTINIB PLUS VENETOCLAX RESULTS IN A HIGH RATE OF MRD NEGATIVITY
IN PREVIOUSLY UNTREATED CLL: THE RESULTS OF THE PLANNED INTERIM ANALYSIS OF THE PHASE
III NCRI FLAIR TRIAL
P. Hillmen1,*, A. Pitchford2, A. Bloor3, A. Pettitt4, P. Patten5, F. Forconi6, A.
Schuh7, C. Fox8, N. Elmusharaf9, S. Gatto10, B. Kennedy11, J. Gribben12, N. Pemberton13,
O. Sheehy14, G. Preston15, D. Howard16, A. Hockaday2, D. Cairns2, S. Jackson2, N.
Greatorex2, N. Webster17, S. Dalal17, J. Shingles17, K. Cwynarski18, S. Paneesha19,
D. Allsup20, A. Rawstron17, T. Munir21
1St James’s University Hospital; 2Leeds Cancer Research UK Clinical Trials Unit, Leeds
Institute of Clinical Trials Research, Leeds; 3University of Manchester, Manchester;
4Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool; 5King’s College Hospital,
London; 6University of Southampton, Southampton; 7Oxford University Hospital NHT Trust,
Oxford; 8Nottingham University Hospitals NHS Trust, Nottingham; 9Wales Teaching Hospital;
10University Hospital of Wales, Cardiff; 11Leicester Royal Infirmary, Leicester; 12St
Bartholomew’s Hospital, London; 13Worcestershire Acute Hospitals NHS Trust, Worcester;
14Belfast City Hospital, Belfast; 15Aberdeen Royal Infirmary, Aberdeen; 16Roche, Reading;
17HMDS, Leeds Cancer Centre, Leeds; 18University College London, London; 19Birmingham
Heartlands Hospital, Birmingham; 20Castle Hill Hospital, Hull; 21UK NCRI CLL sub-group,
Leeds, United Kingdom
Background: Ibrutinib (I) and venetoclax (V) improve outcome in CLL. I rarely eradicates
measurable residual disease (MRD), whereas V (alone or with anti-CD20) can eradicate
MRD permitting time limited therapy. Small studies suggest synergy between I and V,
as I+V results in MRD negativity in many patients (pts).
Aims: The primary aim was to compare the MRD eradication rate between I and I+V. Key
secondary aims were IWCLL overall (ORR), complete response (CR) and safety.
Methods: FLAIR (ISRCTN01844152), a phase III, randomised, controlled trial for previously
untreated CLL requiring therapy by IWCLL criteria. Pts >75 yrs or with >20% 17p del
were excluded. FLAIR was adapted in July 2017 to add two arms, I monotherapy and I+V.
I was given at 420mg/day. For I+V, V was added after two months of I with dose escalation
to 400mg/day over 5 weeks. The duration of therapy (DOT) was defined by MRD with treatment
for up to 6 years. MRD was assessed centrally by flow cytometry, MRD negativity was
defined as <1 CLL cell in 10,000 leucocytes (IWCLL criteria), was assessed in peripheral
blood (PB) and bone marrow (BM) at 9 months post-randomisation, in PB at 12 months
and then 6 monthly. When PB was MRD negative, this was repeated after 3 months and
then in both PB & BM 3 months later. If PB & BM were negative the time to MRD negativity
was calculated (treatment start to first MRD negative PB) and DOT was twice this.
The earliest therapy could stop was 2 years post-randomisation. A formal interim analysis
was performed when 50% pts in I and I+V arms had reached 2 yrs post-randomisation
and a p-value of <0.005 was statistically significant.
Results: 523 pts were randomised to I or I+V. We report the interim analysis in the
first 274 pts (I [n=138] and I+V [n=136]) reaching 2 yrs post-randomisation from 83
UK Centres from 13/07/17 to 15/03/19. 72.3% male, median age 63 yrs (34.3% >65yo)
and 40.9% Binet C. IGHV were available for 256 (93.4%) pts - 48.2% IGHV unmutated
(≥98% homology to germline), 45.3% IGHV mutated and 9.1% Subset 2. Hierarchical FISH
testing revealed 16.1% 11q del, 19% trisomy 12, 21.9% normal and 36.9% 13q del with
6.2% failed. The arms were well-balanced for all variables. For I+V arm, MRD negativity
was achieved within 24 months in BM in 89/136 (65.4%) and PB in 97/136 (71.3%) compared
to no pts for I (p<0.0001). MRD negativity for I+V in BM within 24 months was 51/64
(79.7%) for IGHV unmutated and 31/55 (56.4%) for IGHV mutated. At 9 months post-randomisation
49/136 (36%) I+V pts were MRD negative in BM and 56/136 (41.2%) negative in PB compared
to 0/138 with I (p<0.0001). ORR at 9 months in 120/136 (88.2%) I+V pts and 119/138
(86.2%) I pts (p=0.6157). At 9 months CR in 81/136 (59.6%) for I+V and 11/138 (8%)
for I (p<0.0001). For I+V CR at any time was 93.4%. At 24 months, 54/136 (39.7%) stopped
I+V due to meeting MRD stopping criteria.
SAEs were reported in 41.5% I+V and 38.2% I pts. Infectious SAEs 14.8% vs 19.9% and
cardiac SAEs 11.9% vs 8.1% of pts for I+V & I respectively. Laboratory TLS was reported
in 6/136 (4.4%) of I+V pts and none with I. There were no cases of clinical TLS. Most
frequent any grade AEs within 12 months of randomisation differing between I+V & I
were diarrhoea (52.6% I+V pts, 29.4% I), anaemia (28.9% vs 16.9%), leucopenia (36.3%
vs 8.8%), thrombocytopenia (23.7% vs 14%). Leucopenia was the only grade ≥3 SAE reported
in >10% of pts (27.4% I+V and 5.1% I).
Image:
Summary/Conclusion: Ibrutinib plus venetoclax is an effective and well tolerated combination
resulting in a high rate of MRD negativity in blood (71.9%) and marrow (65.4%) in
the first 2 years of treatment.
S146: VENETOCLAX IN PATIENTS WITH CHRONIC LYMPHOCYTIC LEUKEMIA WITH 17P DELETION:
6-YEAR FOLLOW-UP AND GENOMIC ANALYSES IN A PIVOTAL PHASE 2 TRIAL
S. Stilgenbauer1,*, E. Tausch1, A. W. Roberts2, M. S. Davids3, B. Eichhorst4, M. Hallek4,
P. Hillmen5, C. Schneider1, S. Böttcher6, R. Popovic7, M. T. Ghanim7, M. Moran7, W.
J. Sinai7, X. Wang7, N. Mukherjee7, B. Chyla7, W. G. Wierda8, J. F. Seymour2
1Division of CLL, Internal Medicine III, Ulm University, Ulm, Germany; 2Peter MacCallum
Cancer Centre, Royal Melbourne Hospital, and University of Melbourne, Melbourne, Australia;
3Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United
States of America; 4Department of Internal Medicine, Center of Integrated Oncology
Köln Bonn, University Hospital of Cologne, Cologne, Germany; 5Leeds Teaching Hospitals,
NHS Trust, Leeds, United Kingdom; 6Division of Internal Medicine, Medical Clinic III-Hematology,
Oncology and Palliative Medicine, Rostock University Medical Center, Rostock, Germany;
7AbbVie Inc, North Chicago, IL; 8The University of Texas MD Anderson Cancer Center,
Houston, TX, United States of America
Background: Patients (pts) with chronic lymphocytic leukemia (CLL) with del(17p) and/or
mutated TP53 have adverse prognosis and are a population of interest for improving
outcomes. In a pivotal Phase 2 study (NCT01889186) evaluating venetoclax (Ven; a selective
oral BCL2 inhibitor) monotherapy in del(17p) CLL (N=158), overall response rate (ORR)
was 77% (median time on study, 26.6 mo; Stilgenbauer. J Clin Oncol. 2018;17:1973).
Aims: To report final analyses after 5 y from last pt enrolled, including post-hoc
subgroup analyses of prognostic markers.
Methods: Adults with R/R or previously untreated del(17p) CLL received Ven 400 mg
(via ramp-up) orally daily until progressive disease (PD) or intolerance. Main cohort
comprised pts with R/R CLL; safety expansion cohort included pts with R/R or untreated
CLL. Primary endpoints were ORR (main) and safety (safety expansion). Peripheral blood
(PB) and bone marrow (BM) were analyzed pre-Ven for somatic mutations via targeted
next-generation sequencing (NGS). Minimal residual disease (MRD) was assessed by flow
cytometry, ASO-PCR, and/or clonoSEQ NGS; undetectable MRD (uMRD) is reported at <10−4.
Results: In all, 158 pts received Ven (main, n=107; safety expansion, n=51). Data
cutoff was 15 December 2020. Pts had median 2 (range, 0−10) prior lines of therapy
(LOT); 5 had untreated CLL (first-line [1L]). Common mutations (n/N) were TP53 (113/138
[82%]; 35/113 [31%] had >1), SF3B1 (28/137 [20%]) NOTCH1 (22/137 [16%]), ATM (11/120
[9%]), and BIRC3 (6/120 [5%]); 93/115 (81%) had unmutated IGHV and 118/158 (75%) had
≥20% del(17p). Median time on Ven was 27.4 (range, 0–79.3) mo. At study close, 77
pts were alive; 26 remained on Ven. No new safety signals were observed.
With 70 mo median follow-up (f/u), investigator-assessed ORR was 77% (95% CI, 70−84);
21% achieved complete remission (CR)/CR with incomplete blood count recovery (CRi).
Median duration of response was 39.3 mo (95% CI, 31.1−50.5); 28% had ongoing response
at 60 mo. Of 61 evaluable pts with a PB MRD assessment, 25% had uMRD at ~2 y (24−30
mo). Median progression-free survival (mPFS) was 28.2 mo (95% CI, 23.4−37.6; Figure).
Median overall survival (mOS) was 62.5 mo (95% CI, 51.7−NR; 5-y PFS and OS rates,
24% and 52%). Of 5 1L pts, 4 were alive and progression-free. In pts with CR/CRi (n=37),
mPFS was 62.2 mo (95% CI, 53.1−NR); in pts with partial remission (PR)/nodular PR
(n=85), mPFS was 27.6 mo (95% CI, 22.8−38.0). Overall, 98/158 (62%) pts had PD, including
24/158 (15%) with Richter transformation. Of pts with PD (n=98) or whose disease was
refractory to Ven (n=11), 73 received another LOT, most commonly ibrutinib (n=41);
mOS from ibrutinib initiation was 28.0 mo.
In pts with both del(17p) and TP53 mutation (n=111) vs those with either (n=19), ORR
was 75% vs 79%; mPFS was 27.4 vs 22.8 mo (P=.8). In pts with mutated (n=22) vs unmutated
IGHV (n=93), mPFS was 40.4 vs 26.9 mo (P=.11). No significant difference in ORR, PFS,
or OS was observed in pts with vs without ≥20% del(17p), >1 TP53 mutation, NOTCH1,
ATM, or BIRC3 mutations; mPFS was shorter in pts with mutated SF3B1 (n=28) vs without
(n=109; 16.4 vs 30.2 mo [P=.0071]). Multivariate analysis is ongoing.
Image:
Summary/Conclusion: At end of study (median f/u, 70 mo), 48% of pts were alive, 24%
were progression-free, and 16% remained on Ven, confirming the long-term activity
of Ven in this high-risk population with del(17p) CLL and median 2 prior LOT. Except
SF3B1 mutation, other adverse features (eg, >1 TP53 mutation, NOTCH1 mutations, unmutated
IGHV) did not influence outcomes with Ven treatment in this cohort.
S147: PIRTOBRUTINIB, A HIGHLY SELECTIVE, NON-COVALENT (REVERSIBLE) BTK INHIBITOR IN
PREVIOUSLY TREATED CLL/SLL: UPDATED RESULTS FROM THE PHASE 1/2 BRUIN STUDY
A. R. Mato1,*, J. M. Pagel2, C. C. Coombs3, N. N. Shah4, N. Lamanna5, T. Munir6, E.
Lech-Maranda7, T. A. Eyre8, J. A. Woyach9, W. G. Wierda10, C. Y. Cheah11, J. B. Cohen12,
L. E. Roeker1, M. R. Patel13, B. Fakhri14, M. A. Barve15, C. Tam16, D. Lewis17, J.
N. Gerson18, A. J. Alencar19, C. Ujjani20, I. Flinn21, S. Sundaram22, S. Ma23, D.
Jagadeesh24, J. Rhodes25, J. Taylor19, O. Abdel-Wahab1, P. Ghia26, S. J. Schuster18,
D. Wang2, B. Nair2, E. Zhu2, D. E. Tsai2, M. S. Davids27, J. R. Brown27, W. Jurczak28
1Memorial Sloan Kettering Cancer Center, New York; 2Loxo Oncology at Lilly, Stamford;
3University of North Carolina at Chapel Hill, Chapel Hill; 4Medical College of Wisconsin,
Milwaukee; 5Herbert Irving Comprehensive Cancer Center, Columbia University, New York,
United States of America; 6Haematology, St. James’s University Hospital, Leeds, United
Kingdom; 7Institute of Hematology and Transfusion Medicine, Warsaw, Poland; 8Oxford
University Hospitals NHS Foundation Trust, Churchill Cancer Center, Oxford, United
Kingdom; 9The Ohio State University Comprehensive Cancer Center, Columbus; 10MD Anderson
Cancer Center, Houston, United States of America; 11Linear Clinical Research and Sir
Charles Gairdner Hospital, Perth, Australia; 12Winship Cancer Institute, Emory University,
Atlanta; 13Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota; 14University
of California San Francisco, San Francisco; 15Mary Crowley Cancer Research, Dallas,
United States of America; 16Peter MacCallum Cancer Center, Royal Melbourne Hospital,
and University of Melbourne, Melbourne, Australia; 17Plymouth Hospitals NHS Trust
- Derriford Hospital, Plymouth, United Kingdom; 18Lymphoma Program, Abramson Cancer
Center, University of Pennsylvania, Philadelphia; 19University of Miami Miller School
of Medicine, Miami; 20Fred Hutchinson Cancer Research Center, Seattle; 21Sarah Cannon
Research Institute, Nashville; 22Hematology and Medical Oncology, Tisch Cancer Institute,
Icahn School of Medicine at Mount Sinai, New York; 23Robert H. Lurie Comprehensive
Cancer Center of Northwestern University, Chicago; 24Cleveland Clinic, Cleveland;
25Northwell Health Cancer Institute, Donald and Barbara Zucker School of Medicine
at Hofstra/Northwell Health, New Hyde Park, United States of America; 26Università
Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy; 27Dana-Farber
Cancer Institute and Harvard Medical School, Boston, United States of America; 28Maria
Sklodowska-Curie National Research Institute of Oncology, Krakow, Poland
Background: Covalent BTK inhibitors (BTKi) have transformed the management of chronic
lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), but many patients (pts)
will require additional treatment. Pirtobrutinib is a highly selective, non-covalent
(reversible) BTKi that inhibits both wild type (WT) and C481-mutated BTK with equal
low nM potency.
Aims: BRUIN is a phase 1/2 multicenter study (NCT03740529) of oral pirtobrutinib monotherapy
in pts with advanced B-cell malignancies who have received ≥2 prior therapies.
Methods: Pirtobrutinib was dose escalated in a standard 3 + 3 design in 28-day cycles.
The primary objective for phase 1 was to determine the recommended phase 2 dose (RP2D)
and the primary objective of phase 2 was overall response rate (ORR); secondary objectives
included duration of response, progression-free survival, overall survival, safety
and tolerability and pharmacokinetics. Response was assessed every 8 weeks from cycle
3, and every 12 weeks from cycle 13 and was measured according to the iwCLL 2018 criteria,
including PR with lymphocytosis (PR-L). Safety was assessed in all pts.
Results: As of 27 September 2020, 323 pts with B-cell malignancies (170 CLL/SLL, 61
MCL, 26 WM, 26 DLBCL, 13 MZL, 12 FL, 9 RT, and 6 other) were treated on 7 dose levels
(25-300mg QD). Among the 170 pts with CLL/SLL, the median age was 69 (36-88) years.
Median number of prior lines of therapies was 3 (1-11). Majority of the CLL/SLL pts
had received a prior BTKi (86%), an anti-CD20 antibody (90%), or a chemotherapy (82%).
High risk molecular features such as 17p deletion, TP53 mutation, and unmutated IGHV
were present in 25% (20/81), 30% (27/91), and 88% (71/81) of pts, respectively. No
dose-limiting toxicities (DLTs) were reported and maximum tolerated dose (MTD) was
not reached (n=323). 200mg QD was selected as the RP2D. Fatigue (20%), diarrhea (17%)
and contusion (13%) were the most frequent treatment-emergent adverse events (TEAEs)
regardless of attribution or grade seen in ≥10% of pts (n=323). The most common adverse
event of grade ≥3 was neutropenia (10%). Treatment-related hemorrhage and hypertension
occurred in 5 (2%) and 4 (1%) pts, respectively. 1% pts discontinued due to TEAEs.
139 CLL/SLL pts were efficacy-evaluable with a median follow up time of 6 months (0.16-17.8+).
The ORR was 63% (95% CI 55-71) among the 139 efficacy evaluable pts with 69 PRs (50%),
19 PR-Ls (14%), 45 SDs (32%), and 1 PD (1%), and 5 (4%) discontinued prior to first
response assessment. Among the 121 BTKi pretreated pts, the ORR was 62% (95% CI 53-71).
Responses deepened over time with an ORR of 86% among the pts with at least 10 months
follow-up. ORR was similar in pts who discontinued prior BTKi due to progression (67%),
or adverse events or other reasons (52%). Of the 88 responding pts, all except 5 remained
on therapy (4 progressed and 1 achieved a PR and electively discontinued treatment
to undergo transplant). The longest-followed responding patient had been on treatment
for 17.8+ months.
Summary/Conclusion: Pirtobrutinib demonstrated promising efficacy in heavily pretreated
CLL/SLL pts following multiple prior lines of therapy and in pts with BTK C481 mutations.
Pirtobrutinib was well tolerated and exhibited a wide therapeutic index. Updated data,
including approximately 100 new pts with CLL and an additional 10 months since the
prior data cut will be presented.
S148: VENETOCLAX-OBINUTUZUMAB FOR PREVIOUSLY UNTREATED CHRONIC LYMPHOCYTIC LEUKEMIA:
5-YEAR RESULTS OF THE RANDOMIZED CLL14 STUDY
O. Al-Sawaf1 2 3,*, C. Zhang1, S. Robrecht1, A. Kotak4, N. Chang4, A.-M. Fink1, E.
Tausch5, C. Schneider5, M. Ritgen6, K.-A. Kreuzer1, B. Chyla7, B. Eichhorst1, Y. Jiang8,
S. Stilgenbauer5, M. Hallek1, K. Fischer1
1Department I of Internal Medicine and Center of Integrated Oncology Aachen Cologne
Bonn Duesseldorf, University Hospital of Cologne, Cologne, Germany; 2Cancer Institute,
University College London; 3Francis Crick Institute, London; 4Roche Products Ltd,
Welwyn Garden City, United Kingdom; 5Department III of Internal Medicine, Ulm University,
Ulm; 6Department II of Internal Medicine, University of Schleswig Holstein, Kiel,
Germany; 7AbbVie Inc., North Chicago, IL; 8Genentech Inc., South San Francisco, CA,
United States of America
Background: One-year fixed-duration venetoclax-obinutuzumab (Ven-Obi) has demonstrated
significant improvement of progression-free survival (PFS) as compared to chlorambucil-obinutuzumab
(Clb-Obi) in patients with previously untreated chronic lymphocytic leukemia (CLL)
and coexisting conditions in the CLL14 trial. As high rates of undetectable minimal
residual disease (uMRD) suggested deep remissions, long-term efficacy data including
patients with high-risk disease is of particular interest.
Aims: The aim of this report is to provide updated efficacy and safety data from the
ongoing follow-up of the CLL14 study with all patients being off study treatment for
≥ 4 years.
Methods: Patients with previously untreated CLL and coexisting conditions were randomized
1:1 to 12 cycles of venetoclax with 6 cycles of obinutuzumab or 12 cycles of chlorambucil
with 6 cycles of obinutuzumab. The primary endpoint was investigator-assessed PFS.
Secondary endpoints included safety, rates of MRD response (measured every 3-6 months
up to 9 years after last patient enrolment), time to next treatment (TTNT) and overall
survival. Follow-up is ongoing, all patients are off study treatment.
Results: Of the 432 enrolled patients, 216 were randomly assigned to receive Ven-Obi
and 216 to receive Clb-Obi. With a current median follow-up of 65.4 months (interquartile
range 52.6-69.4), PFS remained significantly superior for Ven-Obi compared to Clb-Obi
(median not reached [nr] vs 36.4 months; hazard ratio [HR] 0.35 [95% CI 0.26-0.46],
p<0.0001). At 5 years after randomization, the estimated PFS rate was 62.6% after
Ven-Obi and 27.0% after Clb-Obi. Overall, 52 cases of progressive disease (PD) with
28 required second-line treatments occurred in the Ven-Obi arm and 132 with 86 second-line
treatments in the Clb-Obi arm. TTNT was significantly longer after Ven-Obi (5-year
TTNT 72.1% vs 42.8%; HR 0.42, 95% CI 0.31-0.57, p<0.0001). In both arms, the majority
of next-line therapies were BTK inhibitors (54.3% in the Ven-Obi arm, 47.1% in the
Clb-Obi arm). The PFS and TTNT difference was maintained across all risk groups, including
patients with TP53 mutation/deletion (5-year PFS 40.6% vs 15.6%; 5-year TTNT 48.0%
vs 20.8%) and unmutated IGHV status (5-year PFS 55.8% vs 12.5%; 5-year TTNT 66.2%
vs 25.1%). A multivariable analysis indicated 17p deletion and high disease burden
as independent prognostic factors for PFS in patients treated with Ven-Obi.
Four years after treatment completion, 39 (18.1% of the intention-to-treat population)
patients in the Ven-Obi arm still had uMRD (<10-4 by NGS in peripheral blood), 27
(12.5%) had low (L)-MRD (≥ 10-4 and < 10-2) and 41 (19.0%) high (H)-MRD (≥ 10-2),
compared to 4 (1.9%) uMRD, 13 (6.0%) L-MRD and 24 (11.1%) H-MRD in the Clb-Obi arm.
Overall, 40 deaths were reported in the Ven-Obi arm (8 PD related) and 57 in the Clb-Obi
arm (23 PD related); at 5 years after randomization the estimated OS rate was 81.9%
in the Ven-Obi arm and 77.0% in the Clb-Obi arm (HR 0.72 [0.48-1.09], p=0.12). Second
primary malignancies were reported in 44 (20.8%) patients in the Ven-Obi arm and 32
(15.0%) in the Clb-Obi arm. No new safety signals were observed.
Image:
Summary/Conclusion: These data confirm that over 60% of patients who had received
1-year fixed-duration Ven-Obi have remained in remission four years after end of therapy.
The majority of patients treated with Ven-Obi still have not required a second line
of CLL therapy. Hence, the 1-year Ven-Obi regimen continues to be an effective fixed-duration
option for patients with CLL and coexisting conditions, also in the context of high-risk
disease.
S149: LONG TERM OUTCOMES OF IFCG REGIMEN FOR FIRSTLINE TREATMENT OF PATIENTS WITH
CLL WITH MUTATED IGHV AND WITHOUT DEL(17P)/TP53 MUTATION
N. Jain1,*, P. Thompson1, J. Burger1, A. Ferrajoli1, K. Takahashi1, Z. Estrov1, G.
Borthakur1, P. Bose1, T. Kadia1, N. Pemmaraju1, K. Sasaki1, M. Konopleva1, E. Jabbour1,
N. Garg2, X. Wang3, R. Kanagal-Shamanna4, K. Patel4, W. Wang4, S. Wang4, J. Jorgensen4,
W. Lopez1, A. Ayala1, W. Plunkett5, V. Gandhi5, H. Kantarjian1, S. O’Brien6, M. Keating1,
W. Wierda1
1Leukemia; 2Radiology; 3Biostatistics; 4Hematopathology; 5Experimental Therapeutics,
The University of Texas MD Anderson Cancer Center, Houston; 6Chao Family Comprehensive
Cancer Center, University of California Irvine Medical Center, Orange, United States
of America
Background: Chemoimmunotherapy with FCR has been an effective treatment for patients
(pts) with CLL. Pts with mutated IGHV (IGHV-M) have favorable long-term outcomes after
receiving FCR. We designed an investigator-initiated, phase 2 trial with ibrutinib,
fludarabine, cyclophosphamide, and obinutuzumab (iFCG) for previously untreated pts
with IGHV-M CLL (NCT02629809). We report here long-term outcomes with a median follow-up
of 56.8 months.
Aims: Investigate the role of combined chemoimmunotherapy and targeted therapy in
CLL.
Methods: Eligibility included age ≥18, IGHV-M, no del(17p)/TP53 mutation. Pts received
3 courses of iFCG. Pts achieving CR/CRi with undetectable MRD (U-MRD) in bone marrow
(4-color flow-cytometry, sensitivity 10-4) after 3 courses of iFCG received ibrutinib
with obinutuzumab (iG) for 3 cycles, followed by ibrutinib monotherapy for 6 months.
All other pts received iG for 9 cycles (C4-12). Pts with marrow U-MRD at end of Cycle
12 stop all therapy. Response assessment was per 2008 iwCLL criteria with BM and CT
scans every 3 months during the first year. After completion of all therapy, pts were
followed by exam, blood counts and peripheral blood MRD every 6 months. NGS MRD (sensitivity
10-6) was performed in bone marrow in pts with available samples.
Results: 45 pts initiated treatment. Median age was 60 [range, 25-71]. 69% had del(13q).
After three cycles of iFCG, 39/45 (87%) pts achieved marrow U-MRD. Responses improved
with continued therapy with 40/45 (89%) and 41/45 (91%) achieving marrow U-MRD after
Cycles 6 and 12, respectively. Overall, 44/45 (98%) pts achieved marrow U-MRD as best
response at any time during the study.
The 5-year PFS and OS are 97.7% (95% CI 94–100%) and 97.8% (95% CI 94–100%), respectively.
No pt had CLL progression or Richter transformation. One pt developed therapy-related
MDS; this pt is being monitored for 38+ months without any therapy for MDS with normal
blood counts. The sole event noted on both the PFS and OS curve is a pt death from
heart failure.
41/45 pts completed 12 cycles of treatment (4 pts came off study prior to C12). All
41 pts achieved marrow U-MRD4 by flow-cytometry and per protocol, all 41 pts discontinued
ibrutinib. After a median follow-up of 44.2 mos post-discontinuing ibrutinib, 6 pts
had an MRD recurrence (defined as 2 consecutive values of ≥0.01% in peripheral blood
by flow cytometry) at a median of 27.2 mos (range, 20.7-49.0 mos) after stopping all
therapy. All 6 pts are being monitored with no clinical progression or active therapy;
notably, all 6 pts were MRD+ at 10-6 by marrow NGS after the completion of 3 cycles
of iFCG and 4/5 (1 missed sample) were MRD+ at 10-6 by marrow NGS after the completion
of Cycle 12.
Of the 16 pts who were marrow NGS MRD+ at 10-6 after 3 cycles of iFCG, 6/16 had an
MRD recurrence in blood in follow-up vs. 0/20 who were NGS MRD negative/indeterminate
(p = 0.004). Of the 12 pts who were marrow NGS MRD+ at 10-6 after Cycle 12, 4/12 had
an MRD recurrence in blood in follow-up vs. 1/26 who were NGS MRD negative/indeterminate
(p = 0.02).
Image:
Summary/Conclusion: The iFCG regimen, using only 3 cycles of chemotherapy (as opposed
to 6 cycles of chemoimmunotherapy) achieves a very high rate of U-MRD in previously-untreated
pts with CLL with IGHV-M CLL. No pt had disease progression with a median follow-up
of close to 5 years. The 5-year PFS is 97.7%; this is favorable compared to 5-year
PFS of ≈65% with FCR (CLL10), ≈70% with ibrutinib (A041202 trial), and 81% with ibrutinib
(RESONATE-2) for IGHV-M CLL. Not unexpectedly, MRD recurrence during follow-up correlated
with MRD positivity by NGS during therapy.
S150: BIOLOGY AND FUNCTION OF CIRCULAR PCMDT1 IN CHRONIC MYELOID LEUKEMIA IN BLAST
CRISIS (CML-BC)
A. Prathap Urs1,*, D. Papaioannou2, R. Buisson3, S. Khanal1, M. Karunasiri1, S. Kauppinen4,
A. Dorrance1 5, R. Garzon1 5
1Comprehensive Cancer Center, The Ohio State University, Columbus; 2NYU Langone Health,
Laura and Isaac Perlmutter Cancer Center, New York; 3Department of Biological Chemistry,
University of California, California, United States of America; 4Department of Clinical
Medicine, Aalborg University, Copenhagen, Denmark; 5Department of Internal Medicine,
The Ohio State University, Columbus, United States of America
Background: The prognostic and biologic significance of circular RNAs (circRNAs) in
patients with acute myeloid leukemia (AML) has been reported. Circular PCMTD1 (cPCMTD1)
was among the circRNAs that were prognostic in this disease.
Aims: To study the functional role of cPCMTD1 in leukemias.
Methods: We used RNase H-recruiting, locked nucleic acid-modified oligonucleotides
(gapmers) for knock-down (KD) experiments. Cell cycle analyses were performed with
bromodeoxyuridine and 7-actinomycin D staining. Chloro-deoxyuridine (CldU) and iodo-deoxyuridine
(IdU) were used for DNA fiber assays.
Results: To study whether cPCMTD1 is important for leukemia, we KD-cPCMTD1 using gapmers
that specifically degraded the circular transcript without affecting the levels of
the linear PCMTD1. Among the 8 AML cell lines that were tested, we found that depletion
of the cPCMTD1 led to a decrease in the proliferating fraction and potent G2/M blockade
of only K-562 and LAMA-84 CML-BC cells. In contrast, linear PCMTD1-KD did not have
any effect on cell cycle. Mass cytometry (CyTOF) experiments validated the cell cycle
blockade after cPCMTD1-KD and identified an increase of H2AX phosphorylation. We validated
this finding with western blots and could also show that cPCMDT1-KD led to an increase
in the phosphorylation of the ATM, ATR, CHK1, DNA-PK and RPA32 proteins. Further,
DNA fiber assays detected a global decrease in the length of CldU- and IdU-labeled
fibers as well as of the IdU/CldU ratio in the cPCMTD1-KD cells compared to controls.
These results were confirmed by COMET assays. Taken together these data indicate that
cPCMTD1-KD causes impaired DNA replication in CML-BC. Then, we examined whether we
found in CML-BC cell lines was also true for patients. Thus, we measured cPCMDT1 expression
in a panel of CML patients in chronic phase (n=15), accelerated phase (n=4) and blast
crises (n=7) and we found a significant increase of cPCDMT1 in CML-BC with respect
to chronic and accelerated phases. Next, we performed cPCMDT1-KD in CML-BC patient
samples and found increase of H2AX phosphorylation. Like the cell line data, cPCMDT1-KD
in 3 samples from AML patients had no functional effect. Finally, after confirming
that cPCMDT1 is most abundant in the cytoplasm, in silico analysis and polysome profiling
experiments indicated that cPCMDT1 encodes for a small protein. We generated a custom
antibody that detected the predicted protein of 30kD, which was depleted after cPCMDT1-KD.
Immunoprecipitation experiments followed by mass spectrometry analysis using our custom
antibody showed a strong and specific enrichment of the BTR complex (BLM, TOP3A and
RMI1), which is implicated in DNA replication and repair. cPCMDT1-KD had no effect
on the total amount of each of the three proteins but reduced their interaction and
the amount of the formed BTR complex. To validate the therapeutic potential of this
strategy we developed a CML-BC PDX model by transplanting a CML-BC patient sample
into NSGS mice. This PDX is very aggressive, and mice die from disease by 20 weeks.
We are currently treating these mice in vivo with lipid nanoparticle anti-cPCMDT1
gapmers. Results will be updated in the meeting.
Summary/Conclusion:
cPCMDT1 is a circRNA with protein-coding potential that is over-expressed in CML-BC.
cPCDMT1 is critical for the proliferation of CML-BC through the regulation of DNA
replication and may represent a novel target for therapy.
S151: HIGH-THROUGHPUT EVALUATION OF THE POTENTIAL OF CANCER DRUGS TO ENHANCE NATURAL
KILLER CELL IMMUNOTHERAPY IN CHRONIC MYELOID LEUKEMIA.
P. Nygren1 2,*, J. Bouhlal1 2, E. Laajala1 2, A. Ianevski3, J. Klievink1 2, H. Lähteenmäki1
2, K. Saeed1 2, D. Lee4, T. Aittokallio3 5 6, O. Dufva1 2, S. Mustjoki2 7
1Hematology Research Unit Helsinki, University of Helsinki and Helsinki University
Hospital; 2Translational Immunology Research Program; 3Institute for Molecular Medicine
Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland; 4Division of Hematology,
Oncology, and BMT, Nationwide Children’s Hospital, Columbus, United States of America;
5Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital;
6Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University
of Oslo, Oslo, Norway; 7Hematology Research Unit Helsinki, University of Helsinki
and Helsinki University Hospital, Helsinki, Helsinki, Finland
Background: Natural killer (NK) cell immunotherapies are promising novel cancer treatments
and complete remission has been achieved in patients with relapsed/refractory myeloid
leukemias. The significance of NK cells in chronic myeloid leukemia (CML) patients
has been highlighted in several studies. CML patients with a higher percentage of
mature NK cells have increased relapse-free survival after treatment discontinuation.
NK KIR receptor profiles have also been associated with the achievement of remission
in response to tyrosine kinase inhibitor (TKI) therapy. Moreover, complex interactions
between TKIs and NK cell function have been discovered in CML. For example, dasatinib
and imatinib have been suggested to enhance NK cell cytotoxicity through regulation
of activating and inhibitory receptors. NK cell responses are usually short-lived
and attempts have been made to enhance their function against malignant cells using
cytokines, bi- and tri-specific antibodies, and small molecule inhibitors. However,
a large-scale drug screening evaluating the combinatorial effects of NK cells and
cancer drugs has not been previously conducted.
Aims: To evaluate the potential of small molecule cancer drugs to synergize with NK
cell immunotherapy in CML and to discover which drugs can enhance or inhibit cytotoxicity
in CML cells.
Methods: A high-throughput drug sensitivity and resistance testing (DSRT) screen was
used to evaluate the effect of 528 investigational and approved cancer drugs on the
cytotoxicity of NK cells. CML cells (K562) expressing luciferase and primary expanded
NK cells were co-cultured for 24 hours on 384 well drug plates. Target cell viability
was measured using a luciferase-based readout, and results were run through a custom
pipeline to calculate differential drug sensitivity scores (dDSS) between co-cultured
cells and target cells alone, for each drug. Most promising drugs with highest and
lowest dDSS were selected for further analysis with single-cell RNA sequencing to
determine their mechanism of action.
Results: Out of 528 screened drugs, 161 had an inhibitory effect on cytotoxicity (dDSS
< -2.5), 325 had no effect, and 42 had an enhancing effect (dDSS > 2.5). The SMAC
mimetics birinapant and LCL-161 were the most potent enhancers of NK cell cytotoxicity
and had no effect on target cells alone. Individual drugs with other mechanisms, such
as PKC activators, were also effective enhancers of NK cytotoxicity. On the contrary,
cytotoxicity-inhibiting drugs included previously known immunosuppressive drugs dexamethasone
and prednisolone, as well as novel candidates, such as sotrastaurin, a PKC inhibitor.
Single-cell RNA sequencing of drug-treated co-cultures revealed the transcriptomic
phenotypes of NK cell and target cells under SMAC mimetic treatment. NF-kB target
genes such as BIRC3, NFKB2, MHC II genes, and the T/NK cell-recruiting chemokines
CXCL9, CXCL10, and CXCL11 were activated in target cells exposed to both birinapant
and NK cells, compared to target cells exposed to NK cells alone (p value adj < 0.05).
These data suggest that SMAC mimetics in combination with NK cells may induce an immune-inflamed
phenotype in addition to sensitizing CML cells to NK cell cytotoxicity.
Image:
Summary/Conclusion: We discovered novel drug classes, which had both activating and
inhibitory effects on the NK cell cytotoxicity against CML cells in vitro. Defining
NK cell - drug - CML cell interactions in a high-throughput setting provides a framework
for future combination immunotherapies to further improve treatment-free remission
in CML.
S152: SETD2/H3K36ME3 DEFICIENCY SUSTAINS GENOMIC INSTABILITY AND ENHANCES CLONOGENIC
POTENTIAL OF CHRONIC MYELOID LEUKEMIA (CML) PROGENITORS
M. Mancini1,*, S. De Santis2, C. Monaldi2, S. Bruno2, F. Castagnetti2, G. Gugliotta1,
A. Iurlo3, M. Cerrano4, S. Galimberti5, S. Balducci5, F. Stagno6, G. Rosti7, M. Cavo8,
S. Soverini2
1IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”;
2Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di
Bologna, Bologna; 3UO Onco-ematologia, Fondazione IRCCS Ca’ Granda - Ospedale Policlinico,
Milano; 4A.O.U. Citta della Salute e della Scienza di Torino, Torino; 5Clinical and
Experimental Medicine, Hematology, University of Pisa, Pisa; 6Ematologia Universitaria
e Trapianto Midollo Osseo, Ospedale Gaspare Rodolico, Catania; 7Istituto Romagnolo
per lo Studio dei Tumori “Dino Amadori” - IRST Srl Istituto di Ricovero e Cura a Carattere
Scientifico (IRCCS), Meldola (FC); 8IRCCS Azienda Ospedaliero-Universitaria di Bologna,
Istituto di Ematologia “Seràgnoli”; Dipartimento di Medicina Specialistica, Diagnostica
e Sperimentale, Università di Bologna, Bologna, Italy
Background: Genomic instability is a hallmark of chronic myeloid leukemia (CML) cells
since the chronic phase (CP) of the disease and results in BCR-ABL1 mutations and/or
additional genetic and genomic aberrations that may drive resistance to tyrosine kinase
inhibitors (TKIs) and progression to blast crisis (BC). Genomic instability is also
a feature of CML stem cells and may underlie their persistence. We have recently reported
that virtually all CML patients (pts) in BC display loss of function of SETD2, an
enzyme that trimethylates histone H3 lysine 36 (H3K36me3). SETD2 is a tumor suppressor
implicated in several neoplastic conditions. H3K36me3 is required for homologous recombination
(HR) repair of double strand breaks and for DNA mismatch repair (MMR).
Aims: We investigated SETD2/H3K36me3 status in CD34+ progenitors of CP CML pts and
whether SETD2/H3K36me3 deficiency may play a role in genomic instability in CML models.
Methods: CD34+ progenitor cells were isolated from 20 newly diagnosed CP CML pts and
screened for SETD2 and H3K36me3 by Western blotting (WB). SETD2-proficient (LAMA84)
and -deficient (KCL22) CML cell lines were also studied. SETD2 knock-down and SETD2
forced expression in CD34+ progenitors and cell lines were performed by RNAi and nucleofection,
respectively. DNA damage and DNA repair activation were assessed by WB and immunofluorescence
(IF). Clonogenic capacity was evaluated by clonogenic assays.
Results: Loss of SETD2 protein expression and function (the latter assessed using
loss of H3K36me3 as surrogate marker) were detected by WB in the CD34+ cell fraction
of 20/20 newly diagnosed CP CML pts, as compared to the corresponding total mononuclear
cell fraction or to the CD34+ fraction obtained from a pool of healthy donors. To
investigate whether SETD2/H3K36me3 loss impinges on the activation and proficiency
of HR, we used UV rays to induce DNA damage in SETD2 siRNA-depleted LAMA 84 (SETD2-proficient)
cells. Compared to control cells, cells silenced for SETD2 displayed an increase in
the expression of the DNA damage maker γH2AX associated with a loss of RAD51 (HR)
and MSH6 (MMR) repair foci. To confirm the role of SETD2 as a tumor suppressor implicated
in maintaining genomic stability in CML, we transfected KCL22 (SETD2-deficient) cells
with an ectopic SETD2 plasmid. SETD2 forced expression induced more than 50% reduction
in cell doubling time and a significant reduction in clonogenic potential. Moreover,
SETD2 overexpression was able to restore DNA damage response, as demonstrated by WB
and immunofluorescence detection of H2AX phosphorylation, RAD51 (HR) foci, THEX1 (DNA
replication) and MSH6 (MMR) observed after UV exposure.
In line with the effects of SETD2 deficiency/overexpression observed in cell line
models, CML progenitor cells displaying SETD2/H3K36me3 deficiency showed hyper-phosphorylation
of H2AX and the overexpression of RAD51 (HR) and MSH6 (MMR) did not result in DNA
repair foci. Moreover, forced overexpression of SETD2 restored proliferation control
in CD34+ cells from 5 CP CML pts, since after nucleofection clonogenic assays showed
a >50% reduction in clonogenic potential.
Summary/Conclusion: SETD2/H3K36me3 deficiency is a novel, BCR-ABL1-independent mechanism
of genetic instability in CML. SETD2 behaves like a true tumor suppressor in CD34+
progenitor cells of CP CML pts. Supported by AIRC IG 2019 grant (23001) and Italian
Ministry of Health, “Bando Ricerca Finalizzata 2016”, project GR-2016-02364880.
S153: DECRYPTING THE ROLE OF HSP90Α AND Β ISOFORMS TO OVERCOME RESISTANCE IN BCR-ABL1
LEUKEMIA
M. Vogt1 2,*, N. Dienstbier1, J. Schliehe-Diecks1, K. Scharov1, J.-W. Tu1, P. Gebing1,
J. Hogenkamp1, D. Picard1, T. Lenz3, K. Stühler3 4, R. Wagener1 5, A. Pandyra1 5,
J. Hauer6, U. Fischer1 5, A. Borkhardt1 5, S. Bhatia1 5
1Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty,
Heinrich Heine University Düsseldorf, University Hospital; 2DSO - Düsseldorf school
of oncology; 3Molecular Proteomics Laboratory, Biological-Medical Research Center,
Heinrich Heine University Düsseldorf; 4Institute of Molecular Medicine, Proteome Research,
Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf; 5German
Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf; 6Department of
Pediatrics, Technical University of Munich, School of Medicine, Munich, Germany
Background: The BCR-ABL1 fusion protein, which results from the chromosomal translocation
t(9;22)(q34;q11) is detected in over 95% of CML, 25% of adult BCP-ALL and 3-5% of
childhood BCP-ALL patients. The development of tyrosine kinase inhibitors (TKi) has
enabled targeted treatment, however development of resistance is common and the BCR-ABL1
leukemia subtype is still associated with poor prognosis. Among the chaperone proteins
involved in maintaining the homeostasis of cancerous cells, heat shock protein 90
(HSP90) have been widely studied, due to its crucial involvement in stabilizing a
variety of oncogenic proteins, including BCR-ABL1 and STAT3/5. Therefore, inhibition
of HSP90 can be an effective alternative approach to target TKi resistant and BCR-ABL1T315I
mutant expressing cells. In the past, over 17 HSP90 inhibitors (HSP90i) have been
evaluated in clinical trials, however associated resistances (e.g. HSR) and dose limited
toxicity have thus far precluded their clinical approval.
Aims: In mammalian cells, there are two predominant cytosolic isoforms of HSP90, a
stress-inducible HSP90α and a constitutively expressed HSP90β isoform. In the past,
the distinct roles of the isoforms were studied by applying pan- and isoform-targeted
HSP90i. We aimed to decrypt the role of cytosolic HSP90 isoforms using genetically
edited models and to identify novel therapeutic vulnerabilities to target therapy
refractory BCR-ABL1+ leukemia.
Methods: To study the kinetic implications of the resistance mechanism associated
with pharmacological targeting of HSP90, we generated a transient knockdown (Kd) and
a stable CRISPR-Cas9 based knockout (KO) model of HSP90α/β in BCR-ABL1+ cells. The
edited cells were analyzed in ex vivo functional assays (e.g. IF staining and WB)
and their transplantation efficiency was determined in NSG mice. Global transcriptomic
and proteomic profiling was performed in conjunction with high throughput drug screening
(HTDS) to identify therapeutic vulnerabilities upon loss of HSP90α/β.
Results: HSP90α/β-KO or -Kd did not affect the expression of other HSP90 paralogues
(TRAP1 and GRP94). Only HSP90β-KO cells displayed hyperactive pro-survival HSR. Other
reported HSP90 isoform-dependent clients, such as Survivin, CDK4 and CDK6 were also
not affected upon KO or Kd of HSP90α/β. As HSP90 is involved in stabilizing, proper
folding and subcellular localization of BCR-ABL1 protein, immunofluorescence imaging
demonstrated two fold higher abundance of BCR-ABL1 foci (cytoplasmic/nucleocytoplasmic
region) in HSP90α KO cells, with hyperactive BCR-ABL1 and downstream signaling (pBCR-ABL
and pSTAT5a). When KO cells were transplanted into NSG mice (n=5 mice per arm) the
engraftment of the HSP90α-KO cells was significantly reduced with a prolonged overall
survival of the animals (19 days, p=0.0083) as compared to HSP90β-KO and control group.
To find possible target (s) in order to circumvent resistance associated with the
use HSP90i, we performed HTDS with HSP90α/β KO cells, which revealed distinct sensitivities
toward certain classes of inhibitors. One of these classes was CDK7i, which was differentially
active in HSP90α KO cells. These results can be explained by our global transcriptomic
results, which revealed hyperactive androgen receptor signaling in HSP90α KO cells
(GSEA), which is known to be a prognostic marker for CDK7 inhibition.
Summary/Conclusion: In vivo targeting of HSP90α is a promising approach to target
BCR-ABL1+ leukemia cells. Moreover, the combination with CDK7i may help to overcome
the resistance associated with clinical use of pan HSP90i.
S154: IMATINIB-RESISTANT CLONES ISOLATED FROM A MODEL OF BLAST CRISIS OF CHRONIC MYELOID
LEUKAEMIA DIFFER IN MUTATIONS IN BCR::ABL1 AND OTHER CANCER RELATED GENES AND IN THEIR
SENSITIVITY TO BH3-MIMETICS
A. Laznicka1 2,*, N. Curik1 3, V. Polivkova1, J. Koblihova1, P. Burda1 3, A. Dolnikova3,
E. Pokorna3, C. Salek1, H. Klamova1, D. Srbova1, P. Klener3 4, K. Machova Polakova1
3
1Institute of Hematology and Blood Transfusion; 2Second Faculty of Medicine, Charles
University; 3Institute of Pathological Physiology, First Faculty of Medicine, Charles
University; 41st Dep. of Medicine – Dep. of Hematology, First Faculty of Medicine,
Charles University, and General University Hospital, Prague, Czechia
Background: The treatment of blast crisis of chronic myeloid leukaemia (BC-CML) remains
difficult and the outcome of patients is poor.
A dysregulated apoptosis leads to survival of malignant cells. BH3-mimetics inhibit
antiapoptotic proteins of BCL-2 family. These drugs have been recently introduced
in the treatment of chronic lymphocytic leukaemia and acute myeloid leukaemia. This
work assumed that these drugs may have a treatment potential for myeloid and/or lymphoid
BC-CML.
Aims: The aim is to test the sensitivity of imatinib-resistant (IR) clones of KCL-22,
model of BC-CML, and primary blast cells to BH3-mimetics and describe a mechanism
of sensitivity/resistance using protein analysis of apoptotic pathways.
Methods: Isolated IR-clones (n=10) of KCL-22 were characterized by NGS and cultivated
with 4µM imatinib (IM) and dilution series of BH3-mimetics (venetoclax – anti-BCL-2,
S63845 – anti-MCL-1, A-1155463 - anti-BCL-XL) for 72 hours. IC50 was determined based
on proliferation. Protein expression of BCL-2 family (n=10) was conducted in 4 IR-clones
cultivated with IM and with/without BH3-mimetics. For in vivo experiments NOD-SCID-gamma
mice (n=16) were subcutaneously injected with 106 cells of BCR::ABL1-T315I clone and
divided into control group and treated groups. Primary cells of patients with BC-CML
(n=4) were exposed to IM and BH3-mimetics for 7 days and LC50 was estimated.
Results: IR-clones of KCL-22 differ in their sensitivity to BH3-mimetics (Table 1).
The majority of clones (8/10) including 4 clones with T315I were sensitive to MCL-1
inhibitor. A reduced sensitivity was observed in the T315I clone carrying mutated
GATA2 and other clone with mutation Y253H and mutated BCOR. Moreover, two T315I clones
were sensitive to venetoclax, but other T315I clones with mutations in other cancer
related genes showed insensitivity (n=2) or decreased sensitivity (n=1). Interestingly,
two T315I clones with mutations in other cancer related genes insensitive to venetoclax
were sensitive to BCL-XL inhibitor and in general, it seems that clones insensitive
to venetoclax were sensitive to BCL-XL inhibitor and vice versa.
Protein analysis of parental KCL-22IR cells and 4 clones showed a high expression
of BCL-2 and BCL-XL and undetectable MCL-1 expression. Except for the T315I clone
(B8, Table 1), other analysed clones expressed MCL-1 after exposure to BH3-mimetics.
The sensitivity of T315I clone to anti-MCL-1 can be explained by detection of cBAX
(an active form of apoptotic effector) after exposure to the inhibitor. The parental
cell line showed dissociation of antiapoptotic complex MCL-1/BIM after exposure to
MCL-1 inhibitor. The released BIM accelerates the apoptosis as an apoptotic activator.
In vivo analysis revealed a decreased tumour growth of xenografted T315I clone (B8)
in IM, venetoclax or combined treatment compared to control. An additive effect of
dual therapy against monotherapy was not observed. This can be explained by IM inhibition
of non-mutated BCR::ABL1 in KCL-22IR carrying 2 Ph chromosomes.
LC50 analysis of primary blasts (4 patients) showed anti-MCL-1 to be the most potent
inhibitor. Mutations in other cancer related genes and/or changes in karyotype increased
LC50 for venetoclax and BCL-XL inhibitor or venetoclax, respectively.
Image:
Summary/Conclusion: The preliminary data of this study showed that the MCL-1 inhibitor
S63845 seems to be the most potent BH3-mimetic to induce apoptosis in BC-CML. The
combined therapy of TKI and BH3-mimetics should reflect mutation status of BCR::ABL1
and other cancer related genes.
S155: EFFICACY AND SAFETY RESULTS FROM ASCEMBL, A PHASE 3 STUDY OF ASCIMINIB VS BOSUTINIB
IN PATIENTS WITH CHRONIC MYELOID LEUKEMIA IN CHRONIC PHASE AFTER ≥2 PRIOR TYROSINE
KINASE INHIBITORS: WK 96 UPDATE
D. Rea1,*, A. Hochhaus2, M. J. Mauro3, Y. Minami4, E. Lomaia5, S. Voloshin6, A. Turkina7,
D.-W. Kim8, J. F. Apperley9, J. E. Cortes10, A. Abdo11, L. M. Fogliatto12, D. D. H
Kim13, P. le Coutre14, S. Saussele15, M. Annunziata16, T. P. Hughes17, N. Chaudhri18,
L. Chee19, V. García-Gutiérrez20, K. Sasaki21, S. Kapoor22, A. Allepuz23, S. Quenet23,
V. Bédoucha23, C. Boquimpani24
1Hôpital Saint-Louis, Paris, France; 2Universitätsklinikum Jena, Jena, Germany; 3Memorial
Sloan Kettering Cancer Center, New York, United States of America; 4National Cancer
Center Hospital East, Kashiwa, Japan; 5Almazov National Medical Research Centre; 6Russian
Research Institute of Hematology and Transfusiology, St. Petersburg; 7National Medical
Research Center for Hematology, Moscow, Russia; 8Uijeongbu Eulji Medical Center, Geumo-dong,
Uijeongbu-si, South Korea; 9Centre for Haematology Imperial College London, London,
United Kingdom; 10Georgia Cancer Center, Augusta, United States of America; 11Instituto
do Câncer do Estado de São Paulo (ICESPSP), São Paulo; 12Hospital de Clínicas de Porto
Alegre (HCPA), Porto Alegre, Brazil; 13Princess Margaret Cancer Centre, University
Health Network, University of Toronto, Toronto, Canada; 14Charité–Universitätsmedizin
Berlin, Berlin; 15III. Medizinische Klinik, Medizinische Fakultät Mannheim der Universität
Heidelberg, Mannheim, Germany; 16Division of Hematology, AORN Cardarelli, Naples,
Italy; 17South Australian Health and Medical Research Institute and University of
Adelaide, Adelaide, Australia; 18King Faisal Specialist Hospital & Research Center,
Riyadh, Saudi Arabia; 19Peter MacCallum Cancer Center and The Royal Melbourne Hospital,
Victoria, Australia; 20Servicio de Hematología, Hospital Universitario Ramón y Cajal
(IRYCIS), Madrid, Spain; 21The University of Texas MD Anderson Cancer Center, Houston;
22Novartis Pharmaceuticals Corporation, East Hanover, United States of America; 23Novartis
Pharma AG, Basel, Switzerland; 24HEMORIO, State Institute of Hematology Arthur de
Siquiera Cavalcanti, Rio de Janeiro, Brazil Oncoclínica Centro de Tratamento Oncológico,
Rio de Janeiro, Brazil
Background: Asciminib is the 1st BCR::ABL1 inhibitor to Specifically Target the ABL
Myristoyl Pocket (STAMP). In the ASCEMBL primary analysis, asciminib had superior
efficacy and better safety/tolerability vs bosutinib (BOS) in patients (pts) with
chronic myeloid leukemia in chronic phase (CML-CP) after ≥2 prior tyrosine kinase
inhibitors (TKIs). Major molecular response (MMR) rate at wk 24 was 25.5% on asciminib
vs 13.2% on BOS; the difference in MMR rates after adjusting for baseline major cytogenetic
response (MCyR) was 12.2% (95% CI, 2.19%-22.30%; 2-sided P=.029). Fewer grade ≥3 adverse
events (AEs) and AEs leading to treatment discontinuation occurred on asciminib vs
BOS. After a median follow-up of 2.3 years (16.5 months’ additional follow-up since
the primary analysis), we report updated efficacy and safety results (cutoff: October
6, 2021).
Aims: The key secondary objective was to compare MMR rate at wk 96 on asciminib vs
BOS.
Methods: Eligible pts provided informed consent, were adults with CML-CP after ≥2
prior TKIs, with intolerance or lack of efficacy per 2013 European LeukemiaNet recommendations.
They were randomized 2:1 to asciminib 40 mg twice daily or BOS 500 mg once daily,
stratified by baseline MCyR status (Ph+ metaphases ≤35%).
Results: 233 pts were randomized to asciminib (n=157) or BOS (n=76). At cutoff, treatment
was ongoing in 84 (53.5%) and 15 (19.7%) pts, respectively; the most common reason
for discontinuation was lack of efficacy in 38 (24.2%) and 27 (35.5%) pts, respectively.
MMR rate at wk 96 (per ITT) was 37.6% on asciminib and 15.8% on BOS, meeting the key
secondary objective. The difference after adjusting for baseline MCyR was 21.7% (95%
CI, 10.5%-33.0%; 2-sided P=.001). Preplanned subgroup analyses showed that MMR rate
at wk 96 was consistently higher with asciminib than BOS in all demographic and prognostic
subgroups, including all prior lines of TKI therapy, and regardless of the reason
for discontinuation of the last TKI (Figure).
At wk 96, more pts on asciminib than BOS had BCR::ABL1
IS ≤1% (45.1% vs 19.4%) (Table). Responses were durable, with a probability (95% CI)
of maintaining MMR and BCR::ABL1
IS ≤1% for ≥72 wk of 96.7% (87.4%-99.2%) and 94.6% (86.2%-97.9%), respectively, on
asciminib and 92.9% (59.1%-99.0%) and 95.0% (69.5%-99.3%), respectively, on BOS. Median
time to treatment failure was 24 months on asciminib and 6 months on BOS.
Median duration (range) of exposure was 103.1 (0.1-201.1) wk on asciminib and 30.5
(1.0-188.3) wk on BOS. Despite asciminib’s longer duration of exposure, its safety/tolerability
continued to be better than that of BOS (Table). Fewer pts on asciminib than BOS had
AEs leading to treatment discontinuation (7.7% vs 26.3%). No new on-treatment deaths
were reported since the primary analysis. Most frequent (>10%) grade ≥3 AEs on asciminib
vs BOS were thrombocytopenia (22.4%, 9.2%), neutropenia (18.6%, 14.5%), diarrhea (0%,
10.5%), and increased alanine aminotransferase (0.6%, 14.5%).
Image:
Summary/Conclusion: After >2 years of follow-up, asciminib continued to show clinically
and statistically significant, superior efficacy and better safety/tolerability vs
BOS. Responses were durable, and MMR was more than double on asciminib than BOS. The
difference in MMR rates between the 2 arms increased from 12.2% at wk 24 to 21.7%
at wk 96. A higher proportion of pts had BCR::ABL1
IS ≤1%, a milestone response in later lines that is associated with improved long-term
survival. These results further support the use of asciminib as a new CML therapy,
with the potential to transform standard of care.
S156: INTERNATIONAL, PROSPECTIVE STUDY COMPARING NILOTINIB VERSUS IMATINIB WITH EARLY
SWITCH TO NILOTINIB TO OBTAIN SUSTAINED TREATMENT-FREE REMISSION IN PATIENTS WITH
WITH CHRONIC MYELOID LEUKEMIA
F. Pane1,*, F. Castagnetti2, L. Luciano1, A. Russo Rossi3, E. Abruzzese4, R. Bassan5,
G. Binotto6, G. Caocci7, G. Cimino8, P. Fazi9, A. Gozzini10, M. Lunghi11, R. Marasca12,
B. Martino13, M. Bonifacio14, F. Cavazzini15, F. Paoloni16, G. Saglio17, S. Sica18,
A. Tafuri19, D. Vallisa20, M. Vignetti9, P. Westerweel21, G. Rosti22, M. Breccia23
1Dpt. of Clinical Medicine and Surgery, University of Naples, Naples; 2Department
of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna;
3Hematology and Transplantation Unit, University of Bari, Bari; 4Division of Hematology,
Ospedale S. Eugenio, Rome; 5Hematology Unit, Ospedale Dell’Angelo, Venezia-Mestre;
6Hematology Unit, University of Padova, Padova; 7Department of Medical Sciences and
Public Health, Hematology, Businco Hospital, University of Cagliari, Cagliari; 8UOC
Ematologia e trapianto, S.Maria Goretti Hospital, Latina; 9GIMEMA Data Center, Fondazione
GIMEMA Franco Mandelli Onlus, rome; 10Department of Cellular Therapy and Transfusional
Medicine, AUO Careggi, Florence; 11Division of Hematology, Department of Translational
Medicine, University of Eastern Piedmont, Novara; 12Section of Hematology, Department
of Medical Sciences, University of Modena and Reggio Emilia, Modena; 13Division of
Hematology, Azienda Ospedaliera ‘Bianchi Melacrino Morelli’, Reggio Calabria; 14Section
of Hematology, Department of Medicine, University of Verona, Verona; 15Hematology
Section, Department of Medical Sciences, University of Ferrara, University Hospital
Arcispedale S. Anna, Ferrara; 16Biostatistic Central Office, Fondazione GIMEMA Franco
Mandelli Onlus, Rome; 17Hematology Division, University of Turin, Turin; 18Ematologia,
FONDAZIONE POLICLINICO UNIVERSITARIO AGOSTINO GEMELLI IRCCS, Roma; 19Department of
Clinical and Molecular Medicine, Hematology, Sant’Andrea University HospitalSapienza
University of Rome, Rome; 20Unit of Hematology, Department of Oncology and Hematology,
Guglielmo da Saliceto Hospital, Piacenza, Italy; 21Internal Medicine, Albert Schweitzer
Hospital, Dordrecht, Netherlands; 22Hematology Unit, IRCCS Istituto Romagnolo per
lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola; 23Translational medicine OUC
Hematology, La Sapienza university, Rome, Italy
Background: Treatment free remission (TFR) is one of the most important goals of the
CML treatment but, so far, the best treatment to reach this aim is still undefined.
It is widely accepted that a sustained deep molecular remission (DMR) is the pre-requisite
to discontinue TKI, and it is expected that in patients who start treatment with imatinib
(IMA) and fail early molecular remission (EMR), a switch to a second generation TKI
may improve the probability of achieving a DMR
Aims: We launched in November 2016 an international, prospective, interventional,
randomized, two arms, study to evaluate both the depth of the molecular response and
the rate of TFR in newly diagnosed CP-CML patients treated with a second generation
TKI (Nilotinib, NIL) or with IMA followed by switching to NIL in absence of optimal
response (defined according the ELN 2013 criteria (Clinical Trial number 02602314).
Methods: The patients are randomized 1:1 between NIL and IM according to Sokal risk
score (high versus intermediate/low risk) and country. All the patients who obtain
a residual disease reduction greater than 4.0 logs (MR4.0) within the first three
years of treatment and maintain this level of response up to the end of the fourth
years of therapy qualify for the discontinuation phase of the study. The study has
two primary end-points: a) the rates of molecular response (MR4.5) at 24 months, and
b) the rate of patients who remain in sustained treatment free remission (≥MR3.0)
without molecular relapse 12 months after entering the TFR phase. The molecular relapse
is defined as loss of MMR or confirmed loss of MR3.0 (figure).
Results: From November 2016 to January 2021, 457 patients with newly diagnosed CP-CML
patients were enrolled into the study and 448 of these (228 and 220 randomized to
the NIL and IMA arms, respectively) were evaluable (mean age 54.2 yrs - range 19.4
– 85.8). At baseline, 183 (40.8%), 191 (42.6%) and 72 (16.1%) patients were classified
as low, intermediate, or high-risk Sokal, respectively, while 278 (62.3%), 128 (28.7%)
and 40 (9.0%) had a low, intermediate, or high-risk ELTS risk score. The median follow-up
of the whole cohort of patients is 30.4 mo. Fifthy-six (25.4%) of the 220 patients
of the IMA arm did not fulfill the ELN criteria for optimal response within the first
12 mo. of treatment and, according to the protocol, switched to NIL therapy. At the
last analysis of the protocol database (February 2022), 59 patients had had stopped
the protocol treatment since their decision, death (24), toxicity (23), progression
(9), uncontrolled second neoplasia (2) or protocol violation (1), 69 patients had
not reach 24 mo. of follow-up and other 15 had missing data. Of the remaining 304
patients, 35 showed non optimal response to therapy. At the 24 mo. of follow-up, 76
of the 322 patients with an available molecular response (23.6%), reached a MR4.5
response that showed a significantly higher frequency within the patients randomized
to the NIL arm (48 vs 28; p=0.015) (first primary co-endpoint of the study).
Image:
Summary/Conclusion: This is the first and, so far, the unique prospective study comparing
not only the rate of DMR but, more important, also the rate of TFR according to treatment:
a second generation TKI frontline vs IMA frontline followed by the same second generation
in case of non-optimal response. The analysis of the first co-primary endpoint indicates
that, despite the early switch in the IMA randomized patients, NIL therapy is more
effective to induce DMR. Subsequent analysis will clarify whether the higher rates
of DMR in the NIL arm may translate into a higher rate of TFR.
S157: BCR::ABL1 DIGITAL PCR IDENTIFIES CHRONIC PHASE CML PATIENTS SUITABLE FOR AN
EARLY TKI DISCONTINUATION ATTEMPT: A PATIENT-LEVEL META-ANALYSIS
C. Kockerols1,*, S. Dulucq2, S. Bernardi3, M. Farina3, I. Civettini4, G. Colafigli5,
S. Mori6, P. Valk7, F.-X. Mahon8, C. Gambacorti-Passerini4 9, F.-E. Nicolini10, M.
Breccia5, D. Russo3, P. E. Westerweel1
1Internal Medicine, Albert Schweitzer Hospital, Dordrecht, Netherlands; 2Laboratory
of Hematology, Hôpital Haut Lévêque, University hospital of Bordeaux, Pessac, France;
3Clinical and Experimental Sciences, Hematology; 2and Bone Marrow Transplant Center,
University of Brescia, ASST Spedali Civili of Brescia, Brescia; 4University of Milano
Bicocca, Monza; 5Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza
University, Rome; 6University of Milano-Bicocca, Monza, Italy; 7Molecular Biology,
Erasmus University Medical Center, Rotterdam, Netherlands; 8Hematology, Institut Bergonié,
Bordeaux, France; 9Hematology, S. Gerardo Hospital, Monza, Italy; 10Hematology, Centre
Léon Bérard and CRCL, Lyon, France
Background: Digital PCR (D-PCR) is an emerging technique that delivers a highly accurate
BCR::ABL1 quantification, even in CML patients with low residual disease. This is
crucial in the context of treatment-free remission (TFR) for the selection of patients
who may successfully discontinue TKI therapy. However, it is unclear how its prognostic
value relates to time variables such as treatment duration prior to the TFR attempt.
Current guidelines suggest aiming for a TKI treatment duration >6 years to increase
TFR success rate.
Aims: In current analysis, we aimed to assess the prognostic value of BCR::ABL D-PCR
in relation to prior TKI treatment duration.
Methods: We performed an Individual Patient Data Meta-Analysis (IPD-MA) combining
data from different study cohorts in which BCR::ABL1 was assessed by D-PCR prior to
TKI discontinuation. Eligibility criteria included age ≥18 years and CML diagnosed
in chronic phase. Data of the participating studies were pooled and stratified based
on D-PCR and/or treatment duration. BCR::ABL1 D-PCR was dichotomized based on each
study-defined prediction cut-off. Strata were assessed for molecular relapse (MolR)
with Kaplan-Meier estimates and with cox regression analysis including a frailty term
for correction for between-study heterogeneity and including confounding variables:
age at diagnosis, gender, Sokal score, TKI generation, treatment duration, DMR duration
(model 1) and BCR::ABL1 transcript type (model 2). MolR was defined as a BCR::ABL1
>0,1%IS or a 1-log BCR::ABL1 increase in two consecutive analyses. Patients were censored
at last follow-up.
Results: For this meta-analysis, data were combined from five cohorts: four published
cohorts (STIM2 cohort [Nicolini et al.], n=175; ISAV cohort [Diral et al.], n=107;
Bernardi et al., n=111; Colafigli et al., n=50) and 1 unpublished cohort (Dutch cohort;
n=40). The pooled dataset comprised 483 patients (Table).
A total of 205 patients (42%) experienced MolR with a median time to relapse of 3
months. Median follow-up duration for TFR patients was 27 months. MolR patients had
a significantly shorter treatment duration prior to TKI discontinuation (6,7 vs 7,9
years, p=0.006) and more often presented a BCR::ABL1 D-PCR above the study-defined
prediction cut-off (34% vs 19%, p<0.001). Interestingly, the median treatment durations
were almost identical in patients with a BCR::ABL1 below or above the cut-off (7.0
vs 7.5 years, p = 0.470).
The probability of MolR at 24 months was 38% versus 58% for patients with a D-PCR
BCR::ABL1 below versus above the prediction cut-off (p <0.001). In the cox regression
analysis, the HR of D-PCR BCR::ABL1 below the cut-off for MolR was 0.48 (95% CI 0.35-0.66,
p<0.001). Treatment duration and BCR::ABL1 transcript type also remained independent
predictors, but TKI generation and DMR duration did not.
When stratifying into 4 groups based on the D-PCR result and treatment duration, patients
with a TKI treatment for ≥6 years and low D-PCR result had the lowest MolR rate (33%
at 24 months, figure). Patients treated <6 years and with a low D-PCR result had a
rate of 48% at 24 months, while patients treated <6 years and a D-PCR result above
the cut-off had the highest MolR rate of 72% at 24 months.
Image:
Summary/Conclusion: These combined patient-level data of multiple CML cohorts further
support the independent prognostic value of BCR-ABL1 D-PCR for TFR success rate. Importantly,
patients with a TKI treatment duration <6 years and a low D-PCR result were found
to have a clinically acceptable MolR rate (48%).
S158: FINAL RESULT OF TKI DISCONTINUATION TRIAL WITH DASATINIB FOR SECOND ATTEMPT
OF TREATMENT FREE REMISSION AFTER FAILING FIRST ATTEMPT WITH IMATINIB: TREATMENT-FREE
REMISSION ACCOMPLISHED BY DASATINIB
D. Kim1,*, E. Atenafu2, D. Forrest3, I. Bence-Bruckler4, L. Savoie5, M.-M. Keating6,
L. Busque7, R. Delage8, A. Xenocostas9, E. Liew10, P. Laneuville11, K. Paulson12,
T. Stockley13, J. Lipton1, B. Leber14
1Princess Margaret Cancer Centre, Toronto, Canada; 2Statistics, Princess Margaret
Cancer Centre, Toronto; 3Vancouver General Hospital, Vancouver; 4Ottawa Hospital Research
Institute, Ottawa; 5University of Calgary, Calgary; 6Queen Elizabeth II Health Sciences
Centre, Halifax; 7Hôpital Maisonneuve-Rosemont, Montreal; 8Centre Universitaire d’Hématologie
et d’Oncologie de Québec, Quebec; 9London Health Sciences Centre, London; 10University
of Alberta, Edmonton; 11McGill University Health Centre, Montreal; 12CancerCare Manitoba,
Winnipeg; 13Division of Clinical Laboratory Genetics, University Health Network, Toronto;
14Juravinstki Cancer Centre, Hamilton, Canada
Background: The Canadian tyrosine kinase inhibitor (TKI) discontinuation (DISC) trial
evaluated if Dasatinib (DA) therapy can lead to a successful second treatment-free
remission (TFR2) after failing a Imatinib (IM) DISC for first TFR (TFR1) attempt.
We previously reported that 1) The 12-month molecular relapse free survival (mRFS)
rate for TFR1 was 58.0%; 2) doubling time (DT) at 2 months after IM DISC correlates
with TFR1 failure.
Aims: Here, we report the final result of TFR2 rate after DA DISC. The null hypothesis
was a TFR2 rate of 17.5% while the alternative hypothesis was a TFR2 rate of 35.0%,
and the study was designed to reject our null hypothesis if > 28% of pts remain in
TFR2 after DA DISC.
Methods: This prospective clinical trial (BMS CA180-543, NCT#02268370) had 3 phases:
1) IM DISC phase, 2) DA rechallenge phase, 3) DA DISC phase. Key inclusion criteria
included: 1) CML in chronic phase at original diagnosis, 2) total duration of IM therapy
of minimum 3 years, 3) total duration of MR4.5 or deeper response over 2 years. Molecular
relapse was defined as an increase in BCR-ABL qPCR > MR4 on 2 consecutive occasions,
or a single increase in BCR-ABL qPCR > MR3. DA treatment was started at 100mg once
daily after molecular relapse was confirmed, and continued for at least 12 months
after achieving ≥ MR4 until DISC for TFR2.
Results: The study was launched on March 2015 and completed all participants’ planned
visits as of Feb 2022 with a median follow-up duration of 27.5 months (range 2-51
months).
1) In the IM DISC phase, 58 of 131 pts (44.3%) experienced molecular relapse with
a median onset of 3.53 months, thus the remaining 73 pts achieved TFR1 (55.7%): The
mRFS rate at 12 months was 56.8% (95% CI, 47.8-64.8 %). Distribution of monthly DT
in 6 months is presented in Fig A.
2) In the DA rechallenge phase, out of the 58 pts who failed TFR1, 51 pts received
DA. At 3 months, 98.0%, 87.9%, and 75.4% of pts achieved MMR, MR4 and MR4.5. The halving
time (HT) after DA re-therapy is summarized in Fig B. Notably, HT at 2 and 3 months
was 10.6 and 10.7 days, consistent with rapid reduction of BCR-ABL qPCR in first 3
months.
3) In the DA DISC phase, 35 pts who attained MR4 or deeper response for ≥ 12 months
discontinued DA for a TFR2 attempt. Out of 35 pts, only 4 pts (11.4%) has maintained
the molecular response at last follow-up, while the remaining 31 pts lost the molecular
response with a median 3.65 months. The actuarial mRFS rate at 6 and 12 months was
22.9% (10.8-37.6%) and 10.0% (2.7-23.1%). Monthly DT within 6 months after DA DISC
is presented in Fig C. Based on the final result of 11.4% TFR2 rate, we conclude that
12 months’ DA re-therapy could not improve TFR2 rate significantly.
4) For DT after IM DISC, average DT at 2/3 months was much shorter in those lost molecular
response (10.6 / 10.7 days) than those who did not (23.5 / 30.1 days). For DT after
DA DISC, while average DT was 7.6 and 14.3 days at 2/3 months in overall population,
it was much shorter in those lost molecular response within 6 months (3.2 and 13.3
days at 2/3 months).
Image:
Summary/Conclusion: The final result indicates that re-challenge with DA after TFR1
failure with IM DISC is effective in restoring deep molecular response as most cases
rapidly regained at least MR4. However, 12 months’ DA re-therapy does not significantly
improve TFR2 rate. Further studies should consider increasing MR4 duration before
TFR2 attempt, adding other therapeutics and refining risk factor for TFR2 failure.
S159: QUÉBEC CML RESEARCH GROUP ANALYSIS OF TREATMENT PATTERNS IN CHRONIC MYELOGENOUS
LEUKEMIA: SWITCHING IS DRIVEN BY INTOLERANCE AND SIMILAR ACROSS TYROSINE KINASE INHIBITORS
AND LINES OF TREATMENT
L. Busque1 2,*, M. Harnois2, N. Szuber1 2, R. Delage2 3, L. Mollica1 2, H. Olney2
4, P. Laneuville2 5, S. Sirhan2 6, G. Cournoyer2 7, I. Chamakhi2 8, M. Lalancette2
3, D. Talbot2 9, V. Éthier2 10, P. Desjardins2 11, S. Assouline2 6
1Hematology, Hôpital Maisonneuve-Rosemont, Université de Montréal; 2Groupe Québécois
de Recherche en LMC-NMP, GQR-LMC-NMP, Montreal; 3Hematology, Centre Hospitalier Universitaire
de Québec (CHUQ), Université Laval, Québec; 4Hematology, Centre Hospitalier Universitaire
de Montréal (CHUM), Université de Montréal; 5Hematology, McGill University Health
Center (MUHC), McGill University; 6Hematology, Jewish General Hospital, McGill University,
Montreal; 7Hemato-oncology, Hôpital regional de St-Jérôme, St-Jérôme; 8Hemato-oncology,
Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montreal; 9Hemato-oncology,
Hôpital de la Cité-de-la-Santé, Laval; 10Hemato-oncology, Centre Hospitalier Universitaire
de Sherbrooke (CHUS), Université de Sherbrooke, Sherbrooke; 11Hemato-oncology, Hôpital
Charles-Lemoyne, Université de Sherbrooke, Greenfield Park, Canada
Background: In 2001, imatinib was the first tyrosine kinase inhibitor (TKI) approved
for the treatment of chronic myelogenous leukemia (CML) which translated into a revolution
in the management of this disease. The arsenal of TKIs was progressively reinforced
with the addition of other TKIs such as dasatinib, nilotinib, bosutinib, ponatinib
and recently asciminib. Two decades of clinical trials have provided critical insight
into differential efficacies and specific toxicity profiles of these TKIs supporting
the development of guidelines such as the European Leukemia Net (ELN) and the national
comprehensive cancer network (NCCN). Real-world evidence studies provide unique and
complementary information on treatment patterns, efficacy, side effects and may help
identify unmet medical needs of CML patients
Aims: This study was aimed at studying frequency of TKI switching, reason for switch,
duration of treatment without switching as a function of line of treatment and specific
TKI using the real-world data of the Québec registry created in 2009
Methods: Patients with Philadelphia positive (Ph+) CML were recruited with informed
consent in the Québec registry. 795 patients were included in this analysis. Data
from the registry were extracted July 31ist 2021. Switching was defined as a change
of a specific TKI to another. Reasons to initiate switching were categorized as resistance
(primary and secondary) or intolerance (hematological or non-hematological). Standard
statistical methods were used to evaluated bivariate and continuous variables. Kaplan-Meier
curve were used to evaluate overall survival and survival without switching (JMP®
Pro 14.1 software, SAS Institute, Cary, NC, USA).
Results: At the time of data collection, the median time of follow-up was 7.5 years.
Proportion of switching per line of treatment: 1st: 357/795 (44.9%); 2nd: 157/357
(43,9%); 3rd: 59/157 (37,6%); 4th:23/59 (39%). The reason for switching (ratio intolerance/resistance)
for each line were: 1st: 1.33; 2nd: 4.3; 3rd: 2.4; 4th: 2.7. 20 patients switched
serially across all lines for intolerance and only 3 for serial resistance. Survival
without switching was evaluated using a Kaplan-Meier curve by line of treatment and
by specific TKI (Figure 1). The survival without switching was similar for patients
on imatinib, dasatinib or nilotinib in first line as in second line. In third line,
there was no difference between imatinib, nilotinib, dasatinib or bosutinib although
numbers of patients were small. We then compared the survival from diagnosis of patients
that remained in first or second line versus the patients that reached third or more
lines of treatment. Although the mean age at diagnosis and SD were identical between
the two groups (54,8 years and SD 15,2) there was a statistically significant difference
in survival in favor of patients needing only 1 or 2 lines of treatment (P= 0.0254).
Image:
Summary/Conclusion: We demonstrate: (i) that switching of TKI is frequent and mainly
driven by intolerance in all lines of treatment; (ii) serial intolerance is 6.6 time
more frequent than serial resistance suggesting a class effect for intolerance in
some patients; (iii) all TKIs have a similar «retention level» in all lines of treatment;
(iv) that patients necessitating 3 or more lines of treatment have a survival disadvantage.
Our results suggest that one of the most important unmet medical need in CML management
is availability of better tolerated drugs
S160: MOLECULAR DETERMINANTS OF DISEASE PROGRESSION AFTER HYPOMETHYLATING AGENT THERAPY
IN RAS PATHWAY MUTANT CHRONIC MYELOMONOCYTIC LEUKEMIA AT THE SINGLE-CELL LEVEL
G. Montalban-Bravo1,*, F. Ma2, I. Ganan-Gomez1, R. Kanagal-Shamana3, V. Adema1, N.
Thongon1, H. Yang1, K. A. Soltysiak1, C. Bueso-Ramos3, H. Kantarjian1, G. Garcia-Manero1,
S. Colla1
1Leukemia, The University of Texas MD Anderson Cancer Center, Houston; 2Molecular
Biology Institute, University of California, Los Angeles; 3Hematopathology, The University
of Texas MD Anderson Cancer Center, Houston, United States of America
Background: Most patients (pts) with chronic myelomonocytic leukemia (CMML) have incomplete
or transient responses to hypomethylating agent (HMA) therapy. CMML cases driven by
mutations in RAS pathway signaling genes or ASXL1 have a higher risk of failure and
progression to acute myeloid leukemia (AML). Development of effective alternative
therapies has been delayed, owing to an incomplete understanding of how different
hematopoietic populations contributes to disease maintenance and progression.
Aims: We aimed to dissect the cellular and molecular mechanisms underpinning CMML
maintenance and progression in RAS mutant CMML.
Methods: We performed single-cell RNA sequencing (scRNA-seq) analysis of lineage-negative
(Lin-) CD34+ hematopoietic stem and progenitor cells (HSPCs) and BM mononuclear cells
(MNCs) isolated from RAS pathway mutant CMML pts (n=5 and 6, respectively) and age-matched
healthy donors (HD; n=2 and 3, respectively). CMML samples were obtained at the time
of diagnosis and HMA failure. Additionally, we performed scATAC-seq analysis of Lin-CD34+
HPSCs isolated at the times of diagnosis and progression (n=1).
Results: Our analysis revealed that CMML HSPCs had a predominantly granulomonocytic
differentiation route with increased frequencies of myeloid-monocytic progenitors,
at the expense of hematopoietic stem cells (HSCs) (Fig 1a), and upregulated expression
of genes involved in the oxidative phosphorylation, type I interferon (IFN) and IFNg
pathways. Consistent with these results, scRNA-seq analysis of MNCs revealed expanded
populations of myelomonocytic progenitors and monocytes and upregulated expression
of genes involved in IFNg response and NF-kB activation (Fig 1b), along with upregulation
of the NF-kB transcriptional effector BCL2A1 (Fig 1c). Assessment of ligand-receptor
interactions using the CellPhoneDB repository identified that CMML monocytes established
a high number of cell-cell interactions (n=638) with dendritic cells, NK cells, and
HSPCs via chemokines, cytokines, and inhibitory molecules known to induce NF-kB signaling
and NK-cell exhaustion. Disease progression was associated with expansion of lympho-myeloid
progenitors (LMPPs) (Fig 1d) characterized by the highest levels of IFNg response,
NF-kB survival signaling, and cell cycle regulators. scATAC-seq of Lin-CD34+ confirmed
higher activity of transcriptional factors associated with monocytic differentiation
and NF-kB signaling (Fig 1e-f). Accordingly, scRNA-seq analysis of MNCs showed increased
frequencies of HSPCs and myelomonocytic precursors, a reduction of T cells (Fig 1f),
and emergence of a monocyte population characterized by the highest expression of
NF-kB signaling and its effectors MCL1 and BCL2A1. BCL2A1 protein expression at progression
was confirmed by immunohistochemistry (Fig 1g). CellPhoneDB analysis identified a
high number of cell-cell interactions (n=2978) involving cytokines, chemokines, and
surface proteins known to elicit NF-kB activation and immune evasion between expanded
monocytes, LMPPs, myelomonocytic precursors, and immune cells during progression.
Image:
Summary/Conclusion: Our data suggests that CMML is maintained through metabolically
active HSPCs, which leads to monocytes’ reprograming and survival through NF-kB signaling
activation. We showed that disease progression arises from the expansion of NF-kB
dependent immature myeloid progenitors, which leads to therapy resistance and immune
evasion. This study has implications for the development of therapies targeting downstream
effectors of NF-kB–mediated survival pathway to overcome treatment failure. In vitro
validation is ongoing.
S161: ZRSR2 AND TET2 MUTATIONS PROMOTE MDS BY DYSREGULATING GENE EXPRESSION AND ABERRANT
ALTERNATIVE SPLICING IN MICE
C. Garcia-Ruiz1,*, C. Martínez-Valiente1, A. Liquori1, A. Gutiérrez-Adán2, J. Cervera3,
A. Sanjuan-Pla1
1Hematology Research Group, IIS La Fe, Valencia; 2Animal Reproduction Department,
INIA, Madrid; 3Genetics Unit, Hospital Universitario y Politécnico La Fe, Valencia,
Spain
Background: Mutations in splicing factors and epigenetic regulators are the most frequent
genetic alterations in patients with myelodysplastic syndromes (MDS). The minor spliceosome
factor ZRSR2 and the epigenetic regulator TET2 appear significantly associated in
MDS patients. However, the functional impact of such mutations in the hematopoietic
system and MDS have been scarcely studied. To address this question, we established
a murine model (Zrsr2
m/m
Tet2
−/−
) carrying mutations in both genes, which exhibited signs compatible with MDS disease
in mice. However, the molecular disease mechanism has not yet been elucidated.
Aims: To interrogate the impact of ZRSR2 and TET2 mutations in gene expression and
alternative splicing in the context of hematopoiesis and MDS.
Methods: Whole transcriptome sequencing was performed to investigate changes in gene
expression and alternative splicing patterns. Lin-Sca-1+c-kit+ (LSK) cells (50,000
per sample) were sorted from pools of 3 mice, and mRNA was sequenced in an Illumina
NovaSeq 6000 platform. Differential gene expression was determined using DESeq2 with
adjusted P-value ≤ 0.05, log2FoldChange ≥ 0. Functional enrichment from differentially
spliced genes was identified with KEGG, and the enrichment cut-off p-value was set
as adjusted P-value ≤ 0.05. Relevant alternative splicing events were validated by
RT-PCR.
Results: RNA-seq analysis of Zrsr2
m/m
Tet2
−/−
cells revealed 2952 differentially expressed genes (DEG), compared to 571 in Zrsr2m/m
and 1203 in Tet2−/−. From 2952, 1327 were up-regulated and 1625 were downregulated.
Interestingly, relevant genes for hematopoietic lineage specification were significantly
dysregulated. In particular, genes related to lymphoid lineage (Flt3, Notch1, and
Tlr7) were downregulated, while those related to myeloid lineage (Ccl9, Mpo, Ms4a6b,
and Prtn3) and megakaryocytic-erythroid lineage (Pdfgrb, Itgb3, Optn, and Tgfbr3)
were upregulated. This suggests an early myeloid and megakaryocyte-erythroid priming
of LSK towards the production of cells from these lineages in Zrsr2
m/m
Tet2
−/−
mice. Enrichment analysis of DEG using Go enrichment analysis identified ribosome
function, inflammation, and migration/motility processes as the most significantly
altered. Further, KEGG pathway enrichment pointed ribosome, the MAPK family, and pro-inflammatory
pathways as significantly enriched in Zrsr2
m/m
Tet2
−/−
cells. Finally, KEGG pathway enrichment of alternative splicing targets identified
the MAPK family and the Fanconi anemia pathway as the most altered targets in Zrsr2
m/m
Tet2
−/−
cells. Importantly, a total of 9 genes related to the MAPK pathway were identified
as mis-spliced, from which Dusp1, Tgfbr2, and Fgf11 were validated in this study.
Summary/Conclusion: In this study, we broaden our previous report and show that concurrent
mutations in Zrsr2 and Tet2 dysregulate normal gene expression and cause aberrant
mRNA splicing. Gene expression analysis identified ribosome function, inflammation,
and migration/motility as the most altered processes in Zrsr2
m/m
Tet2
−/−
cells. Alternative splicing analysis identified the MAPK and the Fanconi Anemia pathway
as key targets of aberrant splicing. All in all, gene expression dysregulation and
aberrant mRNA splicing disturb important biological pathways and drive the molecular
pathomechanism in Zrsr2
m/m
Tet2
−/−
mice.
S162: SOMATIC GENETIC LANDSCAPE IN GATA2 DEFICIENCY PATIENTS
L. Largeaud1 2,*, M. Collin3, N. Monselet4, F. Vergez5, L. Larcher6, P. Hirsch7, N.
Duployez8, J. Bustamante9, C. Bellanné-Chantelot10, J. Donadieu11, F. Sicre de Fontbrune12,
M. Nolla13, C. Fieschi14, F. Delhommeau7, E. Delabesse15, M. Pasquet13
1Clinical Haematology laboratory, Toulouse Hospital; 2UMR1037, Cancer Research Center
of Toulouse, Toulouse, France; 3Haematology Department, Institute of Cellular Medicine
Newcastle University, Newcastle, United Kingdom; 4Bureau des essais cliniques, Claudius
Rigaud Institut; 5Haematology Department, Toulouse Hospital, Toulouse; 6Clinical Haematology
laboratory, Saint Louis Hospital APHP; 7Clinical Haematology laboratory, Saint-Antoine
Hospital, APHP, Paris; 8Clinical Haematology laboratory, CHU Lille, Lille; 9Unité
centre d’études des déficits immunitaires, Necker Hospital APHP; 10Centre de génétique
moléculaire et chromosomique, Hôpital Pitié-Salpêtrière, APHP; 11Haematology Department,
Trousseau Hospital, APHP; 12Service d’hématologie greffe, Saint-Louis Hospital, APHP,
Paris; 13Service d’hématologie et immunologie pédiatrique, Toulouse Hospital, Toulouse;
14Immunologie clinique, Saint-Louis Hospital, APHP, Paris; 15Clinical Hematology laboratory,
Toulouse Hospital, Toulouse, France
Background: Heterozygous germline GATA2 mutations strongly predispose to myeloid malignancies,
immunodeficiency, and/or lymphedema. The progression towards haematological diseases
seems to be correlated with the acquisition of molecular and cytogenetic abnormalities.
Aims: Our study therefore focuses on the biological characterization of the different
progression stages of germline GATA2 deficiency patients and their correlation with
clinic features.
Methods: We describe here a cohort of 78 patients (62 from the French-Belgian cohort
and 16 British patients). Molecular analysis by next generation sequencing (targeting
90 genes frequently mutated in myeloid malignancies with a sensitivity = 1%) was performed
in 76 patients. Cytogenetic analyses were determined for 76 patients.
Results: Median age of our cohort was 21.8 years [6months–62years]. 44 had missense
mutations, 32 had null mutations, one intronic and one synonymous mutation. Our results
showed a trend toward a younger age at diagnosis in patients harboring null mutations
compared to patients with missense mutations (13 vs 17 years, p=0,086) and more chronic
infections during follow-up (74% vs 25%, p<0,001).
55 karyotypic abnormalities were identified including monosomy 7 (29%), trisomy 8
(16%) and der (1;7) (9%). Moreover, 141 somatic mutations were identified targeting
STAG2 (38%), ASXL1 (13%), SETBP1 (8%), EZH2 (4%) and RUNX1 (3%) genes.
To better stratify patients, we defined 3 spectra based on morphological bone marrow
analysis: 12 (15%) patients had spectrum 0 (normal bone marrow), 47 (60%) had spectrum
1 (hypoplastic marrow and/or low-grade myelodysplasia (MDS)) and 19 (25%) were classified
as spectrum 2 (MDS with excess blasts, AML and CMML). We found a genotype-phenotype
correlation: spectrum 1 is more associated with null mutations and spectra 0 and 2
with missense mutations (p=0.023). Patients in spectrum 0 exhibited no acquired molecular
and karyotypic abnormalities. Spectrum 2 was enriched with mutations of SETBP1, RAS
pathway genes, and RUNX1 as well as more other cytogenetic abnormalities excluding
-7, tri8 and der(1;7) (p<0,001). The proportion of monosomy 7 was higher in spectrum
2 than in spectrum 1 without reaching the significance level (47% vs 28%).
Conversely, STAG2 mutations were mainly found in spectrum 1. 53 STAG2 mutations were
identified in 25 patients (1 to 8 mutations per patient), mostly at low mutated cell
fraction (median: 6%). Follow-up of 3 patients with STAG2 clones in spectrum 1 showed
no progression toward spectrum 2. Moreover, clonal hierarchy of 3 patients with STAG2
mutations in spectrum 2 showed that STAG2 mutations were subclonal events. Our results
suggested that STAG2 mutation did not act as driver and could be related to clonal
hematopoiesis. Interestingly, Stag2 KO mice showed an enrichment of GATA2 binding
motif (Ochi et al. Cancer Discov 2020) which could lead to an increase of GATA2 activity.
We speculate that in GATA2 deficiency syndrome, STAG2 mutations could induce a mechanism
of indirect Somatic Genetic Rescue (SGR) by compensating the loss of GATA2 activity
induced by the mutated allele.
Image:
Summary/Conclusion: This study reported that STAG2 mutations were recurrent in GATA2
deficiency clonal hematopoiesis. Some genetic abnormalities are associated with the
leukemic transformation stage such as SETBP1, RAS pathway genes and other cytogenetic
abnormalities. An improved ability to identify patients with high risk of developing
leukemic transformation has the potential to improve clinical outcomes and help clinicians
determine the optimal timing of bone marrow transplantation.
S163: EFFICACY OF JAK 1/2 INHIBITION IN MURINE IMMUNE BONE MARROW FAILURE
E. Groarke1,*, X. Feng1, N. Aggarwal1, A. L. Manley1, Z. Wu1, S. Gao1, B. Patel1,
J. Chen1, N. Young1
1Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, United States
of America
Background: Immune aplastic anemia (AA) is a severe blood disorder characterized by
cytotoxic T-lymphocyte mediated stem cell destruction. Therapies including hematopoietic
stem cell transplant and immunosuppression are effective but entail costs and risks,
and are not effective or possible in all patients. The Janus Kinase (JAK) 1/2 inhibitor
ruxolitinib (RUX) suppresses cytotoxic T cell activation and inhibits production of
interferon gamma and tumor necrosis factor alpha in models of graft-versus-host disease.
Aims: Assess RUX in murine immune AA for potential therapeutic benefit.
Methods: In a murine major histocompatibility complex mismatched C67BL/6(B6) to CByB6F1
lymph node (LN) cell infusion AA model, and a C.B10 minor histocompatibility antigen
mismatched B6 to C.B10 LN cell infusion AA model, RUX was administered as a food additive
(Rux-chow), which achieves therapeutic levels in 48-72 hours after administration.
Control BMF mice received chow without RUX. Animals were fed with Rux-chow two days
before LN cell infusion as a prophylaxis (BMF+RUXD-2), or two days after LN cell infusion
as therapy (BMF+RUXD+2). Recipient mice were either bled and euthanized at day 14
following LN infusion to collect tissues or were kept for 56 days to record animal
survival. Blood counts were measured biweekly. Samples for flow cytometry, histology,
and gene expression assays were collected.
Results: In both AA murine models, RUX attenuated bone marrow hypoplasia, ameliorated
peripheral blood pancytopenia, and prevented mortality when used either prophylactically
or therapeutically. Treated mice had significantly higher blood counts (neutrophils,
red blood cells, hemoglobin, platelets) as well as bone marrow (BM) and RBM (residual
BM, excluding T-cells), and cellularity relative to control BMF mice. RUX suppressed
infiltration, proliferation and activation of effector T cells in the bone marrow
and mitigated Fas-mediated apoptotic destruction of target hematopoietic cells. All
mice who received RUX were alive at 56 days while control BMF mice all died (Figure
1). With the exception of two mice with low RBC at day 56, discontinuing Rux-chow
at day 28 or day 42 did not affect animal blood counts measurements, nor survival.
Gene expression in mice who received RUX revealed downregulated T cell function and
JAK/STAT pathway-related genes (Stat1, Stat3, Stat4, Fas, Ly6a, Infg, Gzmb, Gzma,
Gzmk, Infgr1, Il2rb, Il2rg, and Lag3). On network analysis of differentially expressed
genes in downregulated pathways, Stat1 and Ifn-g genes were at the center of the network,
connecting immune responses and cell cycle pathways. When toxicity was assessed, RUX
exerted modest suppression of lymphoid and erythroid hematopoiesis in normal and irradiated
CByB6F1 mice, but the drug showed impressive clinical efficacy despite this.
Image:
Summary/Conclusion: RUX showed striking therapeutic efficacy, improving blood counts
and prolonging survival in two different BMF murine models. In patients, clonal T
cell expansion and activation, IFN-g and TNF-α upregulation, and Fas mediated cell
death, lead to severe BM destruction and the development of AA. Our study demonstrated
that JAK 1/2 inhibition with RUX produced its suppressive effects by inhibiting T
cell activation, reducing inflammatory cytokine secretion, and limiting FasL/Fas-mediated
BM destruction. Given these results, JAK 1/2 inhibition with RUX is an exciting and
novel potential therapy for BMF patients based on a well characterized mechanism of
action.
S164: CLONAL DYNAMICS, IMMUNE PHENOTYPES, AND TARGETS OF BONE MARROW-INFILTRATING
T CELLS IN ACQUIRED APLASTIC ANEMIA
A. Ben Hamza1,*, C. Welters1, M. Brüggemann2, K. Dietze1, L. Bullinger1 3, T. H. Brümmendorf4
5, J. Strobel6, H. Hackstein6, K. Dornmair7, F. Beier4 5, L. Hansmann1 3
1Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin
Berlin, Berlin; 2Department of Medicine II, Hematology and Oncology, University Hospital
Schleswig Holstein, Kiel; 3German Cancer Consortium (DKTK), partner site Berlin, and
German Cancer Research Center (DKFZ), Heidelberg; 4Department of Hematology, Oncology,
Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University;
5Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen;
6Department of Transfusion Medicine and Haemostaseology, University Hospital of Erlangen,
Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen; 7Institute of Clinical
Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilian-University
Munich, Munich, Germany
Background: Acquired aplastic anemia is a hematological disease characterized by hypocellular
bone marrow with insufficient hematopoiesis and pancytopenia. High response rates
to immunosuppressive therapy with anti-thymocyte-globulin and cyclosporin A suggest
a key role for T cells in disease pathogenesis. However, bone marrow-infiltrating
clonal T cell expansion, associated immune phenotypes, and T cell targets remain poorly
understood.
Aims: To defined clonal T cell expansion, associated immune phenotypes, and potential
T cell targets in bone marrow specimens of aplastic anemia patients before and after
immunosuppressive therapy.
Methods: Within bone marrow of 15 patients with acquired aplastic anemia, we determined
T cell clone-associated immune phenotypes by multi-parameter flow cytometry single
cell index sorting for subsequent T cell receptor (TCR) αβ and cytokine/transcription
factor sequencing. T cell clones of 10 patients were monitored before and after immunosuppressive
therapy (median time span: 9 months) by TCRβ repertoire sequencing. Twenty-seven TCRs
of expanded clones were re-expressed in reporter cell lines to determine recognition
of autologous hematopoietic precursor cells.
Results: We detected oligoclonal T cell expansion in all patients and 92.9% of all
expanded clones were CD8+, while only 9.6% were CD4+. Frequencies of dominant T cell
clones varied between patients (1.1-20.6% of CD8+ T cells). Expanded CD8+ clones were
almost exclusively CCR7- PD1- TIM3- CD39- and frequently expressed cytokines and transcription
factors associated with cytotoxic effector differentiation (IFNG, GZMB, PRF1, TBX21).
More than 50% of the CD45RA+ clones (TEMRA) were CD57+, while CD45RA- clones (TEM)
were mainly CD28+ and smaller in size. While TEM clones showed similar frequencies
in bone marrow and peripheral blood, TEMRA displayed higher localization-dependent
differences in size. Interestingly, almost all (91.1%) expanded clones with frequencies
>1% of all T cells persisted in the bone marrow after immunosuppressive therapy independent
of clinical response. Similarly, highly expanded CD4+ clones, albeit low in numbers,
were stable in frequencies before and after therapy. To determine targets of CD8+
T cell clones with strong expansion, we re-expressed twenty-seven TCRs of eight patients
and found five to be specific for immunodominant epitopes of cytomegalovirus. Strikingly,
two TCRs recognized hematopoietic progenitor cells expanded from CD34-enriched autologous
bone marrow.
Summary/Conclusion: We tracked T cell clonotypes and associated immune phenotypes
under immunosuppressive therapy at the single cell level. Expanded CD8+ clones showed
cytotoxic effector differentiation and persisted in the bone marrow after immunosuppressive
therapy even in situations of therapeutic response, challenging the hypothesis that
clinical response follows the decline of highly expanded autoreactive T cell clones.
We identified two expanded T cell clones that recognized hematopoietic progenitor
populations providing experimental proof of concept that expanded autoreactive T cell
clones can be critically involved in aplastic anemia pathogenesis.
FB and LH contributed equally.
S165: INTEGRATED GENETIC DIAGNOSTICS OF PATIENTS WITH EARLY ONSET OF DE NOVO MYELODYSPLASTIC
SYNDROMES.
E. Attardi1,*, L. Tiberi2, D. Formicola3, R. Artuso3, V. Santini1
1MDS Unit, Hematology, AOU Careggi - Department of Experimental and Clinical Medicine;
2Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University
of Florence; 3Medical Genetics Unit, Meyer Children’s University Hospital, Florence,
Italy
Background: Cytogenetic and molecular alterations determine myelodysplastic syndrome
(MDS) prognosis and must be evaluated during disease history. Relevance of germline
(GM) predisposition in MDS was stressed in WHO classification, but its actual incidence
is probably underestimated. Recently, the widespread availability of large-scale genomic
sequencing techniques has facilitated the investigation of GM variants, and the identification
of novel GM variants predisposing to MDS is expected. In de novo MDS patients (pts)
with an early onset of the disease (age ≤55 yrs) when no syndromic signs drive the
genetic assessment, whole exome sequencing (WES) analysis of bone marrow (BM) and
saliva can constitute an optimal diagnostic procedure, improving the performance of
targeted next-generation sequencing (t-NGS) approach.
Aims: We adopted integrated diagnostics to define the cytogenetic profile, and WES
to investigate possible GM predisposition of a cohort of 30 de novo MDS pts with an
unusual young age at disease onset (≤55 yrs). Standard karyotype (SK) was paralleled
by low coverage whole genome sequencing (lc-WGS) on plasma cell-free DNA (cf-DNA),
as non-invasive screening test for detection of structural chromosome abnormalities.
Methods: Patient BM-DNA / peripheral blood DNA (PB-DNA) was analysed by WES and compared
to DNA extracted from saliva (S-DNA) samples as control for GM variants. To identify
high-confidence MDS variants, we attributed an internal deleteriousness score to define
variants, from “of uncertain significance” to “pathogenic” (ACMG guidelines). Variants
were suspected as GM where variant allele frequency was > 0.3 and had been confirmed
for genes detected in both S-DNA and BM/PB-DNA. To confirm the somatic nature of some
variants, deep t-NGS of 27 myeloid custom panel gene was applied in 26/30 MDS cases
on BM-DNA. SK and lc-WGS (0.2x) on plasma cf-DNA were assessed in 27/30 pts.
Results: The small cohort of “young” MDS pts here analysed (Table 1). The majority
of pts, 17/30 (57%) presented at least one high-confidence variant evaluated as GM
for MDS. A total of 26 GM variants were recognized. Patients could be grouped on the
basis of the type of variant as follows: 11/30 pts (37%) presented variants in genes
involved in DNA repair defects and cancer predisposition (ATM, ATR, FANCA, FANCM,
PARN, BRCA1, BRCA2, CHEK2); 4/30 (13%) had variants involved in genetic predisposition
to myeloid neoplasm - GATA2, ANKRD26 and RBBP6 - the latter classically associated
to MPN familial cases; one presented compound heterozygous variants in SBDS; 4 presented
variants related to hereditary red blood cell defects. In our cohort, we identified
cases with a complex mode of inheritance (4/17) as well as high confidence GM variants
in 4 pts in 2 new interesting cancer predisposition genes: RBBP6 and PARN. Lc-WGS
of plasma cf-DNA confirmed all the cytogenetic alterations found by SK (8/27) and
identified a 21q22.12 deletion involving RUNX1 locus in a patient, not revealed by
SK.
Image:
Summary/Conclusion: By means of an integrated diagnostics, defined as the convergence
of diagnostic techniques with advanced information technology (WES, t-NGS, lc-WGS),
as a proof of principle, it was possible to fully characterize the genetic asset of
a particular subgroup of 30 MDS cases. We also demonstrated that lc-WGS of plasma
cf-DNA has an excellent sensitivity allowing to perform periodic cytogenetic evaluations
in MDS pts without invasive maneuvers. We showed that, by the use of WES, GM alterations
were demonstrated in a small cohort of “young” non-syndromic MDS pts in 57% of cases,
a proportion unexpectedly high.
S166: MAGROLIMAB IN COMBINATION WITH AZACITIDINE FOR PATIENTS WITH UNTREATED HIGHER-RISK
MYELODYSPLASTIC SYNDROMES (HR MDS): 5F9005 PHASE 1B STUDY RESULTS
D. A. Sallman1,*, M. M. Al Malki2, A. S. Asch3, E. S. Wang4, J. G. Jurcic5, T. J.
Bradley6, I. W. Flinn7, D. A. Pollyea8, S. N. Kambhampati9, T. N. Tanaka10, J. F.
Zeidner11, G. Garcia-Manero12, D. Jeyakumar13, L. Gu14, A. Tan14, M. Chao14, C. O’Hear14,
I. Lal14, P. Vyas15, N. Daver12
1Moffitt Cancer Center, Tampa; 2City of Hope National Medical Center, Duarte; 3Stephenson
Cancer Center, Oklahoma University Health, Oklahoma City; 4Roswell Park Comprehensive
Cancer Center, Buffalo; 5Columbia University Medical Center, New York; 6Sylvester
Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami;
7Tennessee Oncology, Nashville; 8University of Colorado School of Medicine, Denver;
9Sarah Cannon Research Institute, Kansas City; 10University of California San Diego
Moores Cancer Center, San Diego; 11Lineberger Comprehensive Cancer Center, University
of North Carolina, Chapel Hill; 12The University of Texas MD Anderson Cancer Center,
Houston; 13University of California Irvine, Orange; 14Gilead Sciences, Inc., Foster
City, United States of America; 15Weatherall Institute of Molecular Medicine, University
of Oxford, Oxford, United Kingdom
Background: Magrolimab is a monoclonal antibody that blocks CD47, a “don’t eat me”
signal overexpressed on cancer cells. CD47 blockade by magrolimab induces macrophage-mediated
phagocytosis of tumor cells and is synergistic with azacitidine (AZA) in the upregulation
of “eat me” signals. A high unmet need exists to build on current standard-of-care
AZA frontline therapy to increase efficacy while maintaining a tolerable safety profile
in patients (pts) with HR MDS.
Aims: To report the final safety/tolerability and efficacy data from a phase 1b trial
of magrolimab + AZA in pts with untreated HR MDS (NCT03248479).
Methods: Pts with previously untreated intermediate, high, or very high–risk MDS per
the Revised International Prognostic Scoring System (IPSS-R) received magrolimab IV
as a priming dose (1 mg/kg) followed by ramp-up to a 30-mg/kg QW or Q2W maintenance
dose. AZA 75 mg/m2 was administered IV or SC on days 1-7 of each 28-day cycle. Primary
endpoints were safety/tolerability and complete remission (CR) rate.
Results: A total of 95 pts (median age, 69 y [range, 28-91 y]) were treated. IPSS-R
risk was intermediate, high, or very high in 27%, 52%, and 21% of pts. MDS was therapy
related in 22% of pts; 26% had a TP53 mutation, and 62% had poor-risk cytogenetics.
Median number of cycles was 6 (range, 1-27). The most common treatment-emergent adverse
events (TEAEs) included constipation (68%), thrombocytopenia (55%), anemia (52%),
neutropenia (47%), nausea (46%), and diarrhea (44%). The most common grade 3/4 TEAEs
included anemia (47%), neutropenia (46%), thrombocytopenia (46%), and white blood
cell count decreased (30%). Six pts discontinued treatment due to AEs. The 60-day
mortality rate was 2%. Median hemoglobin change from baseline (BL) at first postdose
sample was −0.7 g/dL (range, −3.1 to 2.4 g/dL). CR and objective response (OR) rates
were 33% and 75%, with 31% of OR-evaluable pts with abnormal cytogenetics at BL having
cytogenetic CR. Median time to first OR, duration of CR (DCR), duration of OR, and
progression-free survival (PFS) were 1.9, 11.1, 9.8, and 11.6 mo. Overall survival
(OS) rates at 12 and 24 mo were 75% and 52%; median OS was not reached (NR) with 17.1
mo of follow-up (figure). In pts evaluated with sequential whole-exome sequencing
with a variant allele frequency (VAF) cutoff of 5%, 3 of 3 pts with TP53 mutation
who achieved CR had TP53 VAF <5% by cycle 5 day 1. Favorable outcomes were observed
both in pts with TP53 mutation (CR rate, 40%; median OS, 16.3 mo) and wild-type TP53
(CR rate, 31%; median OS, NR) (table).
Table.
Outcomes in patients with previously untreated HR MDS treated with magrolimab + AZA
Outcome
All patients
N=95
a
Wild-type TP53
n=61
TP53 mutation
n=25
OR rate, %
b
75
79
68
CR rate (95% CI), %
33 (23-43)
31 (20-44)
40 (21-61)
Marrow CR rate, %
32
38
20
Stable disease with HI rate, %
11
10
8
Median DCR (95% CI), mo
11.1 (7.6-13.4)
12.9 (8.0-NR)
7.6 (3.1-13.4)
Marrow CR rate with HI/any HI, %
17/59
20/61
12/56
Converted to red blood cell transfusion independence, %
14
10
24
Median PFS (95% CI), mo
11.6 (9.0-14.0)
11.8 (8.8-16.6)
11.0 (6.3-12.8)
Median OS (95% CI), mo
NR (16.3-NR)
NR (21.3-NR)
16.3 (10.8-NR)
a a 9 patients had missing TP53 status. b Defined as CR+PR + marrow CR+SD with HI.
HI, hematologic improvement.
Image:
Summary/Conclusion: Magrolimab + AZA was well tolerated with promising efficacy in
pts with untreated HR MDS, including those with TP53-mutated and –wild-type disease.
A phase 3 trial of magrolimab/placebo + AZA (ENHANCE: NCT04313881) is ongoing.
S167: PREDICTION OF RELAPSE AFTER ALLOGENEIC STEM CELL TRANSPLANTATION USING INDIVIDUALIZED
MEASURABLE RESIDUAL DISEASE MARKERS; THE PROSPECTIVE NORDIC STUDY NMDSG14B
M. Tobiasson1,*, T. Pandzic2, J. Illman3, L. Nilsson4, S. Weström2, K. Sollander2,
E. Ejerblad5, A. Olsnes Kittang6, G. Olesen7, O. Werlenius8, A. Björklund9, J. Wiggh1,
C. lindholm1, F. Lorentz10, B. Rasmussen11, J. Cammenga12, D. Weber13, D. Grönnås14,
M. Dimitriou15, S. Kytölä16, G. Walldin15, P. Ljungman9, K. Groenbeck17, S. Mielke9,
S. E. Jacobsen15, F. Ebeling3, L. Cavelier2, L. Smidstrup Friis17, I. Dybedal18, E.
Hellström-Lindberg1
1Hematology, Karolinska University Hospital, Stockholm; 2Department of Immunology,
Genetics and Pathology, Science for life, Uppsala, Sweden; 3Hematology, Helsinki University
Hospital, Helsinki, Finland; 4Hematology, Lund University Hospital, Lund; 5Hematology,
Akademiska sjukhuset, Uppsala, Sweden; 6Hematology, Haukeland University Hospital,
Bergen, Norway; 7Hematology, Arhus University Hospital, Århus, Denmark; 8Hematology,
Sahlgrenska University Hospital, Gothenburg; 9Centre for allogeneic stem cell transplantation,
Karolinska University Hospital, Stockholm; 10Hematology, Norrlands University Hospital,
Umeå; 11Hematology, Örebro University Hospital, Örebro; 12Hematology, Universitetssjukhuset,
Linköping, Sweden; 13Hematology, Odense University Hospital, Odense, Denmark; 14Institution
for environmental medicine; 15Centre for hematology and regenerative medicine, Institution
for medicine, Huddinge, Karolinska Institute, Stockholm, Sweden; 16HUS Diagnostic
Center, HUSLAB, Helsinki University Hospital, Helsinki, Finland; 17Hematology, Rigshospitalet,
Copenhagen, Denmark; 18Hematology, Oslo University Hospital, Oslo, Norway
Background: One third of patients with myelodysplastic syndrome (MDS) relapse after
allogeneic stem cell transplantation (HCT). Early detection of impending relapse would
enable pre-emptive treatment and potentially reduce relapse risk but is limited by
the lack of sensitive markers for measurable residual disease (MRD). We developed
a pipeline where patient-specific mutations, as determined by a myeloid next generation
sequencing (NGS) panel are tracked using digital droplet PCR (ddPCR).
Aims: To evaluate if personalized MRD detection by ddPCR can predict clinical relapse
earlier than conventional methods.
Methods: The prospective study (NCT02872662) enrolled patients with MDS, MDS/MPN or
MDS-AML with < 30% marrow blasts undergoing HCT. Patients were included before HCT,
and serial bone marrow (BM) samples were collected every third month post-HCT for
2 years. Peripheral blood (PB) samples were collected monthly. MRD results were not
available for the treating physician.
Results: We screened 286 pts between 2016 and 2020, whereof 20 were excluded mainly
due to lack of genetic aberration or no HCT performed. 266 pts were included from
12 HCT centers. Median age was 64 (18-78) years and 59% were male. Myeloid panel NGS
screening identified a median of 2 (0-9) mutations. The most common mutations were
TET2 (n=85), ASXL1 (n=73) and SRSF2 (n=59).
Median time of follow up was 886 (4-1934) days. Sixty pts relapsed after a median
of 189 (53-1281) days and 46 died due to non-relapse mortality after a median of 121
(4-1036) days. Remaining pts (n=160) were in continuous complete remission (CCR) after
a median follow-up of 1053 (479-1934) days. Estimated 1 and 2y overall survival was
79%, and 71%, respectively, while estimated 1 and 2y relapse-free survival (RFS) was
75% and 66%, respectively.
MRD data was missing in 46 pts; no post-HCT samples available (n=15), no mutation
detected (n=14) and difficulties to design ddPCR primers (n=11). 221 pts were available
for MRD analysis with a median number of 4 (0-13) and 5 (0-23) samples from BM and
PB, respectively.
Of 53 clinical relapses with MRD results available, 42 were preceded by pos MRD (>0.1%)
with a median of 70 (range 20-425) days between first pos MRD and clinical relapse.
For the 11 remaining pts, 8 were inadequately sampled with a median time of 189 (82-397)
days between last sampling and clinical relapse. One patient had an extramedullary
relapse only.
Of 31 pts who died without relapse, 19 were consistently MRD neg, while 5 were borderline
positive (MRD > 0.1% and <0.5%) during the first 100 days but negative thereafter.
Four MRD+ patients died without clinical relapse. Three pts were initially MRD+ but
turned negative, all of which had chronic GVHD (cGVHD).
Of 136 CCR patients, 94 were consistently MRD neg; 26 were borderline pos (MRD > 0.1%
and <0.5%) during the first 100 days followed by neg samples; 16 were MRD positive
(either > 0.5% during the first 100d or > 0.1% after 100d) of which 10 had a transition
from pos to neg samples (all had cGVHD); one patient was treated for a molecular relapse
detected by clinical routine method (FISH) and five patients were MRD positive at
time of last follow-up.
MRD used as a time-dependent co-variate was negatively associated with RFS (HR 7.1,
p<0.01). Estimated cumulative incidence of relapse and non-relapse mortality 2y after
pos MRD was 60% and 7% respectively (see figure).
Image:
Summary/Conclusion: We report the development of a highly functional personalized
MRD pipeline based on patient-specific mutations showing a high sensitivity to predict
relapse and relapse-free survival.
S168: ERYTHROPOIETIN STIMULATION AGENTS SIGNIFICANTLY IMPROVES OUTCOME IN LOWER RISK
MDS.
H. Garelius1,*, A. Smith2, T. Bagguley2, A. Taylor2, P. Fenaux3, D. Bowen4, A. Symeonidis5,
M. Mittelmann6, R. Stauder7, J. Čermák8, G. Sanz9, S. Langemeijer10, L. Malcovati11,
U. Germing12, R. Itzykson13, A. Guerci-Bresler14, D. Culligan15, I. Kotsianidis16,
K. Koinig Mag17, C. van Marrewijk18, S. Crouch2, T. de Witte19, E. Hellström-Lindberg20
1Section of Hematology, Specialist Medicine, Sahlgrenska University hospital, Göteborg,
Sweden; 2Department of Health Sciences, University of York, York, United Kingdom;
3Service d’Hematologie, Hôpital Saint-Loius, Assistance Publique des Hopitaux de Paris
(AP-HP) and Universite Paris; 7, Paris, France; 4St.James’s Institute of Oncology,
Leeds Teaching Hospitals, Leeds, United Kingdom; 5Department of Medicine, Division
of hematology, University of Patras Medical Scholl, Patras, Greece; 6Department of
Medicine, Tel Aviv Sourasky (Ichilov) Medical Center and Sackler Medical Faculty,
Tel Aviv University, Tel Aviv, Israel; 7Department of Internal Medicine V (Haematology
and Oncology), Innsbruck Medical University, Innsbruck, Austria; 8Dep. of Clinical
Hematology, Institute of Hematology & Blood Transfusion, Prague, Czechia; 9Department
of Haematology, Hospital Universitario y Politécnico La Fe, Valencia, Spain; 10Department
of Hematolog, Radboud university medical center, Nijmegen, Netherlands; 11Department
of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo,University of Pavia,
Pavia, Italy; 12Department of Haematology, Oncology and Clinical Immunology, Universitätsklinik
Düsseldorf, Düsseldorf, Germany; 13Service d’Hématologie, Hôpital Saint-Louis, Assistance
Publique des Hôpitaux de Paris (AP-HP) and Université Paris; 7, Pari, Paris; 14Service
d’Hématologie, Centre Hospitalier Universitaire Brabois Vandoeuvre, Nancy, France;
15Department of Haematology, Aberdeen Royal Infirmary, Aberdeen, United Kingdom; 16Dep.
of Hematology, Democritus University of Thrace Medical School, University Hospital
of Alexandroupolis, Alexandroupolis, Greece; 17Department of Internal Medicine V (Hematology
and Oncology), Medical University Innsbruc, Innsbruck, Austria; 18Department of Haematology,
Radboud university medical center; 19Department of Tumor Immunology - Nijmegen Center
for Molecular Life Sciences, Radboud university medical cente, Nijmegen, Netherlands;
20Department of Medicine, Div. Hematology, Karolinska Institutet, Stockholm, Sweden
Background: The EUMDS Registry started in 2008 as a prospective, non-interventional
longitudinal study, enrolling newly diagnosed patients with IPSS low or intermediate-1
MDS from 16 European countries and Israel.
Aims: The aim of the present analysis was to see how treatment with or without Erythropoietin
Stimulating Agents (ESAs) and/or red blood cell transfusions (RBCT) impact overall
survival (OS) and quality of life (QoL).
Methods: Patient management was recorded electronically every 6 months (“visit”) in
a central database, including treatment, transfusions, blood values, and health related
quality of life (HRQoL) using the EQ-5D 3-Level index and Visual Analog Scale (VAS).
Patients were eligible to be included in the analyses if their hemoglobin was recorded
as less than <10 g/dl at a visit. To overcome potential confounding by non-random
allocation of ESA treatment, propensity score matching was performed to ensure that
treated and untreated patients had similar characteristics. Only patients with comparable
propensity scores were included in the analyses to estimate the effects of ESA treatment
on outcomes using standard time to event
analyses; OS was estimated from the first visit a Hb value of <10g/dl was recorded.
OS was examined for patients treated with ESA stratified by their transfusion status
prior to commencing ESA treatment (no RBCT, <4 units, ≥4 units).
Patients were separated into 4 groups at each clinical visit, depending on the treatment
received in the interval leading up to that visit; no ESA nor RBCT, ESA only, ESA
and RBCT and RBCT only. HRQoL at each visit according to the treatment status was
summarized for patients who had completed a questionnaire at visit 1 and 2; mean values
were examined by treatment group.
Results: Of 2562 patients registered by November 2021, 2448 were diagnosed before
July 2019 and included in the analysis; these patients were divided into two groups:
ESA untreated (n=1265) and ESA treated (n=1183). Patients whose Hb remained above
10g/dl were excluded leaving 529 untreated patients and 749 ESA treated; after propensity
score matching was applied two comparable groups were produced: ESA untreated (n=
426) and ESA treated (n= 742). Median OS from reaching the eligibility criteria in
the ESA treated vs untreated groups were 44.9 and 34.8 months respectively (Fig 1a),
giving a clear survival advantage to the ESA-treated group. (p<0.003). In the ESA-treated
group, OS was poorer in those who had been transfused prior to commencing ESA (Fig
1b, p<0.001).
Fig 1c shows the number of patients at each visit who had been treated with transfusions
or ESA; 647/1278 had received neither at visit 1, the figure shows the “flow” of patients
by treatment for the first 6 visits. HRQoL was examined for the 695 patients who had
completed a questionnaire at both visit 1 and 2 up to visit 6; differences were seen
by treatment (Fig 1d). Patients who had received no treatment reported, on average,
the highest mean HRQoL, in contrast, patients who had RBCT had the lowest (p<0.001).
Image:
Summary/Conclusion: This unique large prospective registry study clearly shows a significant
survival advantage for lower-risk MDS patients exposed to ESA treatment at onset of
anemia (Hb <10g/dL) but before onset of transfusion therapy, strongly supporting recommendations
to start ESA treatment early. The effect on patients with an early transfusion need
warrants further studies. Moreover, ESA exposure is associated with maintained QoL,
while RBCT development with or without ESA exposure is associated with significantly
deterioration in QoL.
S169: CLINICAL AND MOLECULAR MARKERS FOR PREDICTING RESPONSE TO ROMIPLOSTIM TREATMENT
IN LOWER-RISK MYELODYSPLASTIC SYNDROMES
A. S. Kubasch1 2 3,*, A. Giagounidis2 3 4, G. Metzgeroth5, A. Jonasova6, R. Herbst7,
J. M. T. Diaz8, B. De Renzis9, K. S. Götze2 3 10, M.-L. Huetter-Kroenke11, M.-P. Gourin12,
B. Slama13, S. Dimicoli-Salazar14, P. Cony-Makhoul15, K. Laribi16, S. Park17, K. Jersemann18,
D. Schipp19, K. H. Metzeler20, O. Tiebel21, K. Sockel2 22, S. Gloaguen2 3, A. Mies22,
F. Chermat23, C. Thiede22, R. Sapena23, R. F. Schlenk24 25, P. Fenaux3 23 26, U. Platzbecker2
3 20, L. Ades3 23 26
1Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital
Leipzig; 2German MDS Study Group (D-MDS); 3The European Myelodysplastic Syndromes
Cooperative Group (EMSCO), Leipzig; 4Department of Oncology, Hematology and Palliative
Care, Marien Hospital, Düsseldorf; 5Department of Hematology and Oncology, University
Medical Centre, Mannheim, Germany; 61st Medical Department - Hematology, General Hospital,
Prague, Czechia; 7Medizinische Klinik III, Klinikum Chemnitz, Chemnitz, Germany; 8Department
of Hematology and Oncology, CHU de Poitiers, Poitiers; 9Service d’Hématologie Clinique
Adulte, Clermont Ferrand, France; 10Department of Medicine III, Technical University
of Munich, Munich; 11Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin
Berlin, Berlin, Germany; 12CHU Limoges, Limoges; 13Service d’Hématologie, Centre Hospitalier
d’Avignon, Avignon; 14University Hospital Bordeaux, Pessac; 15Centre Hospitalier Annecy-Genevois,
Pringy; 16Centre Hospitalier Du Mans, Le Mans; 17Department of Hematology, CHU Grenoble,
Grenoble, France; 18GWT-TUD GmbH, Dresden; 19DS-Statistics, Rosenthal-Bielatal; 20Department
of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig;
21Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technical
University Dresden; 22Department of Internal Medicine I, University Hospital Carl
Gustav Carus, Technical University Dresden, Dresden, Germany; 23Groupe Francophone
des Myélodysplasies, Paris, France; 24Department of Internal Medicine V, University
Hospital of Heidelberg; 25NCT-clinical trials office, German Cancer Research Center,
Heidelberg, Germany; 26Hématologie Clinique, Hôpital Saint-Louis, Paris, France
Background: In about half of patients with lower-risk (LR) myelodysplastic syndromes
(MDS), thrombocytopenia is present at the time of diagnosis, being associated with
shortened survival and a higher risk of progression to acute myeloid leukemia (AML).
Romiplostim (ROM), a thrombopoietin receptor agonist (TPO-RA), has shown safety and
clinical efficacy in prospective trials in LR-MDS. Post-hoc analyses have demonstrated
hematologic improvement of platelets (HI-P) after ROM treatment contingent on endogenous
thrombopoietin (TPO) levels and platelet transfusion events (PTE) (Sekeres et al.
BJH 2014).
Aims: The prospective EUROPE multicenter phase 2 trial (NCT02335268) investigated
the predictive value of biomarkers like endogenous TPO levels, PTE and molecular markers
on the clinical efficacy of single-agent ROM treatment within the `European Myelodysplastic
Syndromes Cooperative Group` (EMSCO) network. Patients with IPSS low or intermediate
1 risk were eligible if baseline bone marrow blast count was <5% (central morphology)
and platelet count was ≤30 Gpt/L or ≤50 Gpt/L in case of a bleeding history.
Methods: According to a previously published model of response to TPO-RA (Sekeres
et al. BJH 2014), patients were assigned into two different cohorts at the time of
screening based on previous PTE and centrally assessed TPO serum levels (cohort A:
TPO<500 ng/l and PTE<6 units/past year; cohort B: TPO>500 ng/l, and/or PTE36 units/past
year). The primary efficacy endpoint was the rate of HI-P according to IWG 2006 criteria
lasting for 38 weeks. ROM was initiated at a dose of 750 μg weekly by subcutaneous
injection, the dose was adjusted based on the patient`s platelet counts.
Results: From 2015 to 2019, a total of 77 patients were included at 29 different trial
sites in Germany, France and the Czech Republic. Regarding the primary endpoint, 32
out of 77 (42%) responded (HI-P) with a numerically higher response rate in cohort
A (47%, n=24) vs. cohort B (31%, n=8) (p=0.2953). At 16 weeks of ROM treatment, three
(4%) and seven (9%) patients had additional neutrophil (HI-N) and erythroid (HI-E)
responses, respectively. None of the patients achieved trilineage responses (HI-P,
HI-E and HI-N). Median duration of response was significantly longer for patients
in cohort A (351 days) compared to cohort B (315 days) (p=0.006, log-rank-test). Mutated
SRSF2 was significantly more frequent in responders (41%) compared to non-responders
(16%) (p=0.018, Fisher’s exact test). In patients with an SRSF2 mutation, the probability
to achieve HI-P was 65% compared to 33% inpatients with SRSF2 wildtype (Figure 1A).
Comparing responders vs. non-responders, we found no significant changes of variant
allelic burden of variants detected pre- and post-ROM (Figure 1B). Finally, we developed
a response prediction model to ROM therapy with the aim to improve personalized patient
stratification in the future. The percentage of correctly predicted HI-P was highest
for the model, which included the variables platelet count, SRSF2 mutation status
and the hemoglobin level using the threshold of 11.4 g/dl and resulted in an overall
accuracy of 70 % for a correct ROM response prediction (Figure 1A).
Image:
Summary/Conclusion: In conclusion, this prospective study confirms the efficacy and
overall safety of ROM in this subgroup of LR-MDS patients with thrombocytopenia. To
avoid overfitting of variables and to confirm our results, the here presented response
prediction model needs to be validated in an external independent cohort.
* U.P. and L.A. contributed equally to this study as senior authors
S170: DYNAMIC INTERPLAY BETWEEN TUMOR AND MICRO-ENVIRONMENT DURING MYELOMA DISEASE
PROGRESSION
M. C. Köse1,*, I. Bergiers2, M. Malfait3, B. Heidrich4, D. De Maeyer2, N. Fourneau2,
B. Verbist2, J. Van Houdt2, G. Vanhoof2, R. Verona4, M. Delforge5, J. Depaus6, N.
Meuleman7, J. Van Droogenbroeck8, P. Vlummens9, C. J. Heuck4, N. Bahlis10, J. Caers1,
T. Casneuf2
1Laboratory of Hematology, University of Liege, Liege; 2Janssen Research & Development,
Beerse; 3Department of Applied Mathematics, Computer Science and Statistics, University
of Ghent, Ghent, Belgium; 4Janssen Research & Development, Spring House, United States
of America; 5University Hospital Leuven, Leuven; 6Department of Hematology, CHU UCL
Namur, Yvoir; 7Service d’Hématologie, Université Libre, Brussels; 8Departmentof Haematology,
AZ Sint-Jan Brugge-Oostende AV, Brugge; 9Department of Clinical Hematology, Ghent
University Hospital, Ghent, Belgium; 10Department of Hematology and Oncology, University
of Calgary, Calgary, Canada
Background: Multiple Myeloma (MM) is an incurable plasma cell (PC) malignancy that
evolves from two premalignant stages: Monoclonal Gammopathy of Undetermined Significance
(MGUS) and Smoldering Multiple Myeloma (SMM). The disease progression has been characterized
to be driven by intrinsic genomic events in the myeloma cells and by gradual dysregulation
of the immune system.
Aims: We investigated how the interplay between tumor cells with their microenvironment
and the underlying complex and dynamic immune biology evolve during this process.
Methods: Single cell multi-omics profiling, including RNA, B-cell receptor (BCR) and
antibody barcode-tagged 10x sequencing, was conducted on human bone marrow (BM) aspirates
collected at 6 Belgian centers from 4 cohorts: 31 healthy elderly and 28 MGUS, 32
SMM and 32 newly diagnosed MM. Mononuclear cell isolation, freezing and transport
to central facilities was optimizedand data were integrated and filtered using Scanpy
and Scirpy. The main immune cell types were identified from the RNA and antibody data
using SingleR. Further functional subtyping was done using Leiden clustering. Differential
pathway expression analysis was performed with Muscat and FGSEA.
Results: From the tumor cell transcriptomes, our analyses confirmed the previously
documented myeloma molecular hallmarks, such as MYC and IFN-a signaling, cell proliferation,
energy metabolism and oxidative phosphorylation. Evidence was found for transcriptomic
similarities and within-and between-patient malignant PC transcriptomic heterogeneity,
as well as the existence of multiple transcriptomic clones in several patients. We
observed a positive correlation between the antigen processing mechanism in the PCs
with IFN response, suggesting that this mechanism associates with initiation of the
immune recognition and activation against the tumor.The gradually increasing differential
gene expression was also observed in the immune microenvironment: dysregulation of
signaling pathways initiates early in MGUS and spreads throughout the various cell
types surrounding the tumor cells. Cell population shifts were also found. In the
CD1C+ DCs, that play a role in cancer immune control, a functional shift was observed
that correlated with disease progression towards a more mature and antigen presenting
phenotype with higher levels of CD83, HBEGF, MCL1 and CXCL16 as well as increased
TNF-a pathway. Similarly, a shift was observed in the macrophage population, toward
M1 state showing high IFN response along with expression of MS4A4A, STAT1, TNFSF13B
and TRAIL in more severe disease. Interestingly, in the CD8+ T cells, we detected
a pre-dysfunctional subpopulation with high expression of GZMK, activation markers
CD69, CCL4, CXCR4 and genes associated with T cell pre-dysfunctionality NR4A2, RGS1,
TOX and TIGIT, that was found to be associated with progression (Figure). In the CD4+
cytotoxic T cells, a proportion change was observed with more severe disease.
Image:
Summary/Conclusion: With the atlas of healthy, precursor and active MM patient BM
samples, we generated a comprehensive and granular view of the various cell types
involved in disease progression and provide evidence for early and gradually increasing
immune dysregulation and activation of oncogenic driver pathways. Our data evidence
the co-divergence and reciprocal stimulation of transcriptomes of tumor and microenvironment
and support the postulation of microenvironment as a central modulator of cancer cell
growth, survival and metastasis.
S171: MOLECULAR CLUSTERS OF IGM MONOCLONAL GAMMOPATHIES PRESENT DISTINCT BIOLOGIC,
IMMUNE AND METABOLIC FEATURES
P. Mondello1,*, J. Paludo1, J. Novak1, K. Wenzl1, S. Jalali1, J. Krull1, E. Braggio2,
S. Dasari3, M Manske1, J. Abeykoon1, S. Vivekananda1, P. Kapoor1, A. Paulus4, C. Reeder2,
S. Ailawadhi4, A. Chanan-Khan4, R. Kyle1, M. Gertz1, Z.-Z. Yang1, A. Novak1, S. Ansell1
1Medicine, Mayo Clinic, Rochester; 2Medicine, Mayo Clinic, Phoenix; 3Bioinformatics,
Mayo Clinic, Rochester; 4Medicine, Mayo Clinic, Jacksonville, United States of America
Background: IgM MGUS and Waldenstrom Macroglobulinemia (WM) represent a wide range
of conditions whose management varies from observation to immunochemotherapy. The
current classification relies solely on clinical features and does not explain the
heterogeneity that exists within each of these conditions.
Aims: To shed light on the biology that may account for the clinical differences,
we performed the first comprehensive multi-omics analysis of IgM monoclonal gammopathies.
Methods: We used bone marrow (BM) CD19+CD138+ sorted cells and matched BM plasma from
32 pts (7 IgM MGUS, 25 WM) and 5 healthy controls to perform whole exome sequencing,
RNA-seq, proteomic and metabolomic analysis. 7 matched WM and 4 IgM MGUS pts were
also evaluated using mass cytometry (CyTOF).
Results: Applying principal component analysis to gene expression profiling, most
of WM pts clustered together, while a small subset of them grouped separately with
MGUS pts, suggesting a biologic dichotomy within WM. The controls formed a group distinct
from most WM and MGUS pts. Fig1A We then applied a non-negative matrix factorization
consensus clustering to the gene expression data and identified three robust clusters.
Cluster 1 (C1) included only pts with WM, cluster 2 (C2) included pts with both WM
and MGUS, and cluster 3 (C3) included all normal controls as well as a small number
of WM and MGUS pts. Fig1B-C When mutations commonly identified in WM were analyzed,
there was no difference among the three groups (excluding controls) in mutation burden
of MYD88 L265P and CXCR4. Interestingly, aberrant expression of TNFAIP3 was a distinct
feature of C1 as deletion of 6q (which encodes for TNFAIP3) and TNFAIP3 mutations
were each significantly enriched in C1 (47%) compared to C2 (0%) and C3 (20%; p=0.04).
Individual clusters associated with specific transcriptional signatures and clinical
features. While C1 displayed enrichment of RNA processing, downregulation of inflammatory
pathways and aggressive clinical behavior, C2 showed increased inflammatory signaling
and senescence with indolent clinical behavior. C3 had intermediate features with
combined proliferative and antigen response signatures. Fig1D In accordance with transcriptomics,
the proteomic hallmark of C1 was upregulation of proteins involved in proliferation
(eg AKT, MAPK) and downregulation of inflammatory proteins (eg IL4, IL10) while the
opposite was observed in C2. Once more, C3 confirmed intermediate features with combined
upregulation of proliferation and inflammatory proteins. The metabolism was rewired
towards mitochondrial anabolism in C1 and C3, while towards glycolysis in C2. Accordingly,
C1 and C3 showed undetectable concentrations of 3-hydroxybutyric acid as opposed to
C2 which had increased levels of lactic acid, as end products of fatty acid oxidation
and glycolysis respectively. Next, we explored whether C1 and C2 displayed a distinct
immune profile. tSNE analysis showed that CD4+ T cells were more abundant in C2 compared
to C1 while the opposite was observed for CD8+ T cells. Among CD4+ T cells, activated
follicular helper (TFH; p=0.02) and regulatory (Treg; p=0.008) cells were predominantly
expressed in C2. In contrast, C1 showed a higher expression of senescent T effector
memory (TEM) cells. Fig1E In support of this, SPADE clustering analysis identified
three clusters including TFH, Treg and TEM cells.
Image:
Summary/Conclusion: We have identified three molecular clusters in IgM monoclonal
gammopathies with distinct clinical, proteomic, metabolomic and immune features, suggesting
a potential biologic classification that may have therapeutic implications.
S172: FUNCTIONAL SUBSETS OF PLASMA CELLS ASSOCIATED WITH AMYLOID PRODUCTION AND
X. Wang1 2 3,*, H. Han1 3, J. Sun3 4, Q. Wang2, X.-M. Gao1 3, K.-N. Shen1 3, L. Zhang1
3, Y. Zhao2, X.-X. Cao1 3, M. Qian5, Y. Chen2, J. Li1 3
1Department of Hematology, Peking Union Medical College Hospital, Chinese Academy
of Medical Sciences and Peking Union Medical College; 2The State Key Laboratory of
Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute
of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences
and Peking Union Medical College; 3State Key Laboratory of Complex Severe and Rare
Diseases; 4Department of Pathology, Peking Union Medical College Hospital, Chinese
Academy of Medical Sciences and Peking Union Medical College; 5School of Mathematical
Sciences, Peking University, Beijing, China
Background: Light chain amyloidosis (AL amyloidosis, AL) is a life-threatening plasma
cell dyscrasia characterized by misfolded monoclonal immunoglobulin light chain production
by pathogenic bone marrow plasma cells (BMPCs), leading to irreversible damage in
multiple organs. Until now, the pathogenic BMPCs in AL haven’t been elucidated, let
alone the functional roles of BMPC subsets.
Aims: We aimed to identify intra- and inter-individual heterogeneities in BMPCs, especially
those related to AL development, light chain production, organ tropism, and chemotherapy
response.
Methods: Here, we conducted single-cell RNA sequencing and image flow cytometry analysis
of BMPCs from patients with AL (n=3), compared with that from monoclonal gammopathy
of undetermined significance (MGUS) (n=2) and healthy controls (n=21). All the subjects
provided informed consent.
Results: We identified 7 functional subsets of BMPCs in AL, MGUS or healthy controls
(Fig. 1a). These subsets had over-lapping but distinct functions, including DNA repair,
cell proliferation, drug response, osteoclast differentiation, and immunoglobulin
production (Fig. 1b). Subsets enriched in AL showed up-regulation of amyloidosis-associated
genes (Fig. 1c), such as the amyloid-beta binding protein-encoding Apolipoprotein
E (APOE), and showed plasmablastic morphology (Fig. 1d). Subsets defined by aberrant
light chain production up-regulated the pathways related to neutrophil degranulation,
transportation to and modifications in the Golgi apparatus, and asparagine N-linked
protein glycosylation. High expression of Cyclin D1 (CCND1), CD79A, and V-Set Pre-B
Cell Surrogate Light Chain 3 (VPREB3) were observed in the predominant subset of AL
predicted sensitive to venetoclax, while Cyclin D2 (CCND2), S100 Calcium Binding Protein
A6 (S100A6), Cystatin C (CST3) were up-regulated in that of AL predicted resistant
(Fig. 1e). In an independent cohort of bulk RNA sequencing (n=29), clinical subgroups
of patients with AL were defined by the proportion of functional subsets, including
2 major subgroups that were consistent with the aforementioned AL with differential
sensitivity to venetoclax (Fig. 1f). Co-expression of CCND1, CD79A, and VPREB3, and
a larger predominant subset, were in the venetoclax-sensitive subgroup. The up-regulation
of CCND2 and amyloidosis-associated genes, including S100A6 and CST3, were in the
venetoclax-resistant subgroup (Fig. 1g).
Image:
Summary/Conclusion: These functional subsets of BMPCs provide mechanistic insights
into AL pathogenesis, light chain production, and chemotherapy response. These results
suggest potential avenues for the exploration of clinical subgroups among AL patients.
S173: T-CELL ACTIVATION AND MYELOMA CELL KILLING CONFIRM THE MODE OF ACTION OF RG6234,
A NOVEL GPRC5D T-CELL ENGAGING BISPECIFIC ANTIBODY, IN PATIENTS WITH RELAPSED/REFRACTORY
MULTIPLE MYELOMA
I. Dekhtiarenko1,*, I. Lelios2, J. Attig2, N. Sleiman2, D. Lazzaro2, I. Clausen2,
N. Gräfe3, H.-J. Helms2, E. Schindler2, S. Belli2, T. Fauti1, J. Eckmann3, P. Umana1,
W. Jacob3, M. Schneider2, C. Hasselbalch Riley4, M. Hutchings4, S.-S. Yoon5, Y Koh5,
S. Manier6, T. Facon6, S. J. Harrison7, J. Er7, F. Volzone8, A. Pinto8, C. Montes9,
E. M. Ocio9, A. Alfonso-Pierola10, P. Rodríguez Otero10, F. Offner11, A. Guidetti12,
P. Corradini12, C. Titouan13, C. Hulin13, C. Touzeau14, P. Moreau14, R. Popat15, S.
Leong15, R. Mazza16, C. Carlo-Stella16, A.-M. E. Bröske3
1Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Zurich;
2Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel,
Switzerland; 3Roche Pharma Research and Early Development, Roche Innovation Center
Munich, Penzberg, Germany; 4Rigshospitalet, Copenhagen, Denmark; 5Seoul National University
College of Medicine, Seoul, South Korea; 6Lille University Hospital, Lille, France;
7Peter MacCallum Cancer Center and Royal Melbourne Hospital, and Sir Peter MacCallum
Department of Oncology, University of Melbourne, Melbourne, Australia; 8Istituto Nazionale
dei Tumori, Fondazione Pascale, IRCCS, Napoli, Italy; 9Hospital Universitario Marques
de Valdecilla (IDIVAL), Universidad de Cantabria, Santander; 10Clinica Universidad
de Navarra, Navarra, Spain; 11Universitair Ziekenhuis Gent, Gent, Belgium; 12Istituto
Nazionale dei Tumori, Milano, Italy; 13CHU de Bordeaux, Bordeaux; 14CHU de Nantes,
Nantes, France; 15University College London Hospitals NHS Foundation Trust, London,
United Kingdom; 16Department of Biomedical Sciences, Humanitas University and Department
of Oncology and Hematology, IRCCS Humanitas Research Hospital, Milano, Italy
Background: RG6234 is a novel T-cell engaging bispecific antibody targeting G protein-coupled
receptor 5D (GPRC5D) with a unique 2:1 format. GPRC5D is highly expressed on multiple
myeloma (MM) cells and concurrent binding of RG6234 to GPRC5D and CD3 on T cells results
in immunological synapse formation and potent T-cell directed tumor cell killing.
An ongoing Phase 1 dose-escalation study (NCT04557150) is investigating the safety,
clinical activity, pharmacodynamics (PD), and pharmacokinetics of RG6234 monotherapy
in patients (pts) with relapsed/refractory MM (RRMM). Clinical activity was observed
during dose escalation, and the safety profile was manageable (Riley et al. EHA 2022).
Aims: Here, we present preliminary clinical biomarker data highlighting PD effects
after intravenous (IV) administration that confirm the mechanism of action and high
potency of RG6234.
Methods: Exploratory biomarker analyses included data from pts treated with RG6234
doses ranging from 0.006mg to 4.8mg in dose escalation. RG6234 was administered as
an IV infusion under a step-up dosing regimen, reaching the target dose not later
than 2 weeks after the priming dose. Peripheral biomarkers were evaluated using whole
blood flow cytometry (n=28), plasma cytokine Protein Simple ELLA (n=33), and plasma
sBCMA Protein Simple ELLA (n=26). MM cells were assessed at baseline and on-treatment
by bone marrow (BM) aspirate flow cytometry and by BM biopsy CD138/CD8 immunohistochemistry.
Informed consent was obtained from participating pts. The clinical cut-off date for
the current analysis was January 31, 2022.
Results: PD changes were observed in peripheral blood at all tested doses. Cytokines
(IFNg, TNFa, CXCL10, IL6, IL10, IL2, IL8) and sCD25 peaked at 4 to 24 hours (h) following
the first administration, while cytokine peak magnitudes decreased at subsequent administrations.
Cytokine release was followed by transient reduction of circulating T cells at 4 h
after infusion, with partial recovery of peripheral T-cell counts by Day 8 after first
administration. Elevation of sCD25 and IFNg in plasma (~3.2 and 33 median fold change
from baseline, respectively) together with increase in T-cell proliferation (~4 median
fold increase of Ki67+CD8+ T-cells) within 72 h post first infusion indicated T-cell
activation. Analysis of a limited number of paired baseline and on-treatment BM biopsies
(n=13) revealed that the density of CD8+ tumor-infiltrating T cells increased upon
treatment in responders, indicating T-cell recruitment towards the tumor. RG6234 induced
rapid depletion of MM cells, as demonstrated by a decrease of sBCMA in the plasma
of responding pts already 8 days after the first administration (median 33.5% reduction
from baseline in responding pts; n=15). Moreover, at the end of Cycle 1, the majority
of pts (14/15) had <1% of MM cells in BM based on flow cytometry readout. GPRC5D expression
was detected at baseline in all pts with evaluable bone marrow aspirate and >20 detectable
MM cells (n=16). Updated exploratory biomarker data will be presented.
Summary/Conclusion: Cytokine release, T-cell activation, BM infiltration, and MM cell
depletion are early PD changes seen after treatment with RG6234 and precede clinical
responses. These PD changes indicate that RG6234 leads to T-cell engagement in the
BM of pts with RRMM and clearly demonstrate rapid and effective T-cell mediated anti-MM
activity.
S174: HIGH LEVEL OF CIRCULATING TUMOUR DNA AT DIAGNOSIS CORRELATES WITH DISEASE SPREADING
AND DEFINES MULTIPLE MYELOMA PATIENTS WITH POOR PROGNOSIS
M. Martello1,*, A. Poletti1, D. Bezzi2, E. Borsi1, B. Taurisano1, V. Solli1, S. Armuzzi1,
I. Vigliotta1, G. Mazzocchetti1, I. Pistis3, L. Pantani3, S. Rocchi1, K. Mancuso1,
P. Tacchetti3, I. Rizzello1, M. Cavo1, E. Zamagni1, C. Nanni2, C. Terragna3
1IRCCS - Azienda Ospedaliero Universitaria di Bologna - Department of Experimental,
Diagnostic and Specialty Medicine - University of Bologna; 2IRCCS - Azienda Ospedaliero
Universitaria di Bologna - Department of Nuclear Medicine; 3IRCCS - Azienda Ospedaliero
Universitaria di Bologna, Bologna, Italy
Background: Multiple Myeloma (MM) is a plasma cell (PC) disorder characterized by
the presence of skeletal involvement at the time of diagnosis, as detected by MRI
and/or FDG PET/CT, in most of the patients. The patchy nature of the disease is probably
related to the ability of fitter clones to spread into peripheral blood reaching distant
sites, where favourable microenvironment conditions might promote clones’ seeding.
Recently, cell-free DNA (cfDNA) has been proven to resume the heterogeneity of spatially
distributed clones. However, it has to be determined to which extent cfDNA correlates
with disease distribution and its possible implications with patients’ outcome. Moreover,
the potential of cfDNA to track the evolutionary dynamics and the heterogeneity of
MM, possibly anticipating the emergence of therapy resistant residual cells, remains
to be confirmed.
Aims: Aim of this study is to quantitatively and qualitatively evaluate cfDNA at diagnosis
and during follow-up in correlation with imaging data to possibly define the integration
of this approach with molecular bone marrow (BM) and whole-body residual disease assessment.
Methods: A total of 88 newly diagnosed MM patients were screened at baseline with
18F-FDG PET/CT, and molecularly assessed by Ultra Low Pass-Whole Genome Sequencing
(ULP-WGS). In a subgroup of 22 patients, cfDNA was monitored monthly, whereas PET/CT
was reassessed after induction therapy to evaluate metabolic tumor response. For each
pts, ULP-WGS was used to characterize both the neoplastic PC clone(s) in the BM (gDNA)
and the cfDNA from peripheral blood. Data were analysed by ichorCNA and Clonality
R packages.
Results: At diagnosis, the cfDNA tumor fraction (TF) was significantly lower as compared
to gDNA TF [median (M) TF: 4.4 vs. 59.7%, respectively]. Nevertheless, high cfDNA
TF levels (> 4.4% cfDNA TF values; range: 4.4-84.3%) correlated with high gDNA TF
levels (>65.7% gDNA TF values; range: 65.7-96.7%). This observation was further confirmed
by a significant correlation between cfDNA TF and the percentage of BM CD138/CD38
positive plasma cells (r=0.47; p<.0001).
Interestingly, patients with high cfDNA TF at baseline were more likely to present
with extramedullary disease (EMD) and a higher number of focal lesions, and also featured
a more active tumour metabolism, as compared to pts with low TF (EMD 4/44=9% vs. 1/44=2.3%,
p=ns; M n. PET lesions: 1.7 vs. 2.5, p= 0.003; SUVmax: 5.2 vs. 9.6, p = 0.01).
Despite an overall concordance between cfDNA and BM genomic profiles (80/88=90.9%),
those patients with high cfDNA TF showed more frequently evidence of spatial heterogeneity,
as highlighted by a substantial divergence of copy number alterations profiles between
cfDNA and gDNA (7/8=87.5% of pts with divergent profiles). Finally, high cfDNA TF
at diagnosis predicted for poorer prognosis as compared with low cfDNA TF (PFS at
20 months: 67% vs. 86%, p=0.05; OS at 20 months: 90% vs. 100%, p=0.04).
Interestingly, after bortezomib-based induction therapy, imaging data and cfDNA TF
levels were concordant; notably, in 4/10 (40%) patients with median SUVmax 5.8 and
>2 PET lesions, cfDNA TF was still detectable (>1% TF), thus corroborating a possible
role for cfDNA in monitoring response to therapy.
Summary/Conclusion: In conclusion, patients with high cfDNA TF displayed imaging data
that overall suggested a higher propensity to a metastatic spread of the disease,
which finally correlated with poorer prognosis. Future studies will be addressed to
exploit the use of cfDNA in disease monitoring.
Acknowledgements: AIRC IG2019, AIL BOLOGNA ODV
S175: ATLAS: A PHASE 3 RANDOMIZED TRIAL OF CARFILZOMIB, LENALIDOMIDE, AND DEXAMETHASONE
VERSUS LENALIDOMIDE ALONE AFTER STEM-CELL TRANSPLANT FOR MULTIPLE MYELOMA
D. Dytfeld1,*, T. Wróbel2, K. Jamroziak3, T. Kubicki1, P. Robak4, A. Walter-Croneck5,
J. Czyż6, A. Tyczyńska7, A. Druzd-Sitek8, K. Giannopoulos9, A. Nowicki1, T. Szczepaniak1,
A. Łojko-Dankowska1, M. Matuszak1, L. Gil1, B. Puła10, J. Rybka2, M. Majcherek2, L.
Usnarska-Zubkiewicz10, L. Szukalski11, A. Końska10, J. M. Zaucha7, J. Walewski8, D.
Mikulski4, O. Czabak5, T. Robak4, U. Grzybowska1, O. B. Lahoud12, J. A. Zonder13,
K. Griffith14, A. Stefka15, A. Major15, B. A. Derman15, A. J. Jakubowiak15
1Poznan University of Medical Sciences, Poznan; 2Wroclaw Medical University, Wroclaw;
3Medical University of Warsaw, Warsaw; 4Medical University of Lodz, Lodz; 5University
of Medical Sciences in Lublin, Lublin; 6Collegium Medicum, Bydgoszcz; 7Medical University
of Gdansk, Gdansk; 8Maria Sklodowska-Curie National Research Institute of Oncology,
Warsaw; 9Medical University of Lublin, Lublin; 10Institute of Hematology and Blood
Transfusion, Warsaw; 11Medical University of Bydgoszcz, Bydgoszcz, Poland; 12Memorial
Sloan Kettering Cancer Center, New York; 13Karmanos Cancer Institute, Detroit; 14University
of Michigan, Ann Arbor; 15University of Chicago, Chicago, United States of America
Background: Treatment following autologous stem cell transplantation (ASCT) for multiple
myeloma (MM) remains an area of active investigation. We have shown that extended
post-ASCT treatment with carfilzomib, lenalidomide, and dexamethasone (KRd) after
KRd induction improved the depth and duration of response (Jasielec et al, Blood 2020),
suggesting a benefit of post-ASCT KRd therapy.
Aims: We report the results of ATLAS, a multicenter, international, open-label randomized
phase 3 study to determine the efficacy and safety of post-ASCT KRD vs standard lenalidomide
(R) maintenance (NCT02659293).
Methods: Newly-diagnosed MM patients (pts) who received any induction therapy for
up to 12 months (mo) followed by single ASCT and achieved at least stable disease
within 100 days afterward were randomized to receive either KRd or R, stratified by
post-transplant response (≥VGPR vs <VGPR) and cytogenetic risk [standard risk (SR)
vs high [HR: presence of t(4;14), t(14;16), or del(17p)]. Pts randomized to KRd received
carfilzomib 36 mg/m2 on days (D) 1,2,8,9,15,16 for 4 cycles (C) then D1,2,15,16 starting
C5; R 25 mg D1-21, and dexamethasone 20 mg D1,8,15,22 in 28-day cycles. KRd pts with
SR who reached IMWG-defined MRD-negativity after C6 de-escalated therapy to R alone
after C8 (KRd->R); the rest continued KRd through C36 followed by R alone until progression.
Pts randomized to R received lenalidomide 10 mg C1-3 and then 15 mg daily. The primary
endpoint was progression free survival (PFS) rate: based on historical PFS rates,
a sample size of 180 Pts was calculated to provide 85% power with 2-sided alpha 0.05.
Secondary endpoints included minimum residual disease (MRD) negativity response rates
and treatment-related adverse events.
Results: 180 pts were enrolled (R n=87; KRd n=93) through 10/21/20; data cutoff was
12/31/21. Pt characteristics in the KRd and R arms were balanced for median age (58
vs 59 yrs), ≥VGPR (88% vs 92%), and HR (23% vs 21%). After 6 cycles, 47% pts in the
KRd arm and 29% in the R arm achieved MRD-negativity (p=0.017). 34 KRd pts eligible
for de-escalation converted to R alone after C8 and were analyzed on the KRd arm per
intention-to-treat. At median follow-up of 33.8 mo, 23 pts (25%) on the KRd arm and
38 pts (44%) on the R arm progressed; estimated median PFS was 59.0 mo for KRd vs
41.4 mo for R (Hazard Ratio 0.56, logrank p=0.026). At cutoff, 90% of KRd and 87%
of R pts were alive; no deaths were treatment-related. All-grade toxicities were generally
comparable between arms. The most common grade 3+ AEs and those of special interest
were neutropenia (KRd 47%; R 59%), thrombocytopenia (KRd 13%; R 7%), infections (KRd
15%; R 6%), cardiovascular toxicities (KRd 4%, R 5%), and secondary malignancies (KRd
2, R 2).
Image:
Summary/Conclusion: This is the first randomized phase 3 trial demonstrating superior
PFS with extended post-transplant KRd therapy compared to R maintenance. Therefore,
MRD/risk-adapted post-ASCT extended KRd treatment may represent a new standard of
care.
S176: DARATUMUMAB CARFILZOMIB LENALIDOMIDE AND DEXAMETHASONE AS INDUCTION THERAPY
IN HIGH-RISK TRANSPLANT ELIGIBLE NEWLY DIAGNOSED MYELOMA PATIENTS: RESULTS OF THE
PHASE 2 STUDY IFM 2018-04
C. Touzeau1,*, A. Perrrot2, C. Hulin3, S. Manier4, M. Macro5, M.-L. Chretien6, L.
Karlin7, O. Decaux8, C. Jacquet9, M. Tiab10, X. Leleu11, L. Planchce12, H. Avet-Loiseau2,
P. Moreau13
1CHU Nantes - Hotel Dieu, CHU Nantes - Hotel Dieu, Nantes; 2Hematology, IUCT Oncopole,
Toulouse; 3Hematology, CHU Bordeaux, Bordeaux; 4Hematology, CHU Lille, Lille; 5Hematology,
CHU Caen, Caen; 6Hematology, CHU Dijon, DIjon; 7Hematology, CHU Lyon, Lyon; 8Hematology,
CHu Rennes, Rennes; 9Hematology, CHU Nancy, Nancy; 10Hematology, CHD, La Roche sur
Yon; 11Hematology, CHU Poitiers, Poitiers; 12Biostatistic; 13Hematology, CHU Nantes,
Nantes, France
Background: High-risk (HR) cytogenetic is associated with poor outcome in transplant
eligible (TE) newly diagnosed myeloma multiple myeloma (NDMM). The triplet combination
carfilzomib lenalidomide and dexamethasone (KRD) plus transplantation demonstrated
high efficacy with favorable safety profile in TE-NDMM patients (FORTE). The addition
of daratumumab (Dara) to frontline therapy also improved response rate and progression
free-survival in TE-NDMM patients (CASSIOPEIA, GRIFFIN). Double transplant also improved
outcome of HR TE NDMM patients (EMN02, STAMINA).
Aims: The phase 2 trial 2018-04 from the Intergroupe Francophone du Myelome (IFM)
is evaluating an intensive strategy with Dara-KRD induction and consolidation plus
double transplant in HR TE NDMM (NCT03606577).
Methods: HR MM was defined by the presence of del17p, t(4;14) and/or t(14;16). Stategy
includes Dara-KRD induction (6 cycles), autologous stem cell transplantation (ASCT),
Dara-KRD consolidation (4 cycles), second ASCT, Dara-lenalidomide maintenance. The
primary endpoint was the feasibility of this intensive strategy. Here, we report efficacy
and safety analysis of Dara-KRD induction.
Results: Fifty patients with previously untreated NDMM were included from july 2019
to march 2021 in 11 IFM centers Median age was 57 (range 38 -65). ISS stage 3 was
present in 12 (24%) patients. Based on inclusion criteria, all patients had HR cytogenetic,
including 17p deletion (n=20, 40%), t(4;14) (n=26, 52%) or t(14;16) (n=10,20%). Forty-six
patients completed Dara-KRD induction. Two patients discontinued treatment due to
severe adverse event (COVID-19 infection, n=1; drug-induced hepatitis, n=1) and 2
patients discontinued treatment due to disease progression. Grade 3-4 treatment related
adverse event (>5% of patients) were neutropenia (38%), anemia (14%), thrombocytopenia
(8%), infection (6%), renal insufficiency (6%) and deep-vein thrombosis (6%). Two
patients (6%) experienced stem-cell collection failure. Overall response rate was
96%, including 92 % ≥ very good partial response. Among 37/46 evaluable patients post
induction, Minimal Residual Disease negativity rate (NGS, 10-5) was 62%.
Summary/Conclusion: Dara-KRD as induction prior ASCT is safe and allows deep responses
in TE NDMM patients with high-risk cytogenetic profile. IFM 2018-04 study is ongoing
and longer follow-up is needed to evaluate safety and efficacy of the overall strategy
with Dara-KRD induction and consolidation plus double transplant in this subset of
HR patients.
S177: EVALUATING SERUM FREE LIGHT CHAIN RATIO AS A BIOMARKER FOR MULTIPLE MYELOMA
T. Akhlaghi1,*, K. Maclachlan2, N. Korde2, S. Mailankody2, A. Lesokhin2, H. Hassoun2,
S. X. Lu2, D. Patel2, U. Shah2, C. Tan2, A. Derkach3, O. Lahoud4, H. J. Landau4, G.
L. Shah4, M. Scordo4, D. J. Chung4, S. A. Giralt4, S. Z. Usmani2, O. Landgren5, M.
Hultcrantz2
1Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside
and West; 2Myeloma Service, Department of Medicine; 3Department of Epidemiology and
Biostatistics; 4Adult Bone Marrow Transplant Service, Department of Medicine, Memorial
Sloan Kettering Cancer Center, New York; 5Myeloma Service, Sylvester Comprehensive
Cancer Center, University of Miami, Miami, United States of America
Background: In 2014, the definition of multiple myeloma (MM) was updated to include
serum free light chain (FLC) ratio ≥100 as a myeloma defining biomarker, based on
retrospective data indicating a 2-year progression rate of 80% and a median time to
progression (TTP) of 12 months associated with this marker. As a result, patients
previously diagnosed as smoldering MM (SMM), were considered to have MM requiring
treatment if they had a FLC ratio ≥100. However, two recent studies have reported
lower 2-year progression rates, 30-44%, and a longer median TTP of 40 months in patients
with FLC ratio ≥100, raising concern about the FLC criteria.
Aims: To assess the risk of progression in patients with SMM and a FLC ratio ≥100.
Methods: We performed a retrospective analysis of patients diagnosed with SMM at Memorial
Sloan Kettering Cancer Center between January 2000 and December 2017. Diagnosis of
SMM and progression to active MM requiring therapy was defined according to the International
Myeloma Working Group (IMWG) criteria at the time of diagnosis. Kaplan-Meier method
was used to assess TTP and generate survival curves, with log-rank test for comparison
between groups.
Results: A total of 438 patients were included in the study, with a median follow-up
time of 52 months. While all patients with FLC ≥100 (n=66) had elevated involved FLC
levels, 35 (53%) had an involved FLC concentration >100 mg/L. Per current diagnostic
criteria, we only included patients with an involved FLC concentration >100 mg/L in
the FLC ratio ≥100 group and found a median TTP of 31 months (95% confidence interval
[CI] 16-59 months) and a 2-year progression rate of 49% (CI 28-63%). In a sensitivity
analysis including all 66 cases with FLC ratio ≥100 (independent of involved FLC concentration),
we found the median TTP to be 41 months (CI 30-72 months), compared to 101 months
for those with a FLC ratio <100 (CI 78-127 months; p<0.0001). Furthermore, the risk
of progression within 2 years was 35% (CI 22-46%) compared to 18% (CI 14-23%; p<0.0001)
for those with a FLC ratio <100.
Of note, 22 patients with a FLC ratio ≥100 were monitored expectantly for >4 years,
among whom 12 patients had involved FLC levels >100 mg/L. Ten patients (7 with involved
FLC level >100 mg/L) were followed over a period ranging from 4 to 8.5 years before
eventually progressing, and 12 patients (5 with an involved FLC level >100 mg/L) were
followed between 4 and 8 years and did not progress during the study period.
Image:
Summary/Conclusion: While FLC ratio ≥100 is associated with a high risk of progression
in patients with SMM, it does not infer an imminent risk of progression, defined by
the IMWG as median TTP of 12 months and 2-year progression rate of at least 80%. On
the contrary, select patients with FLC ratio ≥100 can be followed for many years without
progressing and some may never progress despite long-term follow-up. These findings
suggest that in patients where FLC ratio ≥100 is the only myeloma-defining event,
other high-risk features as well as the evolution of FLCs over time should be considered
in the decision to start a patient on treatment.
S178: SAFETY AND EFFICACY OF BELANTAMAB MAFODOTIN IN COMBINATION WITH RD IN NEWLY
DIAGNOSED, TRANSPLANT INELIGIBLE MULTIPLE MYELOMA PATIENTS: A PHASE 1/2 STUDY BY THE
HELLENIC SOCIETY OF HEMATOLOGY
E. Terpos1,*, M. Gavriatopoulou1, I. Ntanasis-Stathopoulos1, P. Malandrakis1, D. Fotiou1,
N. Kanellias1, S. Gkolfinopoulos2, K. Manousou2, E. Kastritis1, M.-A. Dimopoulos1
1Department of Clinical Therapeutics, National and Kapodistrian University of Athens,
School of Medicine, Athens, Greece; 2Health Data Specialists, Dublin, Ireland
Background: Belantamab mafodotin (belamaf), a multi-modal antibody-drug conjugate
targeting BCMA, has shown efficacy and tolerability in pretreated patients (pts) with
relapsed/refractory multiple myeloma. In pts refractory to immunomodulatory drugs,
proteasome inhibitors, and antiCD38, belamaf plus pomalidomide and dexamethasone induced
a very good partial response (VGPR) or better of 69.2% and a median progression-free
survival of 16.2 months (Trudel S, ASH 2021). Preclinical evidence suggests that belamaf
plus lenalidomide enhances antimyeloma activity, with no overlapping toxicities.
Aims: To evaluate the safety and efficacy of belamaf plus lenalidomide and dexamethasone
(Rd) in transplant-ineligible (TI) pts with newly diagnosed multiple myeloma (NDMM).
Methods: The ongoing, prospective, open-label, 2-part, phase 1/2 BelaRd study (NCT04808037)
aims to enroll 66 pts with TI NDMM from a Greek center. Eligible are adult pts with
Eastern Cooperative Oncology Group status 0–2 and adequate organ function. Part 1
(dose selection) evaluates the safety/tolerability of 3 belamaf doses (2.5, 1.9, and
1.4 mg/kg on Day 1 of every other 28-day cycle) plus Rd (each dose regimen administered
to a cohort of 6 pts) over ≥4 weeks of follow up; subsequently, an additional 6 pts
are enrolled in each dose cohort to establish the recommended phase 2 dose (RP2D).
Part 2 (dose expansion) evaluates the safety and clinical activity of belamaf RP2D
plus Rd in 30 additional pts. This descriptive analysis presents the safety data for
all Part 1 pts and the efficacy data for all Part 1 pts with ≥2 post-baseline efficacy
assessments by the cut-off date (14/01/2022).
Results: Of 36 pts included, 35 (97.2%) continued study treatment by the cut-off date,
and 1 (2.8%) had died due to a belamaf unrelated adverse event (AE). The pt median
age was 72.5 years (range 64.0–86.0). Of pts with available data (30 [83.3%]), pts
at revised International Staging System stages I, II, and III were 6 [20.0%], 21 [70.0%],
and 3 [10.0%], respectively, and 3 (10.0%) had high-risk cytogenetics (i.e., del17p13,
t(4;14), t[14;16]). Median duration of therapy was 4.2 months (range 0.5–11.9) and
median number of cycles reached was 5.0 (range 1.0–11.0). Twenty-two (61.1%) pts experienced
at least one grade (Gr) 3–4 AE. One (2.8%) pt experienced a Gr 5 AE (pneumonia), unrelated
to belamaf. Most common (≥ 10.0% of pts) Gr 3–4 AE were fatigue (13 [36.1%] pts),
visual acuity reduced (6 [16.7%] pts), and rash (5 [13.9%] pts). Gr 3–4 thrombocytopenias
and infections were not reported, as were any Gr infusion-related reactions (Table).
Pts with Gr 1–2 ocular symptoms, visual acuity reduced, and keratopathy, were 27 (75.0%),
21 (58.3%), and 18 (50.0%), respectively. Pts with Gr 3–4 ocular symptoms, visual
acuity reduced, and keratopathy were 0 (0.0%), 5 (13.9%) and 0 (0.0%), respectively.
Of all pts, 28 (77.8%) were evaluable for efficacy (Table). At a median follow up
of 4.2 months (range 0.5–11.9), the overall response rate was 96.4% (27/28 pts; complete
response [CR]: 14.3% [4/28 pts]; VGPR: 35.7% [10/28 pts]; partial response [PR]: 46.4%
[13/28 pts]); no disease progression was reported. Of pts achieving VGPR, 6/10 (60.0%)
had negative status serum/urine immunofixation electrophoresis. Median time to PR
or better was 1.0 months (range 0.9–2.0).
Image:
Summary/Conclusion: In pts with TI NDMM, belamaf every 2 months plus Rd showed an
improved safety profile, especially at lower doses. Rapid and deep hematological responses
were recorded.
S179: RANDOMIZED COMPARISON BETWEEN KRD AND KTD INDUCTION, FOLLOWED BY K MAINTENANCE
OR OBSERVATION IN TRANSPLANT NON-ELIGIBLE PATIENTS WITH NDMM (AGMT-MM02 TRIAL)
H. Ludwig1,*, T. Melchardt2, S. Sormann3, N. Zojer4, J. Andel5, B. Hartmann6, C. Tinchon7,
E. Gunsilius8, K. Podar9, A. Egle2, W. Willenbacher8 10, E. Wöll11, M. Schreder4,
R. Ruckser12, B Bozic12, M.-T. Krauth13, A. Petzer14, C Schmitt15, S. Machherndl-Spandl16,
H. Agis17, M. Fillitz18, W. Pönisch19, S. Knop20, B. Paiva21, R. Greil2
1Wilhelminen Cancer Research Institute, Vienna; 2Department of Internal Medicine III
with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology,
Oncologic Center, Salzburg Cancer Research Institute – Laboratory for Immunological
and Molecular Cancer Research (SCRI-LIMCR), Paracelsius Medical University, Cancer
Cluster Salzburg, Salzburg; 3Department of Internal Medicine, University Clinic Graz,
Graz; 4Department of Medicine I, Clinic Ottakring, Vienna; 5Department of Internal
Medicine IIr, Pyhrn-Eisenwurzen Klinikum Steyr, Steyr; 6Department of Internal Medicine
II, LKH Rankweil, Rankweil; 7Department of Internal Medicine, LKH Hochsteiermark,
Leoben; 8Department of Internal Medicine V, Medical University Innsbruck, Innsbruck;
9Department of Internal Medicine II, University Hospital Krems, Karl Landsteiner University
of Health Sciences, Krems; 10Oncotyrol, Center for personalized Cancer Medicine, Innsbruck;
11Department of Internal Medicine, KH Zams, Zams; 12Department of Medicine II, Clinic
Donaustadt; 13Department of Internal Medicine I, Division Hematology & Hemostaseology,
Medical University Vienna, Vienna; 14Department of Internal Medicine I, Ordensklinikum
Linz Barmherzige Schwestern; 15Clinic for Internal Medicine; 3, Kepler University
Clinic Linz; 16Ordensklinikum Linz Elisabethinen, Linz; 17Department of Internal Medicine
I, Division of Oncology, Medical University Vienna; 18Department of Internal Medicine,
Hanusch Krankenhaus, Vienna, Austria; 19Medical Clinic and Policlinic I, University
Clinic Leipzig, Leipzig; 20Medical Clinic and Policlinic II, University Clinic Würzburg,
Würzburg, Germany; 21Clinica Universidad de Navarra, Pamplona, Spain
Background: Carfilzomib (K)-based combinations have been established as effective
frontline and relapse regimens in pts with multiple myeloma (MM).
Aims: In this randomized phase II trial we evaluated the impact of either Revlimid
(R) or Thalidomide (T) as combination partner for K and dexamethasone (KRd or KTd)
on outcome in pts with newly diagnosed MM (NDMM) not eligible for autologous transplantation
(TNE). Further, we evaluated the role of one year K maintenance therapy compared to
observation.
Methods: One hundred twenty two pts have been enrolled (ITT population). Median age
was 75 yrs, ISS stage I/II/III: 29 (23.8%)/48 (39.3%)/45 (36.9%), ECOG stage 0/1:
64 (52.5%) / 58 (47.5%). t(4;14) ± del17p was noted in 15 (16.3%) of 92 pts with results
available. Pts were randomized to 9 cycles of KRd or KTd, and 107 pts received at
least one full cycle. Carfilzomib (K) was started with 20mg/m2 at d 1 of cycle 1,
and was continued with 27mg/m2 for the first 2 cycles (d 1 + 2, 8 + 9, 15 + 16 schedule);
followed by K administration at 57mg/m2 once weekly for a 28 d cycle. Thalidomide
100mg/d (50mg in pts >75 yrs of age), d 1-28, or Revlimid 25mg/d (15mg in pts 375
yrs of age) d 1-21. Dexamethasone 40mg (20mg in pts 375 yrs of age) once/week. After
induction, pts with 3SD were randomized to K maintenance (d 1 and 15) for 12 cycles
or observation. MRD was assessed by NGF with a sensitivity of 10-6 in pts with ≥VGPR.
Survival estimates were calculated according to Kaplan-Meier and survival curves were
compared with the log-rank test. PFS and OS results presented are given for the ITT
population. This trial is registered on clinicaltrials.gov (NCT02891811).
Results: Median follow-up was 25.3 mos, 15 pts discontinued therapy within the first
cycle due to patient (3) or investigator (1) decision, AE/toxicity (8), death or progressive
disease (2) or other reason (1). Overall response rate was 91.3% in the entire group
with available data (n=115). Results for sCR, CR, VGPR, PR, and ORR for KRd and KTd
were similar between both groups (7.3%/10.0%, 27.3%/33.3%, 38.2%/35.0%, 14.5/16.7%,
87.3%/95.0%, respectively). Minor response was noted in 4 (3.5%), stable disease in
5 (4.3%) and progressive disease in 1 (0.8%) pts.
PFS (median 26.9 and 23.5 mos, p=0.832) and OS (not reached vs 52.2 mos, p=0.398)
were similar between the KRd and KTd group, respectively. The OS rate at 36 mos was
82% in both groups. MRD testing was performed in 57 pts at time of CR/VGPR. Of those,
43.9%, (20.5% of the ITT group) pts were found to be MRDneg. PFS was significantly
longer in MRDneg vs. MRDpos pts (p=0.003). Seventy six pts were randomized to K maintenance
therapy or observation. Median PFS was numerically higher in the pts with K maintenance
treatment (median 33.0 vs 24.0, p=0.714), but the difference was not statistically
significant. Data on OS are not mature yet (only 9 events).
Grade 3/4 hematologic AEs were anemia (4.1%), leukopenia (0.8%), thrombocytopenia
(7.4%), while non-hematologic grade 3/4 AEs were infection (20.5%), GI-disorders (7.4%),
hypertension (7.4%), renal and cardiac impairment/failure (6.6% and 8.2% respectively).
Image:
Summary/Conclusion: Our data show similar high efficacy of KRd and KTd in elderly
NTE NDMM pts, including no difference in ORR (KRd and KTd, 87.3% and 95%, respectively),
PFS and OS. Overall survival rate at 3 yrs was 82%. Median PFS was significantly longer
in MRDneg pts. PFS was numerically, but not statistically longer in pts on K maintenance
vs observation. Treatment was associated with an acceptable tolerance profile.
S180: RG6234, A NOVEL GPRC5D T-CELL ENGAGING BISPECIFIC ANTIBODY, INDUCES RAPID RESPONSES
IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA: PRELIMINARY RESULTS FROM A
FIRST-IN-HUMAN TRIAL
C. Hasselbalch Riley1,*, M. Hutchings1, S.-S. Yoon2, Y. Koh2, S. Manier3, T. Facon3,
S. J. Harrison4, J. Er4, F. Volzone5, A. Pinto5, C. Montes6, E. M. Ocio6, A. Alfonso-Pierola7,
P. Rodriguez Otero7, F. Offner8, A. Guidetti9, P. Corradini9, C. Titouan10, C. Hulin10,
C. Touzeau11, P. Moreau11, R. Popat12, S. Leong12, R. Mazza13, A.-M. E. Bröske14,
I. Dekhtiarenko15, H.-J. Helms16, S. Belli16, T. Vardar16, T. Fauti15, J. Eckmann14,
T. Moore14, M. Schneider16, W. Jacob14, M. Weisser14, C. Carlo-Stella13
1Rigshospitalet, Copenhagen, Denmark; 2Seoul National University College of Medicine,
Seoul, South Korea; 3Lille University Hospital, Lille, France; 4Peter MacCallum Cancer
Center and Royal Melbourne Hospital, and Sir Peter MacCallum Department of Oncology,
University of Melbourne, Melbourne, Australia; 5Istituto Nazionale dei Tumori, Fondazione
Pascale, IRCCS, Napoli, Italy; 6Hospital Universitario Marques de Valdecilla (IDIVAL),
Universidad de Cantabria, Santander; 7Clinica Universidad de Navarra, Navarra, Spain;
8Universitair Ziekenhuis Gent, Gent, Belgium; 9Istituto Nazionale dei Tumori, Milano,
Italy; 10CHU de Bordeaux, Bordeaux; 11CHU de Nantes, Nantes, France; 12University
College London Hospitals NHS Foundation Trust, London, United Kingdom; 13Department
of Biomedical Sciences, Humanitas University and Department of Oncology and Hematology,
IRCCS Humanitas Research Hospital, Milano, Italy; 14Roche Pharma Research and Early
Development, Roche Innovation Center Munich, Penzberg, Germany; 15Roche Pharma Research
and Early Development, Roche Innovation Center Zurich, Zurich; 16Roche Pharma Research
and Early Development, Roche Innovation Center Basel, Basel, Switzerland
Background: Patients (pts) with relapsed/refractory multiple myeloma (RRMM) are in
need of effective treatment options. RG6234 is a T-cell engaging bispecific antibody
targeting GPRC5D with a novel 2:1 format. NCT04557150 is an ongoing first-in-human
Phase 1 trial. We present initial results from the dose-escalation part using intravenous
(IV) administration.
Aims: To characterize the safety of RG6234 and to identify a recommended Phase 2 dose
(RP2D), and to characterize the pharmacokinetics, pharmacodynamics, and preliminary
clinical activity.
Methods: Eligible pts with RRMM received RG6234 in escalating weekly step-up doses
followed by a q2w regimen at a peak ‘target dose’ for up to 1 year. A mCRM model was
used for overdose control. Adverse events (AEs) were graded per CTCAE v5.0 and cytokine
release syndrome (CRS) per ASTCT criteria (Lee et al. 2019). Response was assessed
by the investigator according to IMWG criteria. All pts provided informed consent.
Results: As of January 31, 2022, 41 pts had received RG6234 IV (0.006mg to 10mg).
Median age was 63 years (range: 27 to 78) and 61% had R-ISS stage II or III at study
entry. High-risk cytogenetics (t(4;14), t(14;16), del(17p), 1q21 gain) were present
in 17/29 (58%) evaluable cases. Median number of prior therapies was 5 (range: 2 to
15). Pts were triple-class refractory in 67% of cases and penta-class refractory in
36%. Six pts (14.6%) had prior BCMA-directed therapy.
CRS occurred in 85.4% of pts (G1 56.1%, G2 24.4%, G3 2.4%). CRS was generally confined
to the first cycle, with a median time to onset of 4.2 hours (range: 0 to 64.2), and
14 pts (34%) requiring tocilizumab. Three pts (7.3%) experienced CNS toxicity related
to RG6234 (G1 and G3 headache; G1 confusion). G3/4 neutropenia, thrombocytopenia,
and anemia occurred in 9.8%, 19.5%, and 12.2% of pts, respectively.
Infections were reported in 46.3% of pts (≥G3 14.6%). One patient died of an E. coli
sepsis not considered related to RG6234.
AEs probably related to target expression included skin-related AEs in 66% of pts
(G3 7.3%), dysgeusia/ageusia (36.6%, all G1/2), dry mouth (36.6%, all G1/2), dysphagia
(17.1%, all G1/2), and nail changes (12.2%, all G1/2).
No discontinuations due to RG6234-related AEs occurred. The maximum tolerated dose
(MTD) was not reached.
Within the tested target-dose range, RG6234 serum exposure generally showed a dose-proportional
increase, with a preliminary IV terminal T½ of ~5 days (mean value, n=22). At the
predicted active-dose range (>0.162mg), RG6234 induced profound and rapid reductions
in serum free light chains (FLC) and sBCMA. After the first treatment cycle, 93% of
evaluable pts (14/15) had <1% of multiple myeloma cells in bone marrow based on flow
cytometry.
Overall response rate in 34 efficacy evaluable pts across all doses was 68%, and 50%
achieved a VGPR or better response, including 2/6 BCMA pre-treated pts. Responses
occurred rapidly, with a median time to first response of 1.3 months (95% CI: 1.1,
1.6).
Median follow-up was 3.5 months (range: 0.03 to 11.7). Treatment was ongoing for 58.5%
of pts at the time of the data cut. Duration of response was not mature. Response
was ongoing in 18/23 (78.3%) responding pts, with a longest duration of response of
10 months.
Summary/Conclusion: RG6234, a novel T-cell engaging bispecific antibody targeting
GPRC5D, demonstrates compelling clinical activity with manageable safety in heavily
pretreated pts with RRMM. Dose escalation via IV and subcutaneous routes continues,
and data will be updated.
S181: MODAKAFUSP ALFA (TAK-573): UPDATED CLINICAL, PHARMACOKINETIC (PK), AND IMMUNOGENICITY
RESULTS FROM A PHASE 1/2 STUDY IN PATIENTS (PTS) WITH RELAPSED/REFRACTORY MULTIPLE
MYELOMA (RRMM)
J. L. Kaufman1,*, S. Atrash2, S. A. Holstein3, O. Nadeem4, D Benson5, K. Suryanarayan6,
Y. Liu7, C. Li8, L. Yang8, X. Parot8, D. T. Vogl9
1Winship Cancer Institute of Emory University, Atlanta; 2Haematology and blood disorders,
Levine Cancer Institute, Charlotte; 3University of Nebraska Medical Center, Omaha;
4Dana-Farber Cancer Institute Boston, Boston; 5Hematology, Ohio State University Comprehensive
Cancer Center, Columbus; 6Oncology Clinical Research; 7Statistical and Quantitative
Sciences; 8Takeda Development Center Americas, Inc. (TDCA), Lexington; 9Hematologic
Malignancies and Bone Marrow Transplant Program, Abramson Cancer Center, University
of Pennsylvania, Philadelphia, United States of America
Background: Modakafusp alfa (TAK-573) is a first-in-class immunocytokine designed
to deliver attenuated interferon alpha-2b to CD38+ cells. In this phase 1/2 study
(NCT03215030), the maximum tolerated dose of modakafusp alfa was defined as 3 mg/kg
every 4 weeks (Q4W); preliminary data from 29 pts treated at 1.5 mg/kg Q4W (5 in dose
escalation, 24 in dose expansion) showed single-agent, anti-myeloma activity with
an overall response rate (ORR) of 38% after a median follow-up of 4.2 months (mos)
(Vogl ASH 2021, #898).
Aims: We present a clinical update on the efficacy and safety of modakafusp alfa 1.5 mg/kg
Q4W after a median follow-up of 4.5 mos. We also present PK, immunogenicity, and dose-exposure-response
data from the overall study.
Methods: Eligible pts had received ≥3 prior lines of treatment. Pts received modakafusp
alfa as a 1–4-hour IV infusion at 10 dose levels from 0.001–6 mg/kg at weekly (cycles
1–2 only; 0.001–0.75 mg/kg), every-2-week (0.2–0.3 mg/kg), every-3-week (Q3W; 0.4–0.75 mg/kg),
or Q4W (0.75–6 mg/kg) dosing intervals; an expansion cohort was opened at 1.5 mg/kg
Q4W after anti-myeloma activity was observed in 3/5 pts in the escalation cohort at
that dose. Serum samples prepared from blood were used for PK and anti-modakafusp
alfa antibody (ADA) assessments. Modakafusp alfa concentrations were determined via
a validated enzyme-linked immunosorbent assay; ADA were detected using a validated
bridging electrochemiluminescence assay.
Results: As of Oct 2021, 30 pts had received modakafusp alfa 1.5 mg/kg Q4W. The median
number of prior lines was 7 (range 3–16); 90% of pts were refractory to an anti-CD38
monoclonal antibody (mAb), while 37% were exposed to an anti-B-cell maturation agent
(BCMA). Grade (G) 3–4 treatment-emergent adverse events (TEAEs) were reported in 25
pts (83%); the most frequent G3–4 TEAEs were neutropenia in 19 (63%), thrombocytopenia
in 13 (43%), leukopenia in 12 (40%), anemia in 9 (30%), and decreased lymphocyte count
in 9 (30%) pts. As reported previously, 1 pt in the 1.5 mg/kg Q4W cohort had a G3
bleeding event, while 3 pts had G3 infections. Among all pts, the ORR was 40% (complete
response, 7%; very good partial response, 20%; partial response, 13%); among anti-CD38
mAb-refractory and anti BMCA-exposed pts, the ORR was 37% and 27%, respectively. Median
progression-free survival was 6 mos and median duration of response had not been reached
(Kaplan-Meier estimate, 91% at 6 mos). Based on pooled available data for the Q3W
and Q4W cohorts (all doses) within the escalation and expansion phases, there was
a strong trend for a dose-exposure-response relationship for ORR, with the apparent
inflection point at 1.5 mg/kg Q4W dosing. No apparent relationship between dose-exposure-response
and G3/4 thrombocytopenia or neutropenia was observed across all doses, whereas a
correlation between dose-exposure-response and incidence of infusion-related reactions
across the 0.4–6 mg/kg Q3W and Q4W dosing cohorts was found. There was an apparent
non-linear (more than dose proportional) increase in exposure in the dose range of
0.1–3 mg/kg with a geometric mean terminal half-life of 13 hours in serum for modakafusp
alfa 1.5 mg/kg. Based on the available data, 14% and 60% of pts were ADA positive
at baseline and post-treatment, respectively.
Summary/Conclusion: Modakafusp alfa has a novel mechanism of action and has shown
encouraging activity with a manageable safety profile in pts with RRMM. Characterization
of the potential clinical impact of immunogenicity is ongoing. The optimal dose for
single-agent modakafusp alfa will be further investigated in a phase 2 study.
S182: TALQUETAMAB, A G PROTEIN-COUPLED RECEPTOR FAMILY C GROUP 5 MEMBER D X CD3 BISPECIFIC
ANTIBODY, IN RELAPSED/REFRACTORY MULTIPLE MYELOMA: UPDATED EFFICACY AND SAFETY RESULTS
FROM MONUMENTAL-1
M. C. Minnema1,*, A. Krishnan2, J. G. Berdeja3, A. Oriol4, N. W. van de Donk5, P.
Rodríguez-Otero6, D. Morillo7, M.-V. Mateos8, L. J. Costa9, J. Caers10, D. Vishwamitra11,
J. Ma11, S. Yang11, B. W. Hilder11, J. Tolbert11, J. D. Goldberg12, A. Chari13
1University Medical Center Utrecht, Utrecht, Netherlands; 2City of Hope Comprehensive
Cancer Center, Duarte, CA; 3Sarah Cannon Research Institute and Tennessee Oncology,
Nashville, TN, United States of America; 4Institut Català d’Oncologia and Institut
Josep Carreras, Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain; 5Amsterdam
University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; 6Clínica
Universidad de Navarra, Navarra; 7Hospital Universitario Fundación Jiménez Díaz, Madrid;
8University Hospital of Salamanca/IBSAL/CIC/CIBERONC, Salamanca, Spain; 9University
of Alabama at Birmingham, Birmingham, AL, United States of America; 10University of
Liège, Liège, Belgium; 11Janssen Research & Development, Spring House, PA; 12Janssen
Research & Development, Raritan, NJ; 13Mount Sinai School of Medicine, New York, NY,
United States of America
Background: G protein-coupled receptor family C group 5 member D (GPRC5D) is a promising
therapeutic target for multiple myeloma (MM) immunotherapy. GPRC5D has limited expression
in healthy human tissue but is highly expressed on malignant plasma cells. Talquetamab
(JNJ-64407564) is a first-in-class, GPRC5D x CD3 bispecific IgG4 antibody that induces
T cell–activated lysis of GPRC5D+ MM. MonumenTAL-1 is a phase 1 trial evaluating the
safety and preliminary efficacy of talquetamab in patients with relapsed/refractory
MM (RRMM) (NCT03399799).
Aims: We report updated results from MonumenTAL-1 with additional patients and longer
follow-up.
Methods: Patients with RRMM or who were intolerant to standard therapies were eligible;
prior treatment with B-cell maturation antigen-directed therapies was allowed. The
primary objectives were to identify the recommended phase 2 doses (RP2Ds) (part 1)
and assess the safety and tolerability of talquetamab at the RP2Ds (part 2). Based
on the collective safety, preliminary efficacy, pharmacokinetics (PK), and pharmacodynamics
(PD) data, 2 RP2Ds of talquetamab were identified: 405 μg/kg SC QW (n=30) and 800
μg/kg SC Q2W (n=44). To mitigate severe cytokine release syndrome (CRS), step-up dosing
was used. Administration of required premedications were limited to step-up doses
and the first full dose of talquetamab. Adverse events (AEs) were graded by CTCAE
v4.03, and CRS events were graded per Lee et al 2014 criteria. Responses were assessed
by the investigators as per International Myeloma Working Group criteria.
Results: As of Jan 17, 2022, patients in the 405 μg/kg /800 μg/kg groups, respectively,
received a median of 6/5 prior lines of therapy, 100%/98% were triple-class (TC) exposed,
and 77%/75% were TC refractory, and median follow-up (range) was 11.7 (1.0–21.2)/4.2
(0.7–13.7) months. AEs were mostly grade 1 or 2, and cytopenias and CRS were the most
commonly reported AEs. Cytopenias, including neutropenia (67%/36%; grade 3/4: 53%/23%),
were mostly limited to step-up and cycle 1–2 doses, reversible, and generally resolved
within 1 week. CRS (77%/80%; grade 3: 3%/0%) were mostly reported during step-up dosing.
Infections were reported in 47%/34% (grade 3/4: 7%/9%) of patients. 83%/75% of patients
reported skin-related and nail disorder AEs (most commonly skin exfoliation: 37%/39%
[all grade 1 and 2]). Dysgeusia (63%/57%) was generally mild and managed with dose
adjustments. Response-evaluable patients had overall response rates of 70% (21/30
patients)/64% (28/44 patients). Very good partial response or better rates were 57%/52%,
and median time to first confirmed response (range) was 0.9 (0.2–3.8)/1.2 (0.3–6.8)
months. Median duration of response will be reported. No deaths from drug-related
AEs were reported. Both RP2Ds showed comparable PK and PD profiles.
Summary/Conclusion: Both RP2Ds of talquetamab appear tolerable with comparable safety
and PK profiles. Talquetamab demonstrated highly promising efficacy as a novel, first-in-class
therapy for heavily pretreated patients with RRMM.
S183: NOVEL COMBINATION IMMUNOTHERAPY FOR THE TREATMENT OF RELAPSED/REFRACTORY MULTIPLE
MYELOMA: UPDATED PHASE 1B RESULTS FOR TALQUETAMAB (A GPRC5D X CD3 BISPECIFIC ANTIBODY)
IN COMBINATION WITH DARATUMUMAB
N. W. van de Donk1,*, N. Bahlis2, M.-V. Mateos3, K. Weisel4, B. Dholaria5, A. L. Garfall6,
H. Goldschmidt7, T. G. Martin8, D. Morillo9, D. E. Reece10, D. Hurd11, P. Rodríguez-Otero12,
M. Bhutani13, A. D’Souza14, A. Oriol15, E. Askari16, J. F. San-Miguel17, K. M. Kortüm18,
D. Vishwamitra19, S. Xin Wang Lin19, T. J. Prior19, L. Vandenberk20, M.-A. D. Smit21,
J. D. Goldberg22, R. Wäsch23, A. Chari24
1Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands;
2Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada; 3University
Hospital of Salamanca/IBSAL/CIC/CIBERONC, Salamanca, Spain; 4University Medical Center
Hamburg-Eppendorf, Hamburg, Germany; 5Vanderbilt University Medical Center, Nashville,
TN; 6Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA, United States of America; 7University Hospital Heidelberg, Internal
Medicine and National Center for Tumor Diseases, Heidelberg, Germany; 8University
of California, San Francisco, San Francisco, CA, United States of America; 9Hospital
Universitario Fundación Jiménez Díaz, Madrid, Spain; 10Princess Margaret Cancer Centre,
Toronto, ON, Canada; 11Section on Hematology and Oncology, Department of Internal
Medicine, Comprehensive Cancer Center of Wake Forest University, Winston-Salem, NC,
United States of America; 12Clínica Universidad de Navarra, Navarra, Spain; 13Levine
Cancer Institute/Atrium Health, Charlotte, NC; 14Medical College of Wisconsin, Milwaukee,
WI, United States of America; 15Institut Català d’Oncologia and Institut Josep Carreras
and Hospital Germans Trias i Pujol, Badalona, Barcelona; 16Department of Hematology,
Fundacion Jimenez Diaz University Hospital, Madrid; 17Clínica Universidad de Navarra,
CCUN, CIMA, CIBERONC, IDISNA, Pamplona, Spain; 18Medizinische Klinik II, Universitätsklinikum
Würzburg, Würzburg, Germany; 19Janssen Research & Development, Spring House, PA, United
States of America; 20Janssen Research & Development, Beerse, Belgium; 21Janssen Research
& Development, Los Angeles, CA; 22Janssen Research & Development, Raritan, NJ, United
States of America; 23University of Freiburg, Freiburg, Germany; 24Mount Sinai School
of Medicine, New York, NY, United States of America
Background: Talquetamab (tal; JNJ-64407564) is a first-in-class, bispecific IgG4 antibody
that binds both to G protein-coupled receptor family C group 5 member D (GPRC5D),
a receptor highly expressed on malignant plasma cells but with limited expression
in healthy tissue, and CD3 to mediate T-cell–activated lysis of GPRC5D+ multiple myeloma
(MM) cells. Daratumumab (dara) is an anti-CD38 mAb with direct on-tumor and immunomodulatory
actions. Initial clinical results from the phase 1b multicohort TRIMM-2 study identified
the recommended phase 2 doses (RP2Ds) of tal as 400 μg/kg weekly or 800 μg/kg Q2W
and support the combination of tal + dara for the treatment of RRMM, with manageable
safety, no overlapping toxicities, and promising efficacy.
Aims: Here we report updated results for both RP2Ds of tal + dara in TRIMM-2 with
additional patients (pts) and longer follow-up.
Methods: Eligible MM pts (aged ≥18 years) had received ≥3 prior lines of therapy (LOT;
including a PI and IMiD) or were double refractory to a PI and an IMiD, and could
not have received anti-CD38 therapy within 90 days. Pts received dara SC 1800 mg per
approved schedule and tal (400 μg/kg weekly or 800 μg/kg Q2W) with step-up dosing.
The primary objectives were to identify the RP2D(s) of tal for combination therapy
and evaluate safety of the combination. AEs were graded per CTCAE v5.0; cytokine release
syndrome (CRS) and immune effector cell–associated neurotoxicity syndrome (ICANS)
were graded per ASTCT guidelines. Responses were assessed by IMWG criteria.
Results: At data cutoff (Jan 13, 2022, N=46), median follow-up was 4.0 months (range
0.4-16.3), median age was 65 years (range 47-81), and 48% were female. Pts received
a median of 5 prior LOT (range 2-16); 83% were triple-class exposed, 61% penta-drug
exposed, 37% anti-BCMA non–CAR-T exposed, and 4% anti-BCMA CAR-T exposed. 96% of pts
had ≥1 AE (gr 3/4: 67%). The most frequently reported AEs (≥30% across tal + dara
cohorts) were CRS (65%; all gr 1/2; median time to onset: 2 days; median duration:
2 days), dysgeusia (57%), thrombocytopenia (35%; gr 3/4: 20%), anemia (39%; gr 3/4:
20%), and dry mouth (44%). Infections occurred in 50% of pts (gr 3/4: 13%). Skin disorders
were reported in 72% of pts (gr 3/4: 11%): skin exfoliation in 26% (all gr 1/2) and
nail disorders in 11% (all gr 1/2). Two ICANS events were reported in the 800 Q2W
group (both gr 1 and resolved within 1 day). 3 pts discontinued due to AEs. Response
rates were consistent across both RP2Ds supporting their equivalence (Table). Median
time to first response across dosing cohorts was 0.95 months (range 0.9-9.7); median
duration of response was not reached. Upregulation of CD38+/CD8+ T cells and proinflammatory
cytokines was observed with tal + dara, supporting potential synergy of the combination
in pts with prior anti-CD38 exposure. Updated results will be presented.
Image:
Summary/Conclusion: Longer follow-up with additional patients shows comparable efficacy
and safety across both RP2Ds, with no new safety signals, strengthening the benefit-risk
profile of tal + dara as a novel immunotherapy-based approach for heavily pretreated
pts with RRMM.
S184: EVALUATING TECLISTAMAB IN PATIENTS WITH RELAPSED/ REFRACTORY MULTIPLE MYELOMA
FOLLOWING EXPOSURE TO OTHER B-CELL MATURATION ANTIGEN (BCMA)-TARGETED AGENTS
C. Touzeau1,*, A. Krishnan2, P. Moreau1, A. Perrot3, S. Z. Usmani4, S. Manier5, M.
Cavo6, C. Martinez-Chamorro7, A. Nooka8, T. Martin9, L. Karlin10, X. Leleu11, N. Bahlis12,
B. Besemer13, L. Pei14, R. Verona15, S. Girgis15, C. Uhlar15, R. Kobos14, A. Garfall16
1University Hospital Hôtel-Dieu, Nantes, France; 2City of Hope Comprehensive Cancer
Center, Duarte, United States of America; 3Centre Hospitalier, Universitaire de Toulouse,
Service d’Hematologie, Toulouse, France; 4Levine Cancer Institute/Atrium Health, Charlotte,
United States of America; 5University of Lille, Lille, France; 6IRCSS Aziena Ospedaliero-Universitaria
di Bologna, Bologna University School of Medicine, Bologna, Italy; 7University Hospital
Quirónsalud, Pozuelo de Alarcón, Madrid, Spain; 8Winship Cancer Institute, Emory University,
Atlanta; 9University of California, San Francisco, San Francisco, United States of
America; 10Service d’Hématologie Clinique, Centre Hospitalier Lyon Sud, Pierre-Bénite;
11Centre Hospitalier Universitaire de Poitiers, Poitiers, France; 12Arnie Charbonneau
Cancer Institute, University of Calgary, Calgary, Canada; 13University of Tuebingen,
Tuebingen, Germany; 14Janssen Research & Development, Raritan; 15Janssen Research
& Development, Spring House; 16Abramson Cancer Center, Perelman School of Medicine,
Philadelphia, United States of America
Background: Teclistamab (JNJ-64007957) is a T cell redirecting bispecific antibody
that targets both B-cell maturation antigen (BCMA) and CD3 receptors to induce T cell
mediated cytotoxicity of BCMA-expressing myeloma cells. MajesTEC-1 is an open-label,
multicohort, phase 1/2 study evaluating teclistamab in patients (pts) with relapsed/
refractory multiple myeloma (RRMM) previously treated with ≥3 prior lines of therapy
(LOT). An overall response rate (ORR) of 62.0% in pts with no prior anti-BCMA treatment
(tx) was previously reported in a pooled analysis from phase 1 and phase 2 cohort
A at a median follow-up of 7.8 mo.
Aims: We report efficacy and safety results of teclistamab from cohort C, which enrolled
patients who had prior exposure to anti-BCMA treatment.
Methods: Eligible pts (age ≥18 y) had multiple myeloma (MM) per IMWG criteria and
were previously treated with ≥3 prior LOT, including a PI, IMiD, anti-CD38 antibody,
and anti-BCMA treatment (chimeric antigen receptor T cell therapy [CAR-T] or Ab drug
conjugate [ADC]). Pts were enrolled using a Simon’s stage design to receive weekly
subcutaneous teclistamab 1.5 mg/kg (step-up doses of 0.06 and 0.3 mg/kg). ORR per
IMWG 2016 criteria was the primary endpoint. All AEs were graded per CTCAE v4.03;
immune effector cell–associated neurotoxicity syndrome (ICANS) and cytokine release
syndrome (CRS) were graded per ASTCT guidelines.
Results: In cohort C, 38 pts (median age 63.5 y [range 32–82]; 63% male) received
teclistamab (median prior LOT 6 [range 3–14]) at the data cutoff date of Sep 7, 2021.
Of the 38 patients, 25 (66%) were refractory to an anti-BCMA treatment, and 32 (84%)
were refractory to last LOT. Among 25 efficacy-evaluable patients, 16 (64%) received
prior ADC, 11 (44%) received prior CAR-T, and 2 pts received both. The ORR was 40%
(95% CI, 21–61) at a median follow-up of 6.9 mo (range 0.7–8.7). Complete response
or better were observed in 5 pts (20%). In ADC-exposed and CAR-T-exposed pts, the
ORR was 38% (95% CI, 15–65) and 45% (95% CI, 17–77), respectively. Responses were
rapid in most, with deepening of responses over time in 7 of 25 pts. While the median
duration of response was not reached, median time to first response was 1.2 mo (range,
0.2–4.9) and to best response was 2.1 mo (range, 1.1–5.7). No new safety concerns
were observed, and the safety profile was comparable with that of BMCA tx-naive pts.
Infections were reported in 16 pts (42%; grade 3/4, 26%). Most common AEs (n=38) were
CRS (63%; all grade 1/2; median time to CRS onset: 3 d [range, 2–6], duration of CRS:
2 d [range, 1–4]), thrombocytopenia (42%; grade 3/4, 29%), neutropenia (55%; grade
3/4, 50%), lymphopenia (40%; grade 3/4, 37%), and anemia (39%; grade 3/4 29%), Grade
3 ICANS was reported in 1 pt which resolved with supportive care and the pt remained
on tx. Anti-teclistamab Abs were not detected in any pts. Baseline BCMA expression
levels were comparable with those reported in BCMA tx-naive pts. Updated efficacy
and safety results will be presented for 40 pts.
Summary/Conclusion: These preliminary results observed with serial targeting of BCMA
with teclistamab following ADC or CAR-T tx suggest a promising ORR with early responses
that deepen over time. Additionally, a well-tolerated safety profile was observed
in patients treated with anti-BCMA tx.
S185: CARTITUDE-2 COHORT B: UPDATED CLINICAL DATA AND BIOLOGICAL CORRELATIVE ANALYSES
OF CILTACABTAGENE AUTOLEUCEL IN PATIENTS WITH MULTIPLE MYELOMA AND EARLY RELAPSE AFTER
INITIAL THERAPY
M. E. Agha1,*, N. W. van de Donk2, A. D. Cohen3, Y. C. Cohen4, S. Anguille5, T. Kerre6,
W. Roeloffzen7, J. M. Schecter8, K. C. De Braganca8, H. Varsos8, P. Mistry9, T. Roccia9,
E. Zudaire10, C. Corsale8, M. Akram11, D. Geng11, T Nesheiwat11, L. Pacaud11, P. Sonneveld12,
S. Zweegman2
1UPMC Hillman Cancer Center, Pittsburgh, PA, United States of America; 2Amsterdam
UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; 3Abramson Cancer Center,
University of Pennsylvania, Philadelphia, PA, United States of America; 4Tel-Aviv
Sourasky (Ichilov) Medical Center and Sackler School of Medicine, Tel Aviv University,
Tel Aviv, Israel; 5Vaccine and Infectious Disease Institute, University of Antwerp,
Edegem, Belgium, Center for Cell Therapy and Regenerative Medicine, Antwerp University
Hospital, Edegem, Belgium; 6University Hospital Ghent, Ghent, Belgium; 7University
Medical Center Groningen, Groningen, Netherlands; 8Janssen R&D, Raritan, NJ, United
States of America; 9Janssen R&D, High Wycombe, United Kingdom; 10Janssen R&D, Spring
House, PA, United States of America; 11Legend Biotech USA, Piscataway, NJ, United
States of America; 12Erasmus MC University and Medical Center, Rotterdam, Netherlands
Background: CARTITUDE-2 (NCT04133636) is a phase 2, multicohort study evaluating the
efficacy and safety of the chimeric antigen receptor (CAR) T-cell therapy ciltacabtagene
autoleucel (cilta-cel) in patients (pts) with multiple myeloma (MM). Cohort B enrolled
pts who had early relapse after initial therapy. These pts have functionally high-risk
disease, as early relapse following autologous stem cell transplantation (ASCT) is
associated with poor prognosis, representing an unmet medical need.
Aims: To present updated results from CARTITUDE-2 cohort B.
Methods: All pts provided informed consent. Pts had MM, received 1 prior line of therapy
(proteasome inhibitor and immunomodulatory drug required), had disease progression
per International Myeloma Working Group criteria (either ≤12 months after ASCT or
≤12 months after initiation of anti-myeloma therapy for pts not treated with ASCT),
and were naive to CAR-T or other anti-B-cell maturation antigen therapies. After lymphodepletion,
a single cilta-cel infusion was administered at a target dose of 0.75×106 CAR+ viable
T cells/kg. Efficacy and safety were evaluated. The primary endpoint was minimal residual
disease (MRD) negativity at 10-5. Patient management strategies were used to minimize
risk of movement and neurocognitive adverse events (MNTs). Pharmacokinetic (PK) analyses,
including Cmax and Tmax of CAR+ T-cell transgene levels in blood are being performed.
Additional assessments include levels of cytokine release syndrome (CRS)-related cytokines
(eg, IL-6) over time, peak levels of cytokines by response and CRS, association of
cytokine levels with immune effector cell-associated neurotoxicity syndrome (ICANS),
and CAR+ T cell CD4/CD8 ratio by response, CRS, and ICANS.
Results: 19 pts (median age: 58.0 years [range: 44-67]; 74% male) received cilta-cel
as of January 2022; 79% received prior ASCT. Median follow-up was 13.4 months (range:
5.2-21.7). Overall response rate was 100.0%, with 90% of pts achieving ≥complete response,
and 95% achieving ≥very good partial response. Median time to first response was 0.95
months (range: 0.9-9.7) and median time to best response was 5.1 months (range: 0.9-11.8).
Among 15 MRD-evaluable pts, 14 (93%) achieved MRD 10-5 negativity. Median duration
of response was not reached. The 12-month progression-free survival rate was 90%;
12-month event-free rate was 88.9%. CRS occurred in 16 (84.2%) pts (1 grade 4); median
time to onset was 8 days (range: 5-11) and all events resolved. ICANS (grade 1) and
MNT (grade 3, previously reported) occurred in 1 pt each. 1 pt died post cilta-cel
(progressive disease, day 158). Preliminary PK data showed peak expansion of CAR-T
cells on day 13.1 (range: 8.96-209.9); median persistence was 76.9 days (range: 40.99-221.8).
Summary/Conclusion: This functionally high-risk pt population with early relapse after
initial therapy experienced deep and durable responses with manageable safety following
a single cilta-cel infusion. Cilta-cel led to responses in pts with ineffective or
insufficient response to ASCT. Follow-up is ongoing and responses continue to deepen.
We will present updated and detailed PK, cytokine, and CAR-T subset analyses as well
as clinical correlation to provide insight into biological correlates of efficacy
and safety in these pts.
S186: UPDATED RESULTS OF A MULTICENTER FIRST-IN-HUMAN STUDY OF BCMA/CD19 DUAL-TARGETING
FAST CAR-T GC012F FOR PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA (RRMM)
J. Du1,*, H. Jiang1, B. Dong2, L. Gao3, L. Liu4, J. Ge5, A. He6, L. Li1, J. Lu1, X.
Chen2, M. Sersch7, H. Zhang7, L. Shen7, J. Liu7, W. Fu1
1Changzheng Hospital, Shanghai; 2Xijing Hospital, Xian; 3Chongqing Xinqiao hospital,
Chongqing; 4Tangdu Hospital, Xian; 5The first affiliated hospital of Anhui medical
University, Anhui; 6The second affiliated hospital of Xian Jiaotong University, Xian;
7Gracellbiotechnologies Ltd, Shanghai, China
Background: GC012F is a B cell maturation antigen (BCMA) and CD19 dual-targeting CAR-T
developed on the novel FasT CAR-T platform enabling 22-36h manufacturing designed
to improve depth of response and overall efficacy. Data was presented at ASCO and
EHA 2021 for initial 19 pts. Here we present updated data for study (NCT04236011;
NCT04182581) with longer follow up and 9 additional pts treated (total n=28) in 3
different dose levels.
Aims: The study aims to assess safety and preliminary efficacy for FAST-CAR GC012F
in RRMM patients.
Methods: From October 2019 to November 2021, 28 heavily pretreated RRMM pts (age 27-76)
with a median of 5 prior lines (range 2-9) were treated on this single-arm, open label,
multicenter Investigator Initiated Trial receiving a single infusion of GC012F. 89.3%
(25/28) were high risk (HR- mSMART), 8 pts had EM disease, 3 had never achieved a
CR including after transplant, 1 pts presented with plasma cell leukemia, 24/28 pts
were refractory to last therapy, 3 pts primary refractory. 9/28 pts had received prior
anti-CD38, 27/28 pts prior IMiDs. 26/28 pts were refractory to PI, 26/28 pts to IMiDs.
After lymphodepletion over 2-3 days (30 mg/m2/d, 300mg/ m2/d Flu/Cy) GC012F was administered
as single infusion at 3 dose levels: 1x105/kg (DL1) n=2, 2x105/kg (DL2) n=10 and 3x105/kg
(DL3) n=16.
Results: As of Jan 26th 2022 cut-off, 28 pts - median follow-up (f/u) 6.3 mths (1.8-29.9)
- had been evaluated for response. Overall response rate (ORR) in DL1 was 100% (2/2)-
DL 2 -80% (8/10) DL 3 -93.8% (15/16) with 27 pts MRD negative by flow cytometry (sensitivity
10-4-10-6). 100% of MRD assessable pts (27/27) achieved MRD negativity. One patient
out of 28 could not get assessed. At d28, 21/24 assessable patients were MRD negative
(81.5%), 4/28 pts could not get d28 MRD assessment f/u due to COVID-19 restrictions
however were assessed at a later timepoint. To date best response is MRD- sCR in 21/28
patients (75.0%) across all dose levels. Some pts after short f/u show responses that
are still deepening. Cytokine Release Syndrome (CRS) was mostly low grade: gr 0 n=3
(10.7%), gr 1-2 n=23 (82.1%), gr 3 n=2 (7.1%) – no gr 4/5 CRS and no ICANs were observed
(Graded by ASBMT criteria). Median duration of CRS was 3 d (1-8 d). PK results showed
no difference amongst dose levels DL1 to DL3. Overall, CAR-T median Tmax was 10 d
(range 8-14 d), median peak copy number (Cmax) was 97009 (16,011-374,346) copies /ug
DNA with long duration of persistence of up to d793 (data cut-off). CAR-T geometric
mean AUC0-28 for DL1, DL2 and DL3 were 468863, 631540 and 581620 copies/ug DNA×day,
respectively. Pts continue to be monitored for safety and efficacy including DOR.
Summary/Conclusion: BCMA-CD19 dual FasT CAR-T GC012F continues to provide deep and
durable responses with a favorable safety profile in additional RRMM pts across all
dose levels demonstrating a very high MRD negativity rate including in pts refractory
to anti-CD38, PI and IMIDs. Based on these promising results GC012F is being studied
in earlier lines of therapy as well as additional indications.
S187: UPDATED PHASE 1/2 DATA OF SAFETY AND EFFICACY OF CT103A, FULLY HUMAN BCMA-DIRECTED
CAR T CELLS, IN RELAPSED/REFRACTORY MULTIPLE MYELOMA
C. Li1,*, D. Wang1, Y. Song2, J. Li3, H Huang4, B. Chen5, J. Liu6, K. Hu7, H. Ren8,
X. Zhang9, Z. Li10, D. Zou11, Q. Yin2, L. Chen12, Y. Hu4, Y. Xu5, Q. Cheng6, Y. Guo7,
Y. Dong13, L. Gao9, S. Chen14, A. Xu15, S. Cai15, M. Wu15, J. Guo15, Z. Yao15, W.
Wang15, J. Wang1, L. Chen1, J. Zhou1, L. Qiu11
1Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University
of Science and Technology, Wuhan; 2Department of Hematology, Affiliated Cancer Hospital
of Zhengzhou University and Henan Cancer Hospital, Zhengzhou; 3Department of Hematology,
Pukou CLL Center, The First Affiliated Hospital of Nanjing Medical University, Jiangsu
Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine,
Nanjing; 4Department of Hematology, Bone Marrow Transplantation Center, the First
Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou; 5Department
of Hematology, Nanjing University Medical School, the Affiliated Nanjing Drum Tower
Hospital, Nanjing; 6Department of Hematology, The Third Xiangya Hospital of Central
South University, Changsha; 7Department of Hematology, Beijing Boren Hospital; 8Department
of Hematology, Peking University First Hospital, Beijing; 9Department of Hematology,
Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn
and Combined Injury, Army Medical University, Chongqing; 10Department of Hematology,
Affiliated Hospital of Xuzhou Medical University, Xuzhou; 11Department of Hematology,
State Key Laboratory of Experimental Hematology, National Clinical Research Center
for Blood Diseases, Blood Diseases Hospital & Institute of Hematology, Chinese Academy
of Medical Sciences & Peking Union Medical College, Tianjin; 12Department of Hematology,
The First Affiliated Hospital of Nanjing Medical University, Nanjing; 13Department
of Hematology, Peking University First Hospital, Beijing; 14Department of Hematology;
15Nanjing IASO Biotherapeutics Ltd, Nanjing, China
Background: CT103A, a fully human BCMA-directed CAR-T therapy, showed excellent safety
and promising efficacy in the ongoing phase 1/2 FUMANBA-1 study (ChiCTR1800018137,
NCT05066646) in patients with relapsed/refractory multiple myeloma (Wang, ASH, 2021).
Aims: We report updated data from the FUMANBA-1 study with more subjects and a longer
duration of follow-up.
Methods: Enrolled patients had received three or more prior lines of anti-myeloma
therapy, including a proteasome inhibitor and an immunomodulatory agent, and were
required to be refractory to the last line of therapy. CT103A was administered at
RP2D of 1.0×106 CAR+ T cells/kg. Lymphodepletion with cyclophosphamide (500 mg/m2)
and fludarabine (30 mg/m2) was used for 3 consecutive days. Study objectives included
safety and efficacy profile. Pharmacokinetics, pharmacodynamics, and immunogenicity
were also explored. MRD in bone marrow aspirate was detected by EuroflowTM standardized
flow cytometry with the sensitivity of 10-5. Adverse events (AEs) were graded using
CTCAEv5.0, except that CRS and ICANS were graded by ASTCT criteria.
Results: As of Jan 21, 2022, 79 patients received CT103A with the median follow-up
of 9.6 months (range 1.2, 40.0). 28 (35.4%) patients had high-risk cytogenetic abnormality
defined as del(17p), t(4;14), or t(14;16), and 11 (13.9%) had extramedullary plasmacytomas.
Patients received a median of 5 (range 3, 23) prior lines of therapy. Notably, 13
(16.5%) had received prior non-human BCMA-targeted CAR-T cell therapy.
ORR was 94.9% (75/79), with 55 (69.6%) patients achieving CR/sCR, and 71 (89.9%) achieving
≥VGPR. Median TTR was 16 days (range 11, 123), and the median time to CR/sCR was 95
days (range 14, 557). For 13 patients who had prior CAR-T therapy, ORR was 98.5%,
with 6 (46.2%) achieving CR/sCR. For 11 patients with extramedullary disease at baseline,
ORR was 100%, with 9 (81.8%) achieving CR/sCR. All patients with CR/sCR were MRD-negative.
The median duration of MRD negativity was not reached. The probability of MRD negativity
at month 12 was 88.7% (95% CI 65.9%, 96.6%).
CRS occurred in 75 (94.9%) patients, of which only 2 (2.5%) had grade 3 CRS. The median
time to onset of CRS was six days (range 1, 12) with a median duration of 5 days (range
2, 30). ICANS occurred in only 2 (2.5%) patients, of which one was grade 1, and the
other was grade 2. All CRS were resolved, with the use of tocilizumab in 19 (25.3%)
patients and steroids in 47 (62.7%). The most common ≥ grade 3 treatment-related AEs
were hematological toxicities, including neutropenia (78.5%), leukopenia (74.7%),
lymphopenia (55.7%), thrombocytopenia (54.4%), and anemia (46.8%).
CT103A reached the peak at a median of 12 days (range 0, 26) after infusion and were
still detectable in 40 of 61 evaluable patients (65.6%) at month 6 and 13 of 21 (61.9%)
at month 12. Soluble BCMA decreased to the lower limit of quantification (LLOQ) at
a median of 2 months and can maintain bellow LLOQ over 24 months. 11 (13.9%) patients
were positive for anti-drug antibodies, of whom 1/76 (1.3%) developed anti-drug antibodies
at month 3, 5/43 (11.6%) at month 6 and 5/16 (31.3%) at month 12. Among these, neutralizing
antibodies were only identified in one patient (at month 12).
Summary/Conclusion: Updated data from the Phase 1/2 FUMANBA-1 study continue to show
encouraging efficacy of CT103A with a favorable safety profile in relapsed/refractory
multiple myeloma. CT103A led to a deepening and durable response with robust expansion
and prolonged persistence. Patients with prior BCMA CAR-T therapy could still benefit
from CT103A.
S188: TECLISTAMAB IN COMBINATION WITH DARATUMUMAB, A NOVEL, IMMUNOTHERAPY-BASED APPROACH
FOR THE TREATMENT OF RELAPSED/REFRACTORY MULTIPLE MYELOMA: UPDATED PHASE 1B RESULTS
P. Rodriguez Otero1,*, A. D’Souza2, D. Reece3, N. W. van de Donk4, A. Chari5, A. Krishnan6,
T. Martin7, M. V. Mateos8, D. Morillo9, D. Hurd10, L. Rosinol11, A. S. Balari12, R.
Wäsch13, D. Vishwamitra14, S. X. Wang Lin14, T. Prior14, L. Vandenberk15, M.-A. D.
Smit16, A. Oriol17, B. Dholaria18
1University of Navarra, Navarra, Spain; 2Medical College of Wisconsin, Milwaukee,
WI, United States of America; 3Princess Margaret Cancer Centre, Toronto, ON, Canada;
4Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands;
5Mount Sinai School of Medicine, New York, NY; 6City of Hope Comprehensive Cancer
Center, Duarte, CA; 7UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco,
CA, United States of America; 8University Hospital of Salamanca/IBSAL/CIC/CIBERONC,
Salamanca; 9Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain; 10Comprehensive
Cancer Center of Wake Forest Baptist Health, Winston-Salem, NC, United States of America;
11Hospital Clínic, Institut d’investigacions Biomèdiques August Pi i Sunyer, Barcelona;
12Institut Català d’Oncologia – Hospitalet, IDIBELL, University of Barcelona, Barcelona,
Spain; 13Freiburg University Medical Center, Freiburg, Germany; 14Janssen Research
and Development, Spring House, PA, United States of America; 15Janssen Research &
Development, Antwerp, Belgium; 16Janssen Research & Development, Los Angeles, CA,
United States of America; 17Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain;
18Vanderbilt University Medical Center, Nashville, TN, United States of America
Background: Teclistamab (JNJ-64007957) is a B-cell maturation antigen (BCMA) × CD3
T-cell redirecting bispecific antibody currently under investigation in patients with
relapsed/refractory multiple myeloma (RRMM). Daratumumab is a CD38-targeting monoclonal
antibody with direct on-tumor and immunomodulatory mechanisms of action. The preliminary
results from the phase 1b multicohort TRIMM-2 study showed tolerable safety with no
overlapping toxicities, and encouraging efficacy, supporting the combination of teclistamab
with daratumumab for the treatment of RRMM.
Aims: We report updated results from the TRIMM-2 study with additional patients and
longer follow-up.
Methods: Eligible patients were ≥18 years of age with a MM diagnosis and previously
treated with ≥3 prior lines of therapy (including a proteosome inhibitor [PI] and
immunomodulatory drug [IMiD]) or were double-refractory to a PI and IMiD. Patients
who had received anti-CD38 therapy ≤90 days prior were excluded. Written informed
consent was obtained from all eligible patients. Patients received subcutaneous (SC)
daratumumab 1800 mg per approved schedule and teclistamab SC 1.5–3 mg/kg once weekly
or every 2 weeks. Primary objectives of the study were to identify the recommended
phase 2 dose for the teclistamab and daratumumab combination and to assess safety
of the combination. Responses were assessed by IMWG criteria. Adverse events (AEs)
were graded per CTCAE v5.0, except for cytokine release syndrome (CRS) and immune
effector cell–associated neurotoxicity syndrome (ICANS), which were graded per ASTCT
guidelines.
Results: At the Jan 13, 2022 data cutoff, the median follow-up was 7.2 months (range
0.1–16.6). Among the safety population (N=46), 52% were females, and the median age
was 67 years (range 50–79). Patients received a median of 6 prior lines of therapy
(range 2–17); 74% of patients were triple-class exposed; 63% were penta-drug exposed,
and 15% were anti-BCMA exposed. Overall, 91% of patients had ≥1 AE of any grade; 78%
had grade 3/4 AEs. The most common AE was CRS (61%; all grade 1/2); median time to
onset was 2 days and median duration was 2 days. Other AEs included neutropenia (54%;
grade 3/4 50%), anemia (46%; grade 3/4 28%), thrombocytopenia (33%; grade 3/4 28%),
and diarrhea (33%; grade 3/4 2%). Infections occurred in 29 patients (63%; grade 3/4
28%). One patient had grade 1 ICANS that was fully resolved. Among 37 response-evaluable
patients, the overall response rate was 78% (29/37); 27 patients (73%) had very good
partial response (VGPR) or better (Table). While the median duration of response was
not reached, median time to first response across dosing cohorts was 1.0 month (range
0.9–2.8). Upregulation of CD38+/CD8+ T cells and proinflammatory cytokines was observed
after teclistamab dosing in combination with daratumumab, supporting potential synergy
of the combination in patients with prior anti-CD38 exposure. Updated results will
be presented.
Image:
Summary/Conclusion: Teclistamab in combination with daratumumab is a novel immunotherapy
approach that may yield improved clinical efficacy in heavily pretreated patients
with RRMM.
S189: EARLY, DEEP, AND DURABLE RESPONSES, AND LOW RATES OF CRS WITH REGN5458, A BCMAXCD3
BISPECIFIC ANTIBODY, IN A PHASE 1/2 FIRST-IN-HUMAN STUDY IN PATIENTS WITH RELAPSED/REFRACTORY
MULTIPLE MYELOMA
J. A. Zonder1, J. Richter2,*, N. Bumma3, J. Brayer4, J. E. Hoffman5, W. I. Bensinger6,
K. L. Wu7, L. Xu8, D. Chokshi8, A. Boyapati8, D. Cronier8, Y. Houvras8, K. Rodriguez
Lorenc8, G. S. Kroog8, M. V. Dhodapkar9, S. Lentzsch10, D. Cooper11, S. Jagannath2
1Karmanos Cancer Institute, Detroit; 2Icahn School of Medicine at Mount Sinai, New
York; 3The Ohio State University Comprehensive Cancer Center, Columbus; 4H. Lee Moffitt
Cancer Center, Tampa; 5University of Miami Health System, Miami; 6Swedish Center for
Blood Disorders and Stem Cell Transplants, Seattle, United States of America; 7Ziekenhuis
Netwerk Antwerpen Stuivenberg, Antwerp, Belgium; 8Regeneron Pharmaceuticals, Inc,
Tarrytown; 9Emory University School of Medicine, Atlanta; 10Columbia University Medical
Center, New York; 11Rutgers Cancer Institute of New Jersey, New Brunswick, United
States of America
Background: Despite recent advances in treatment options, multiple myeloma (MM) remains
incurable. REGN5458 is a BCMAxCD3 bispecific antibody currently under investigation
in relapsed/refractory MM (RRMM) in an ongoing Phase 1/2 trial (NCT03761108). Preliminary
data suggest that REGN5458 has a manageable safety profile with early, deep and durable
responses in heavily pretreated patients (pts).
Aims: Here we describe updated safety, overall response and response durability in
pts treated with REGN5458 in the Phase 1 portion of this ongoing trial.
Methods: The Phase 1 primary objectives are to assess safety, tolerability and occurrence
of dose-limiting toxicities of REGN5458 and to determine a recommended Phase 2 dose
regimen (RP2DR). Key secondary objectives include: assessment of objective response
rate as determined by the investigator, duration of response (DOR) and minimal residual
disease status; pharmacokinetic evaluation; and characterization of immunogenicity.
Pts with progressive RRMM, who are double- or triple-refractory, or intolerant to
prior lines of systemic therapy including a proteasome inhibitor, immunomodulatory
agent, and anti-CD38 antibody are treated with REGN5458 monotherapy following a modified
3 + 3 dose-escalation design (4 + 3). Treatment consists of 16 weekly infusions of
REGN5458 followed by q2w dosing until disease progression. Response assessments are
measured using modified International Myeloma Working Group criteria.
Results: At data cut-off (September 30, 2021), 73 pts were treated with REGN5458 in
the dose escalation cohort with full doses ranging from 3–800 mg. Median age at enrollment
was 64 years (range, 41–81) and 20.5% pts were ≥75 years. As per Revised International
Staging System, stage was 1, 2 or 3 in 15.0%, 57.5% and 23.3% of pts respectively.
Pts had a median of 5 prior lines of systemic therapy (range, 2–17) with 38.4% of
pts being penta-refractory (Table). Median duration of follow-up was 3.0 months (range,
0.7–22.1).
Treatment-emergent adverse events (TEAE) were reported in 73 pts (100%), Grade (Gr)
3 in 31 pts (42.5%), Gr 4 in 24 pts (32.9%). The most common Gr 3/4 TEAEs were hematologic
(39.0%). The most frequent TEAEs were fatigue in 33 pts (45.2%), Gr 1/2 in 31 pts
(42.5%), Gr 3 in 2 pts (2.7%); cytokine release syndrome (CRS) in 28 pts (38.4%),
Gr 1 in 25 pts (34.2%), Gr 2 in 3 pts (4.1%). No pt had Gr ≥3 CRS or discontinued
treatment due to CRS. There were no Gr ≥3 neurotoxicity events. Nausea was reported
in 24 pts (32.9%), Gr 1 in 17 pts (23.3%), Gr 2 in 7 pts (9.6%).
Responses were observed at all dose levels. Across all dose levels, 86.5% (n=32/37)
of all responders achieved at least a very good partial response and 43.2% (n=16)
of responders had a complete response (CR) or stringent CR. Amongst pts treated at
the 200–800 mg dose levels, the response rate was 75.0% (n=18/24). The Kaplan–Meier
estimated probability of responders being in response for 8 months or more was 90.2%
(95% confidence interval: 72.6–96.7), and median DOR was not reached.
Image:
Summary/Conclusion: REGN5458 shows a manageable safety and tolerability profile, with
Gr 2 CRS in only 4.1% of pts and no Gr ≥3 CRS or neurotoxicity events. No new safety
signals were observed during the additional follow-up period. Early, deep and durable
responses were seen in triple- to penta-refractory pts with RRMM, with a 75.0% response
rate at the combined 200–800 mg dose levels. The Phase 2 portion of the study is currently
recruiting.
S190: ASXL1 MUTATIONS ACCELERATE BONE MARROW FIBROSIS VIA EGR1-TNFA AXIS MEDIATED
INFLAMMATION AND FIBROCYTE GENERATION IN MYELOPROLIFERATIVE NEOPLASMS
Z. Shi1 2,*, J. Liu1, Y. Zhao3, L. Yang1, Y. Cai1, P. Zhang4, Z. Xu1, T. Qin1, S.
Qu1, L. Pan1, J. Wu1, X. Yan1, Z. Li3, W. Zhang1, Y. Yan1, H. Huang1, G. Huang5, B.
Li6, X. Wu7 8, Z. Xiao1
1MDS/MPN center, State Key Laboratory of Experimental Hematology, National Clinical
Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital,
Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin; 2Department
of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu
Province Hospital, Nanjing; 3Department of Cell Biology, Tianjin Medical University;
4pathology center, State Key Laboratory of Experimental Hematology, National Clinical
Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital,
Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin, China;
5Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati
Children’s Hospital Medical Center, Cincinnati, United States of America; 6MDS/MPN
center, State Key Laboratory of Experimental Hematology, National Clinical Research
Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese
Academy of Medical Sciences & Peking Union Medical College; 7Department of Cell Biology,
Tianjin Medical University; 8State Key Laboratory of Experimental Hematology, National
Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases
Hospital, Chinese Academy of Medical Sciences, Tianjin, China
Background: Apart from the central role of activated JAK/STAT signaling pathway, ASXL1
mutations are the most recurrent additional mutations in myeloproliferative neoplasms
(MPNs) and occurred much more common in myelofibrosis (MF) than essential thrombocythemia
(ET) and polycythemia vera (PV) patients. However, the mechanism of the association
with ASXL1 mutations and bone marrow (BM) fibrosis remains unknown.
Aims: To investigate the role of ASXL1 mutations in MF acceleration and explore the
underlying mechanism.
Methods: We studied the clinical data of 302 consecutive MF patients in our hospital.
For mouse model study, Vav1-Cre mice, Asxl1
flox/flox
and Jak2
V617F/+
knockin alleles were used to achieve hematopoietic cell-specific Jak2
V617F/+
/Asxl1
flox/flox
(Asxl1-/-Jak2
VF
), Jak2
V617F/+
(Jak2
VF
), and Asxl1
flox/flox
(Asxl1-/-) mice. Integrated analysis of RNAseq, transposase-accessible chromatin using
sequencing (ATAC-Seq) and chromatin immunoprecipitation (ChIP)-seq were performed
to explore the underlying mechanism.
Results: 98 of 302 (32.5%) patients harbored ASXL1 mutations. ASXL1 mutations are
correlated with higher monocyte counts, increasing CD34+ cells in peripheral blood
(PB), larger spleen sizes, and higher MF grades. Consistent with clinical findings,
Asxl1-/-Jak2
VF
mice showed lower platelet counts, higher monocyte counts and c-kit+ cells in PB compared
with Jak2
VF
mice. Hematopoietic stem and progenitor cells (HSPCs) compartment analysis revealed
increased granulocyte/macrophage progenitors (GMPs), and megakaryocyte /erythroid
progenitors (MEPs) in spleens of Asxl1-/-Jak2
VF
, not in BM compared with Jak2
V617F
mice (Figure 1A), in line with increased spleen weights of Asxl1-/-Jak2
VF
mice (Figure 1B). Histology analysis revealed reticulin infiltration in BM of Asxl1-/-Jak2
VF
mice, not in other age-matched genotypes.
Both ASXL1
MT
MF patients and Asxl1-/-Jak2
VF
mice showed increased monocytes/macrophages in BM (Figure 1C-D), accounting for more
severe inflammation in ASXL1
MT
MF patients and mouse models. In vitro differentiation assay revealed monocyte/ macrophage
differentiation bias of Asxl1-/-Jak2
VF
HSPCs (Figure 1E). In fibrosis driving cells analysis, ASXL1
MT
MF patients showed comparable mesenchymal stromal cells (MSCs) derived myofibroblasts
while increased monocyte derived fibrocytes in BM compared with ASXL1
WT
MF patients (Figure 1F). In vitro fibrocyte differentiation assay (Figure 1G) and
flow cytometry analysis confirmed increased fibrocytes in BM of Asxl1-/-Jak2
VF
mice compared with other genotypes.
Mechanismly, Asxl1 deletion in Jak2
VF
HSPCs leads to enhancer activation of polycomb group (PcG) target genes, such as Egr1
(Figure 1H). The upregulation of Egr1, which was validated in mouse models and patients
(Figure 1I-J), in turn accounts for increased HSPCs commitment to monocyte/macrophage
lineage (Figure 1K). Moreover, Egr1 induces the activation of Tnfa
(Figure 1L) and thereby further drives the differentiation of monocytes to fibrocytes
(Figure 1M). Accordingly, combined TNFR antagonist (R-7050) with ruxolitinib significantly
reduces fibrocytes production in vitro in Asxl1-/-Jak2
VF
mice and ASXL1
MT
MF patients (Figure 1N-O).
Image:
Summary/Conclusion: Our study illustrated the crucial role of ASXL1 mutation in MPN
phenotypes and BM fibrosis. ASXL1 mutations activate the EGR1-TNFA axis in MPNs, leading
to monocyte/macrophage mediated inflammation and fibrocyte induced BM fibrosis. Ruxolitinib
along with TNFR antagonist may ameliorate fibrocytes, providing an attractive theoretical
approach for antifibrosis treatment.
S191: CALRETICULIN-MUTATED HEMATOPOIETIC CELLS ARE VULNERABLE TO THE COMBINED INHIBITION
OF THE PROTEASOME AND THE IRE1A-XBP1 AXIS OF THE UNFOLDED PROTEIN RESPONSE
J. S. Jutzi1,*, A. E. Marneth1, M. J. Jimenez-Santos2, A. Guerra-Moreno1, S. A. Myers3
4, S. A. Carr3, P. van Galen1, F. Al-Shahrour5, A. S. Nam6, A. Mullally1
1Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston,
United States of America; 2Bioinformatics Unit, Structural Biology Programme, Spanish
National Cancer Research Centre (CNIO), Madrid, Spain; 3The Broad Institute of MIT
and Harvard, Cambridge; 4Center for Autoimmunity and Inflammation, La Jolla Institute
for Immunology, La Jolla; 5Bioinformatics Unit, Structural Biology Programme, Spanish
National Cancer Research Centre (CNIO), Madrid; 6Weill Cornell Medicine, New York,
United States of America
Background: Mutations in the endoplasmic reticulum (ER) chaperone calreticulin (CALR)
are frequent and disease-initiating in myeloproliferative neoplasms (MPN). These mutant
CALR proteins have impaired chaperone function. Concordant with this, transcriptional
upregulation of the unfolded protein response (UPR) has been reported in patients
with CALR-mutated MPN. However, it is not understood how CALR-mutated cells counter-balance
ER stress resulting from impaired chaperone function. Despite the frequency of CALR
mutations in MPN, there are currently no treatment strategies to preferentially target
CALR-mutant cells over healthy cells.
Aims: To determine the mechanisms by which mutant CALR cells alleviate ER stress and
to exploit these mechanisms selectively by pharmacological intervention in vivo.
Methods: Long-term HSCs (LT-HSCs) from Calr
Δ52 knockin mice were isolated for RNA-Seq. Calr
Δ52 bone marrow (BM) was differentiated ex vivo, subsequently enriched for megakaryocytes,
and subjected to quantitative proteomic analysis. Pathway analyses of CALR-mutant
megakaryocyte progenitors (MkPs) were performed on Genotyping of Transcriptome (GoT)
data from patients with Essential Thrombocytosis (ET). Calr
Δ52 VAVCre mice and chimeric transplanted Calr
Δ52 mice were treated with a proteasome inhibitor, an IRE1a inhibitor, or both.
Results: To identify dysregulated pathways in Calr
Δ52/Δ52 LT-HSCs, we performed RNA-seq and found that Xbp1s downstream of IRE1a and
the proteasome pathway was significantly upregulated in Calr
Δ52/Δ52
animals. We confirmed elevated XBP1s protein level in heterozygous Calr
Δ52
knockin mice by flow cytometry. Since elevated and abnormal megakaryopoiesis are hallmarks
of CALR-mutant MPN, we performed quantitative proteomics on megakaryocyte-enriched
cells from Calr
Δ52
mice. We confirmed upregulation of both proteasome and ER chaperone proteins in Calr-mutant
cells. To verify these findings in human MPN, we interrogated a published GoT data
set and found that in addition to XBP1s, the proteasome pathway was also differentially
upregulated in CALR-mutated patient MkPs compared to the wildtype MkPs (p<2e-16).
To investigate the functional consequences of IRE1a or proteasome inhibition, we treated
CALR-mutant hematopoietic cell lines with KIRA6 (IRE1a inhibitor) and/or bortezomib
(proteasome inhibitor) and found CALR-mutant cells to be differentially sensitive
as compared to isogenic controls. We subsequently found that treatment with bortezomib
leads to the accumulation of misfolded and ubiquitinylated proteins in mutant Calr
BM, resulting in pro-apoptotic priming. In vivo treatment of Calr
Δ52/+ mice with bortezomib resulted in a significant reduction in platelets. Finally,
we combined IRE1a and proteasome inhibition in a chimeric BM transplantation in vivo
model using CD45.2 Calr
Δ52/+ MxCre GFP and CD45.1 wild-type competitor cells. Following combination therapy,
we found a significant reduction in Calr-mutant donor chimerism, specifically in LT-HSCs
and platelets, accompanied by reductions in LT-HSC frequency and platelet count.
Summary/Conclusion: In summary, we have found that disrupted proteasis in CALR-mutated
MPN cells results in a dependency on the IRE1-XPB1 axis of the UPR and proteasome
activity. Using pre-clinical MPN models, we found that combined inhibition of both
pathways in vivo normalizes important features of MPN and preferentially targets Calr
Δ52 over wild-type cells. These findings highlight combined inhibition of IRE1 and
the proteasome as a promising therapeutic strategy in CALR-mutant MPN.
S192: SINGLE-CELL RNA PROFILING OF MYELOFIBROSIS PATIENTS REVEALS PELABRESIB-INDUCED
DECREASE OF MEGAKARYOCYTIC PROGENITORS AND NORMALIZATION OF CD4+ T CELLS IN PERIPHERAL
BLOOD
O. Zavidij1,*, N. J. Haradhvala2, R. Meyer1, J. Cui1, S. Verstovsek3, S. Oh4, A. Mead5,
P. Taverna1
1Constellation Pharmaceuticals a MorphoSys Company; 2Harvard Graduate Program in Biophysics,
Harvard University, Boston, MA; 3Leukemia Department, University of Texas MD Anderson
Cancer Center, Houston, TX; 4Hematology Division, Washington University, St Louis,
MO; 5NIHR Biomedical Research Centre, University of Oxford, Oxford, United States
of America
Background: Abnormal differentiation of the megakaryocytic (MK) lineage in conjunction
with overproduction of proinflammatory cytokines (Ck), resulting in bone marrow fibrosis,
anemia, extramedullary hematopoiesis and often hepatosplenomegaly, are the main biological
and clinical characteristics of myelofibrosis (MF). Bromodomain and extraterminal
domain (BET) proteins play a key role in the regulation of neoplastic myeloproliferation
and proinflammatory Ck production. Pelabresib (CPI-0610) is a potent, selective BET
inhibitor under investigation in MF patients (pts) as monotherapy and in combination
with the Janus kinase inhibitor (JAKi) ruxolitinib (RUX) in the ongoing MANIFEST Phase
2 study (NCT02158858). Arm 1: pelabresib as monotherapy in RUX-intolerant, ineligible
or refractory MF pts; Arm 2: pelabresib in combination with RUX as an ‘add-on’ in
MF pts with suboptimal response to RUX; Arm 3: pelabresib combination therapy with
RUX in JAKi-naïve MF pts. Pelabresib as monotherapy and in combination with RUX improved
spleen volume reduction, symptoms and hemoglobin levels (Talpaz M, ASH 2020). The
effects of pelabresib on hematopoietic stem/progenitor cells (HSPCs) and immune cell
populations in MF pts are presented here.
Aims: We aimed to characterize the cellular composition and transcriptional alterations
occurring in peripheral blood (PB) obtained from MF pts enrolled in the MANIFEST trial.
Methods: We performed single-cell RNA sequencing on 234,904 CD34+ HSPCs and 135,970
CD34- mature PB cells (Figure). Cells were obtained from a random pool of 20 pts (Arm
1 n=5, Arm 2 n=8, Arm 3 n=7), which included a baseline sample (BL) and samples collected
during treatment (range: C3 through C12) for each pt. Mobilized PB cells from healthy
donors (HD) were used as a control (n=11).
Results: Analysis of CD34+ HSPCs at BL demonstrated a statistically significant increase
of MK, neutrophilic and erythroid progenitors in MF pts compared with HDs and decreased
numbers of myeloid and B cell lineage progenitors. Pelabresib as monotherapy and in
combination with RUX led to a significant reduction of MK-, neutrophilic and erythroid
progenitors as compared with BL in all pts analyzed.
Analysis of CD34- cells from MF pts identified a significantly lower proportion of
CD4+ T cells and increased numbers of erythroid cells at BL compared with HD. Individual
pts also exhibited reduction in natural killer cells and CD16+ monocytes as well as
elevated MK lineage cells. Pelabresib as monotherapy and in combination with RUX increased
the proportion of CD4+ T cells, and more importantly, reduced MK lineage cells compared
with BL in both treatment-naïve and RUX relapsed/refractory (r/r) pts.
In MF pts at BL, a larger spleen volume was observed in pts with lower numbers of
CD4+ T cells and increased numbers of MK- and myeloid CSF3R+ cells.
Updated results of the cell composition analysis and transcriptional reprogramming
during pelabresib treatment, as well as correlation with effects on cytokine plasma
levels and clinical efficacy data, will be presented.
Image:
Summary/Conclusion: The single-cell profiling of a subset of MF pts enrolled in the
MANIFEST study suggests that pelabresib alone and in combination with RUX induces
an improvement of the myeloid-lymphoid imbalance in both JAKi-naïve and r/r MF pts.
The observed effects of pelabresib on HSPCs from MF pts confirm the ex vivo activity
on erythroid and MK differentiation of stem cells (Keller P, EHA 2021; Verstovsek
S, ASH 2021) and implies a potential disease-modifying effect warranting further investigation.
S193: GENOMIC AND FUNCTIONAL IMPACT OF TP53 INACTIVATION IN JAK2V617F MYELOPROLIFERATIVE
NEOPLASMS: A TRANSGENIC MOUSE MODEL APPROACH.
P. GOU1, D. LIU1,*, E. Lauret2, N. Maslah3, V. Montcuquet4, V. meignin5, J.-J. Kiladjian6,
B. Cassinat3, S. Giraudier3
1U1131, 25Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR 8104, INSERM;
3U1131, INSERM Univrsité de Paris and APHP; 4Animal Facility Unit, Université de Paris;
5Histo-pathological department, Hôpital Saint Louis; 6Centre Investigations Cliniques,
Hôpital Saint-Louis, Paris, France, INSERM Univrsité de Paris and APHP, Paris, France
Background: MPN are characterized by increased proliferation of myeloid cells and
a risk of transformation to AML. MPN are due to the acquisition of mutations (JAK2,
CALR, MPL) leading to JAK2 pathway activation but cooperating mutations involved in
MPN evolution have been described. Mutations of TP53 in JAK2V617F patients are linked
to AML transformation, illustrated by development of leukemias after retroviral overexpression
of JAK2V617F in TP53 inactivated (KO) cells transplanted in irradiated animals, thus
a possible cooperation between TP53 inactivation and JAK2 signaling in transformation
process has been postulated. However, JAK2V617F signaling has also been reported as
able to reduce p53 expression per sesuggesting a redundant role of p53 inactivation
in JAK2V617F cells.
Aims: Determine the role of TP53 in JAK2V617F induced genetic and phenotypic modifications.
Methods: To better understand the role of TP53 in MPN phenotype and evolution in steady
state hematopoiesis, we developed a transgenic model of vav-cre induced JAK2V617F
expression in TP53 knock-out (KO) animals and analyzed phenotype, survival, genetic
modifications using stem cell compartments RNA-Seq and response to therapy of this
triple transgenic mice.
Results: JAK2V617F vav-inductible-Tg/TP53 KO mice were generated to recombine JAK2V617F
at an endogenous level in TP53 inactivated hematopoietic cells.
During follow-up, JAK2V617F/TP53KO mice developed the same phenotype than JAK2V617F
transgenic mice without leukemia. In competitive repopulations with grafts with different
ratio of JAK2V617F, JAK2V617F/TP53KO and wild type cells, we observed that JAK2V617F/TP53KO
cells outcompete JAK2V617F cells.
In order to better define TP53-dependent and independent pathways linked to these
phenotypes, LT-HSC, ST-HSC, MPP, CMP, MEP and GMP from normal, JAK2V617F and JAK2V617F/TP53
KO mice were cell sorted and RNA-Seq analysis was performed. Principal component analysis
demonstrated that JAK2V617F/TP53KO cells were closer to normal cells than JAK2V617F
cells whatever the (stem) cell compartment, illustrating that JAK2V617F-induced modifications
are largely p53 dependent. Further analysis demonstrated that approximatively half
of JAK2V617F deregulated genes in the different compartments are TP53 dependent including
the IFN pathways. In order to validate this finding, mice repopulated with a mix of
WT and JAK2V617F (either p53 KO or wild-type) cells were treated for 8 weeks with
recombinant murine pegylated IFN-a. JAK2V617F reconstituted animals entered in complete
hematological remission while JAK2VF/TP53KO reconstituted animals did not, illustrating
that loss of TP53 in this context induced resistance to IFN-a.
On the other hand, since MPN develop in the same way in JAK2V617F-only and in JAK2V617F/TP53KO
mouse, this suggests that JAK2V617F-specific pathways also found differentially expressed
in JAK2V617F/TP53KO are linked to the MPN phenotype. KEGG and GO analysis demonstrated
that these genes were mainly implicated in cytokine response, cell proliferation,
differentiation, and leukemia evolution illustrating that the development of MPN and
its possible risk of transformation in this mouse model is largely TP53 independent.
Summary/Conclusion: Taken together, our results show that a large part of genetic
modifications induced by JAK2V617F mutation are p53 dependent but MPN phenotype is
not, TP53 loss is insufficient to induce quick leukemic transformation in steady-state
hematopoiesis in Jak2 V617F MPN despite it increases LT-HSC cell proliferation and
finally that TP53 loss could be involved in IFN resistance in MPN.
S194: INTEGRATIVE CLINICAL PROTEOTYPING AND DRUG RESPONSE PROFILING IDENTIFIES TARGETABLE
BIOLOGY UNDERLYING MYELOPROLIFERATIVE NEOPLASMS
M. H. Wildschut1 2 3,*, M. van Oostrum2, J. Settelmeier2 4, J. Mena1, C. Dördelmann5,
Y. Festl1, A. Ring3, R. C. Skoda6, M. Lopes5, B. Wollscheid2 4, B. Snijder1, A. Theocharides3
1Institute of Molecular Systems Biology, Department of Biology; 2Institute of Translational
Medicine, Department of Health Science and Technology, ETH Zurich; 3Department of
Medical Oncology and Hematology, Division of Hematology, University Hospital Zurich,
Zurich; 4Swiss Institute of Bioinformatics, Lausanne; 5Insititute of Molecular Cancer
Research, University of Zurich, Zurich; 6Department of Biomedicine, Experimental Hematology,
University Hospital Basel and University of Basel, Basel, Switzerland
Background: Myeloproliferative neoplasms (MPN) are a family of hematopoietic stem
cell diseases characterized by frequent driver mutations in Calreticulin (CALR) and
JAK2. While patients with essential thrombocythemia (ET) have a more favorable outcome,
the prognosis of patients with advanced myelofibrosis (MF) is particularly poor with
limited therapeutic options.
Aims: Here, we set out to elucidate biology underlying MPN and translate this into
vulnerabilities that can be exploited by targeted therapies. By integration of proteomic
analyses and drug response profiling we aimed to correlate MPN protein alterations
directly to targeted drug responses, and subsequently validate pathway alterations
using dedicated follow-up experiments.
Methods: We gathered extended MPN patient and matched healthy donor (HD) cohorts (n
= 119) and a follow-up MF-specific cohort (n = 44) to investigate both general and
subgroup-specific alterations. We performed proteomics on isolated granulocytes, hematopoietic
stem and progenitor cells (HSPCs), and T-cells, and drug screening on peripheral blood
mononuclear cells (PBMCs). For follow-up experiments, we established a CALR mutant-specific
antibody for use in high-content immunofluorescence imaging.
Results: We identify a specific signature of disease- and mutation-specific protein
alterations present in MPN patients by machine learning. Among the identified alterations,
we find strong upregulation of proteins belonging to the MCM complex, a helicase involved
in DNA replication, to be specifically present in a subgroup of MF patients that are
clinically characterized by a worse prognosis. In these patients, a proteomic signature
related to DNA replication and cell cycling is found to be elevated across blood cell
types. We proof that cells from these patients are a) characterized by high proliferation
rates, and b) respond well to drugs targeting this process (e.g. topoisomerase inhibitors).
Furthermore, we find upregulation of proteins involved in endoplasmic reticulum (ER)
stress to be significantly correlated to the CALR mutation burden across HSPCs, granulocytes,
and T-cells. Using an advanced imaging approach we show that on a single cell level,
the amount of ER stress corresponds to the level of CALR mutant protein expression.
This results in a higher sensitivity of CALR-mutated cells to drugs targeting ER stress
or the corresponding unfolded protein response (UPR).
Summary/Conclusion: Using large-scale MPN patient cohorts, we are able to elucidate
biology underlying MPN disease. We here show specific proteomic signatures of proliferation
and ER stress to be present in subsets of MPN patients and translate this to specific
biological alterations and correlated therapeutic vulnerabilities. Our findings can
be instrumental to both improved stratification of response and discovery of novel
targeted therapeutics for MPN patients.
S195: MOMENTUM: PHASE 3 RANDOMIZED STUDY OF MOMELOTINIB (MMB) VERSUS DANAZOL (DAN)
IN SYMPTOMATIC AND ANEMIC MYELOFIBROSIS (MF) PATIENTS PREVIOUSLY TREATED WITH A JAK
INHIBITOR
S. Verstovsek1,*, A. Vannucchi2, A. Gerds3, H. K. Al-Ali4, D. Lavie5, A. Kuykendall6,
S. Grosicki7, A. Iurlo8, Y. T. Goh9, M. Lazaroiu10, M. Egyed11, M. L. Fox12, D. McLornan13,
A. Perkins14, S.-S. Yoon15, V. Gupta16, J.-J. Kiladjian17, R. Donahue18, J. Kawashima18,
R. Mesa19
1The University of Texas MD Anderson Cancer Center, Houston, TX, United States of
America; 2Center Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi,
University of Florence, Florence, Italy; 3Cleveland Clinic Department of Hematology
and Medical Oncology, Avon, OH, United States of America; 4University Hospital of
Halle, Halle, Germany; 5Hadassah Hebrew University Medical Center, Jerusalem, Israel;
6Moffitt Cancer Center, Tampa, FL, United States of America; 7Medical University of
Silesia, Katowice, Poland; 8Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico,
Milan, Italy; 9Singapore General Hospital, Singapore, Singapore; 10Policlinica de
Diagnostic Rapid Brasov, Brasov, Romania; 11Somogy County Mór Kaposi General Hospital,
Kaposvár, Hungary; 12Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Hospital
Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain; 13Guy’s and
Saint Thomas’ NHS Foundation Trust, London, United Kingdom; 14Monash University, Melbourne,
Australia; 15Seoul National University Hospital, Seoul, South Korea; 16Princess Margaret
Cancer Centre, Toronto, ON, Canada; 17Saint-Louis Hospital (AP-HP), Paris, France;
18Sierra Oncology, Inc., San Mateo, CA; 19UT Health San Antonio Cancer Center, San
Antonio, TX, United States of America
Background: MMB, a novel oral ACVR1/ALK2 and JAK1/2 inhibitor, showed clinical activity
on MF symptoms, red blood cell (RBC) transfusion requirements (anemia), and spleen
volume in the SIMPLIFY trials.
Aims: This pivotal phase 3 study of MF patients (pts) previously treated with a JAK
inhibitor (JAKi) tested MMB vs DAN on key symptom, anemia, and spleen volume endpoints
at 24 weeks (wks).
Methods: Eligibility: Primary or post-ET/PV MF; DIPSS high risk, Int-2, or Int-1;
MF Symptom Assessment Form Total Symptom Score (MFSAF TSS) ≥10; hemoglobin (Hgb) <10 g/dL;
prior JAKi for ≥90 days, or ≥28 days if RBC transfusions ≥4 units in 8 wks or Gr 3/4
thrombocytopenia, anemia, or hematoma; palpable spleen ≥5 cm. Stratification: TSS
(≥22 vs <22), palpable spleen (≥12 cm vs <12 cm), and RBC units transfused (0, 1-4,
and 5+). JAKi taper and washout was ≥21 days. Randomization: 2:1 to MMB 200 mg QD
plus DAN placebo or DAN 600 mg QD plus MMB placebo for 24 wks, after which pts could
receive open-label MMB. Assessments: Pt reported symptoms using a daily eDiary and
spleen volume by MRI or CT. The primary endpoint was TSS response (≥50% reduction
from baseline [BL]) rate at wk 24. Secondary endpoints, assessed sequentially at wk
24, were RBC transfusion independence (TI) rate, splenic response rate (SRR; ≥25%
reduction in volume from BL), change from BL in TSS, SRR (≥35% reduction from BL)
and rate of zero transfusions since BL. Informed consent was obtained from all participants.
Results: 94 of 130 (72%) MMB pts and 38 of 65 (58%) DAN pts completed the 24-wk randomized
treatment (RT) phase. Median BL TSS were 28 (MMB) and 26 (DAN), Hgb were 8.1 (MMB)
and 7.9 (DAN) g/dL, and platelets were 97 (MMB) and 94 (DAN) x109/L. BL TI was 13%
(MMB) and 15% (DAN). BL mean spleen volume was 2367 (MMB) and 2288 (DAN) cm3. Prior
JAKi was ruxolitinib in 195 pts (100%) and fedratinib in 9 pts (5%); mean duration
of prior JAKi was 134 weeks. All primary and key secondary endpoints were met (Table).
Most common Gr ≥3 treatment-emergent adverse events (TEAEs) in the RT phase of the
study were thrombocytopenia (MMB, 22%; DAN, 12%) and anemia (MMB, 8%; DAN, 11%). Gr
≥3 infections occurred in 15% of MMB and 17% of DAN pts. Peripheral neuropathy (PN)
occurred in 5 (4%) of MMB (all Gr ≤2) and 1 (2%) of DAN (Gr ≤2) pts in the RT phase,
and none discontinued study drug due to PN. Overall, TEAEs led to study drug discontinuation
in 18% of MMB and 23% of DAN pts, and serious TEAEs were reported in 35% of MMB and
40% of DAN pts, in RT phase. A trend toward improved OS up to wk 24 was seen with
MMB vs DAN (HR=0.506, p=0.0719).
Image:
Summary/Conclusion: In symptomatic and anemic MF pts, MMB was superior to DAN for
symptom responses, transfusion requirements, and spleen responses with comparable
safety and favorable survival. MMB may address a critical unmet need, particularly
in MF pts with anemia. NCT04173494.
S196: ROPEGINTERFERON ALFA-2B ACHIEVES PATIENT-SPECIFIC TREATMENT GOALS IN POLYCYTHEMIA
VERA: FINAL RESULTS FROM THE PROUD-PV/CONTINUATION-PV STUDIES
H. Gisslinger1,*, C. Klade2, P. Georgiev3, D. Krochmalczyk4, L. Gercheva-Kyuchukova,
M. Egyed6, P. Dulicek7, A. Illes8, H. Pylypenko9, L. Sivcheva10, J. Mayer11, V. Yablokova12,
V. Empson2, K. Krejcy2, H. Hasselbalch13, R. Kralovics14, J.-J. Kiladjian15
1Department of Internal Medicine I, Division of Hematology and Blood Coagulation,
Medical University Vienna; 2AOP Orphan Pharmaceuticals GmbH, Vienna, Austria; 3Medical
University of Plovdiv, Plovdiv, Bulgaria; 4Teaching Unit of the Hematology Department,
University Hospital in Krakow, Krakow, Poland; 5Clinical Hematology Clinic, Multiprofile
Hospital for Active Treatment “Sveta Marina”, Varna, Bulgaria; 6Department of Internal
Medicine II, Kaposi Mor County Teaching Hospital, Kaposvar, Hungary; 7Department of
Clinical Hematology, University Hospital Hradec Kralove, Hradec Kralove, Czechia;
8Department of Hematology, Faculty of Medicine, University of Debrecen, Debrecen,
Hungary; 9Department of Hematology, Regional Treatment and Diagnostics Hematology
Centre, Cherkasy Regional Oncology Centre, Cherkasy, Ukraine; 10First Department of
Internal Medicine, Multiprofile Hospital for Active Treatment - HristoBotev, Vratsa,
Bulgaria; 11Clinic of Internal Medicine - Hematology and Oncology, University Hospital
Brno, Brno, Czechia; 12Department of Hematology, Yaroslavl Regional Clinical Hospital,
Yaroslavl, Russia; 13Department of Hematology, Zealand University Hospital, University
of Copenhagen, Roskilde, Denmark; 14Department of Laboratory Medicine, Medical University
of Vienna, Vienna, Austria; 15Centre d’Investigations Cliniques, INSERM, CIC1427,
Université de Paris, AP-HP, Hôpital Saint-Louis, Paris, France
Background: Treatment of polycythemia vera (PV) aims to prevent thromboembolic complications,
reduce the risk of progression to acute leukemia or myelofibrosis - of particular
concern to patients - and ameliorate the symptom burden; specifically, to improve
quality of life, therapy should address the most clinically important symptoms while
reducing phlebotomies to avoid iron-deficiency symptoms. Long-term efficacy and safety
of ropeginterferon alfa-2b have been demonstrated in PROUD-PV/CONTINUATION-PV; the
final analysis applied a patient-focused approach.
Aims: To analyze the patient-relevant benefit of ropeginterferon alfa-2b versus hydroxyurea
(HU)/best available treatment (BAT) over 6 years.
Methods: Patients diagnosed with PV according to WHO 2008 criteria who were cytoreduction-naïve
or hydroxyurea pre-treated and gave written informed consent were randomized 1:1 to
ropeginterferon alpha-2b or control treatment (HU) for one year in PROUD-PV. In CONTINUATION-PV,
control arm patients could switch from HU to BAT. Patient-reported PV symptom burden
was assessed based on adverse events documented in the patient diary and recorded
at each visit; items defined in the Myeloproliferative Neoplasm Symptom Assessment
Form Total Symptom Score (MPN-SAF TSS; fatigue, concentration problems, early satiety,
inactivity, night sweats, itching, bone pain, abdominal discomfort, weight loss, and
fevers) and medical synonyms were evaluated post-hoc. Efficacy assessments included
Kaplan-Meier analysis of event-free survival, phlebotomy need and JAK2V617F allele
burden. Analyses were conducted on the CONTINUATION-PV full analysis set over 6 years
of treatment.
Results: The full analysis set comprised 95 patients in the ropeginterferon alfa-2b
arm and 74 in the control arm.
Patient-reported symptoms defined in the MPN-SAF TSS were present in a small minority
(9.5%) of patients per arm at baseline (up to Week 4 of treatment) in this early-stage
PV population. Occurrence of the defined symptoms remained low over long-term treatment,
reported in 15.7% of patients in the ropeginterferon alfa-2b arm and 20.7% in the
control arm during the 6th year of treatment.
No phlebotomies were required to maintain hematocrit <45% in the 6th year of treatment
in 81.4% of patients receiving ropeginterferon alfa-2b compared with 60.0% of patients
in the control arm (p=0.005).
Depletion of the JAK2V617F alle burden, which may lower the risk of progression to
myelofibrosis, was observed in ropeginterferon alfa-2b treated patients; JAK2V617F
allele burden <1% at 6 years was achieved in 19/92 (20.7%) patients in the ropeginterferon
alfa-2b arm with baseline allele burden >10%. One patient met this threshold in the
control arm (1/70 [1.4%]; p=0.0001).
Event-free survival (risk events: disease progression, death and thromboembolic events)
over ≥6 years of treatment was significantly higher among ropeginterferon alfa-2b
treated patients than the control group (risk events reported in 5/95 vs. 12/74 patients,
respectively; p=0.04 [Log-Rank]; Fig 1).
Image:
Summary/Conclusion: Long-term ropeginterferon alfa-2b therapy fulfils treatment goals
important to patients with PV: a good quality of life as indicated by a low symptom
burden and phlebotomy requirement, the potential to influence myelofibrosis risk,
and better event-free survival versus BAT.
S197: NAVITOCLAX PLUS RUXOLITINIB IN JAK INHIBITOR-NAÏVE PATIENTS WITH MYELOFIBROSIS:
PRELIMINARY SAFETY AND EFFICACY IN A MULTICENTER, OPEN-LABEL PHASE 2 STUDY
F. Passamonti1,*, J. Foran2, A. Tandra3, V. De Stefano4 5, M. Laura Fox6, A. Mattour7,
M. F. McMullin8, A. C. Perkins9, G. Rodriguez-Macías10, H. Sibai11, Q. Qin12, J. Potluri12,
J. How13
1Department of Medicine and Surgery, University of Insubria, Varese, Italy; 2Mayo
Clinic, Jacksonville, FL; 3Indiana Blood and Marrow Transplant, Indianapolis, United
States of America; 4Section of Hematology, Department of Radiological and Hematological
Sciences, Catholic University; 5Fondazione Policlinico Universitario Agostino Gemelli
IRCCS, Rome, Italy; 6Department of Hematology, Hospital Universitari Vall d’Hebron,
Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron
Hospital Campus, Barcelona, Spain; 7Henry Ford Hospital, Detroit, MI, United States
of America; 8Centre for Medical Education, Queenʼs University Belfast, Belfast, United
Kingdom; 9Australian Centre for Blood Diseases, Monash University and The Alfred Hospital,
Melbourne, Australia; 10Department of Hematology, Hospital General Universitario Gregorio
Marañón, Madrid, Spain; 11Medical Oncology and Hematology, Princess Margaret Cancer
Centre, University of Toronto, Toronto, Canada; 12AbbVie Inc, North Chicago, IL, United
States of America; 13Division of Hematology, McGill University Health Center, Montreal,
Canada
Background: Ruxolitinib, a Janus kinase (JAK) 1/2 inhibitor, is the current standard
of care for patients with myelofibrosis (MF) that improves disease symptoms in approximately
40% of patients with limited impact on disease biology. Many patients lose response
over time, highlighting an unmet need for novel therapies. Navitoclax is a first-in-class,
oral, small molecule that binds with high affinity to BCL-XL and BCL-2, key pro-survival
proteins in the apoptotic pathway, and has a synergistic effect when used in combination
with JAK inhibitors to enhance malignant cell death in MF. This ongoing, open-label,
multicenter, phase II trial (NCT03222609) is evaluating the efficacy and safety of
navitoclax with or without ruxolitinib in patients with MF and previously published
data suggest clinically meaningful activity of the combination in patients with prior
ruxolitinib failure (Pemmaraju et al., ASH 2020). Here, we report results from patients
who were JAK inhibitor-naïve and treated with navitoclax and ruxolitinib combination
therapy.
Aims: N/A
Methods: Enrolled patients had primary or secondary MF with splenomegaly (DIPSS ≥
Intermediate-1) and did not receive prior therapy with JAK-2 or bromodomain and extraterminal
motif (BET) inhibitors. Patients initiated navitoclax at 100 mg once per day (QD)
or 200 mg QD if baseline platelet count was ≤150 × 109/L or >150 × 109/L, respectively.
Ruxolitinib was orally administered twice daily at a starting dose based on baseline
platelet count as per the local ruxolitinib label. The primary endpoint was spleen
volume reduction of ≥35% (SVR35) from baseline at week 24. SVR35 was assessed at weeks
12, 24, and once every 24 weeks thereafter. Key secondary endpoints were ≥50% reduction
in total symptom score (TSS50), bone marrow fibrosis reduction, and anemia response
(per International Working Group criteria). Adverse events (AEs) were monitored throughout
the study.
Results: As of Oct 04, 2021, 32 patients received navitoclax plus ruxolitinib. The
median duration of follow-up was 6.1 (range, 1.9 to 18.6) months. Twenty-eight (88%)
patients received a navitoclax dose of 200 mg, and 4 (13%) received 100 mg OD. The
median age was 69 years (range, 44 to 83), median spleen volume was 1889.08 cm3 (range,
645.6 to 7339.6), and 38% had at least 1 prior line of therapy.
The median navitoclax and ruxolitinib exposures were 24.1 (range, 5.1 to 80.9) and
24.1 (5.1 to 80.9) weeks, respectively. Thirty-one (97%) patients experienced ≥1 AE,
25 (78%) had Grade ≥3 AEs, and 6 (19%) had serious AEs. The most frequent Grade ≥3
AEs were anemia (34%), thrombocytopenia (31%), and neutropenia (19%). Three (9%) and
2 (6%) patients experienced an AE that led to navitoclax and ruxolitinib discontinuation,
respectively, and 2 (6%; 1 progressive disease, 1 cardiac disorder unrelated to navitoclax)
AEs led to death ≤30 days after the last dose of navitoclax. SVR35 was achieved by
52% of evaluable patients at week 24 (SVR35 in DIPSS Intermediate-2, 50%; High-risk,
33%) and by 76% at any time on the study (Table). The median time to first SVR35 was
12.1 (range, 11 to 47) weeks.
Image:
Summary/Conclusion: The combination of navitoclax and ruxolitinib was well tolerated
and demonstrated early and robust reductions in spleen volume, anemia, and BM fibrosis
in patients without prior JAK-2 inhibitor exposure. SVR35, anemia, and bone marrow
fibrosis improved over time.
S198: BET INHIBITOR PELABRESIB (CPI-0610) COMBINED WITH RUXOLITINIB IN PATIENTS WITH
MYELOFIBROSIS — JAK INHIBITOR-NAÏVE OR WITH SUBOPTIMAL RESPONSE TO RUXOLITINIB — PRELIMINARY
DATA FROM THE MANIFEST STUDY
J. Mascarenhas1,*, M. Kremyanskaya1, A. Patriarca2, C. Harrison3, P. Bose4, R. K.
Rampal5, F Palandri6, T. Devos7, F. Passamonti8, G. Hobbs9, M. Talpaz10, A. Vannucchi11,
J.-J. Kiladjian12, S. Verstovsek13, R. Hoffman1, M. E. Salama13, D. Chen14, P. Taverna15,
A. Chang15, G. Colak15, S. Klein15, V. Gupta16
1Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United
States of America; 2Hematology Unit, Department of Translational Medicine, University
of Eastern Piedmont and AOU Maggiore della Carità, Novara, Italy; 3Guy’s and St Thomas’
NHS Foundation Trust, London, United Kingdom; 4MD Anderson Cancer Center, Houston,
TX; 5Memorial Sloan-Kettering Cancer Center, New York, NY, United States of America;
6IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”,
Bologna, Italy; 7University Hospitals Leuven and Laboratory of Molecular Immunology
(Rega Institute), KU Leuven, Leuven, Belgium; 8University of Insubria, Varese, Italy;
9Division of Hematology/Oncology, Massachusetts General Hospital, Harvard Medical
School, Boston, MA; 10University of Michigan Comprehensive Cancer Center, Ann Arbor,
MI, United States of America; 11Azienda Ospedaliero-Universitaria Careggi, University
of Florence, Florence, Italy; 12Hôpital Saint-Louis, Université de Paris, Paris, France;
13Leukemia Department, University of Texas MD Anderson Cancer Center, Houston, TX;
14Mayo Clinic, Rochester, MN; 15Constellation Pharmaceuticals a MorphoSys Company,
Boston, MA, United States of America; 16Princess Margaret Cancer Centre, University
of Toronto, Toronto, Canada
Background: Myelofibrosis (MF) is characterized by bone marrow (BM) fibrosis, anemia,
splenomegaly and constitutional symptoms. Progressive BM fibrosis results from aberrant
megakaryopoiesis and proinflammatory cytokine expression, two processes that are tightly
regulated by bromodomain and extraterminal domain (BET) protein-mediated expression
of genes (e.g. NF-κB, MYC and BCL-2) and often lead to myeloproliferation and cytopenias.
While many MF patients (pts) receive the Janus kinase inhibitor (JAKi) ruxolitinib
(RUX) as the current standard of care, the depth and durability of responses, along
with the percentage of pts that achieve ≥35% spleen volume reduction from baseline
(BL; SVR35) and ≥50% total symptom score reduction from BL (TSS50) are limited; thus,
a significant unmet medical need exists. Pelabresib (CPI-0610) is an oral, small-molecule,
investigational BET inhibitor that downregulates NF-κB gene expression and other relevant
genes involved in MF disease pathways.
Aims: Data (Sep 2021 data cut) on the safety and efficacy of pelabresib in combination
with RUX in pts with MF from Arms 2 and 3 of the ongoing, open-label Phase 2 MANIFEST
study (NCT02158858). In Arm 2, MF pts with suboptimal response to RUX are treated
with pelabresib as ‘add-on’ to RUX (Arm 2A: transfusion dependent [TD]; Arm 2B: non-TD).
In Arm 3, JAKi-naïve MF pts are treated with pelabresib in combination with RUX.
Methods: The primary endpoints are SVR35 at Week (Wk) 24 for Arm 3 and Arm 2B (non-TD
cohort) and TD to transfusion independence (TI) in Arm 2A (TD cohort). The key secondary
endpoint is TSS50 per Myelofibrosis Symptom Assessment Form v4.0 at Wk 24; in Arm
2A (TD cohort), SVR35 is an additional key secondary endpoint. BM biopsies to assess
BM fibrosis and safety data are also evaluated.
Results: At Wk 24 in Arm 3 (N=84), 68% (57/84) pts achieved SVR35 (median change:
–50%; Figure), and 56% (46/82) pts achieved TSS50 (median change: –59%). Twenty-four
percent of pts had a mean hemoglobin increase ≥1.5 g/dL from BL over 12 weeks without
transfusions. At Wk 24 in Arm 2 (N=86), 20% (16/81) pts achieved SVR35 (median change:
–18%; Figure), and 37% (30/81) pts achieved TSS50 (median change: –47%). In Arm 2A,
the TD to TI rate was 16% (6/38).
BM fibrosis improvement by ≥1 grade was achieved in 31% (16/52) and 25% (9/36) pts
in Arm 3 and 2, respectively. Further central review and exploratory analyses of BM
pathology from a more mature data set, including clinical correlations, will be presented.
The most common hematologic treatment-emergent adverse event (TEAE) of any grade was
thrombocytopenia, reported in 52% (≥Grade 3: 12%) and 52% (≥Grade 3: 33%) pts in Arm
3 and 2, respectively. Anemia was reported in 42% (≥Grade 3: 35%) and 27% (≥Grade
3: 19%) pts in Arm 3 and 2, respectively. Low-grade gastrointestinal TEAEs and respiratory
infections in Arm 3 and 2 were observed but rarely a reason for treatment discontinuation.
Image:
Summary/Conclusion: Based on these interim Phase 2 data, pelabresib in combination
with RUX, in both RUX treatment-naïve and -experienced pts with MF, resulted in splenic
and symptom responses and BM fibrosis improvement, and appeared to be well tolerated.
Based on data from MANIFEST Arm 3, the randomized, double-blind, active-control Phase
3 MANIFEST-2 study was initiated to further evaluate the safety and efficacy of pelabresib
in combination with ruxolitinib in JAKi treatment-naïve pts with MF (NCT04603495).
S199: A NATIONAL RETROSPECTIVE COHORT STUDY OF MPN-SVT: RESULTS FROM THE UK MYELOPROLIFERATIVE
NEOPLASMS ASSOCIATED SPLANCHNIC VEIN THROMBOSIS (MASCOT) REGISTRY
R. Hargreaves1,*, M. Subhan2, S. Alimam1 2, S. Shapiro3 4, A. Robinson5, M. Carter5,
A. Godfrey5, M. Ul-Haq6, M. Jain6, G. Greenfield7, A. McGregor8, H. K. Hussein9, D.
Tripathi10, H. Network11, A. Doyle12, K. White12, N. Curto-Garcia12, D. Patch13, M.
Sekhar1 2
1Haematology, Royal Free Hospital NHS Foundation Trust; 2Haematology, University College
London Hospitals NHS Foundation Trust, London; 3Haematology, Oxford University Hospitals
NHS Foundation Trust, NIHR Biomedical Research Centre; 4Radcliffe Department of Medicine,
University of Oxford, Oxford; 5Haematology, Cambridge University Hospitals NHS Foundation
Trust, Cambridge; 6Haematology, Leeds Teaching Hospitals NHS Trust, Leeds; 7Patrick
G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast; 8Haematology,
Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne; 9Haematology;
10Hepatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham; 11Haematology
Specialty Trainee Audit and Research, HaemSTAR Network, Nationwide; 12Haematology,
Guy’s and St Thomas’ NHS Foundation Trust; 13Hepatology, Royal Free Hospital NHS Foundation
Trust, London, United Kingdom
Background: Patients with myeloproliferative neoplasms (MPN) are at increased risk
of splanchnic vein thrombosis (SVT), defined as abdominal thrombosis in portal, splenic,
mesenteric or hepatic veins. MPN-SVT carries a significant mortality and morbidity
burden and poses clinical management challenges. Detailed studies of this patient
population are lacking.
Aims: To establish a national web-based clinical registry for MPN-SVT (MASCOT Registry)
and use this to investigate demographics, clinical features, co-morbidities, outcome
& UK treatment practices for MPN-SVT.
Methods: The MASCOT Registry is a UK-wide retrospective cohort of patients with MPN-SVT
from 9 large haematology/hepatology centres. Participating centres entered pseudo-anonymised
data for patients with MPN-SVT. New and historical cases were added; demographic,
radiologic, clinical and outcome data were collected.
Results: 232 patients with MPN-SVT were registered on the online database between
May 2019 and January 2022. 1 centre registered 92 cases (40%) and 2 centres each registered
39 cases (17%) with the remaining 62 cases (27%) from 6 centres. 57% of patients were
female and 43% male and the age of SVT onset was 49 years or less in 70% of patients.
Median follow up was 7.3 years (range 35 days-31.8 years). SVT was diagnosed in 2009
or later in 72%.
MPN was diagnosed first in 30% of patients, SVT first in 42% of patients and simultaneous
diagnosis in 28%. Figure 1 shows the subtypes of MPN associated with SVT.
Multiple splanchnic vein thromboses were observed in 42% of patients. Of those with
single vein thrombosis, the portal vein was affected in 26% and the hepatic vein in
23%.
Initial anticoagulation treatment was in line with local policy. TIPSS (transjugular
intrahepatic portosystemic shunt) was performed in 21% and thrombolysis in 7%. Warfarin
only was the preferred long-term anticoagulant (53%).
Cytoreduction at registration comprised 44% of patients on hydroxycarbamide, 18% on
Pegylated interferon, 16% on ruxolitinib and 8% on no therapy.
37 patients required major abdominal surgery and 17 received liver transplants.
71 patients (33%) suffered from other non-SVT thromboses (34% pre-SVT, 9% simultaneously
and 57% post-SVT), comprising 84 thrombotic events (60 venous, 24 arterial). Grade
3+ haemorrhage was observed in 60 patients (28%), of which the majority (48%) occurred
within the first year of SVT diagnosis.
111 patients (51%) were re-imaged at least once within 12 months post-SVT, demonstrating
clot extension in 12%, recanalization in 25% and stable appearances in 54%. Overall
survival was 88% with 14 deaths, of which 6 were disease-related. Median age at death
was 72 years and death occurred at a median of 12 years post SVT diagnosis.
Image:
Summary/Conclusion: This is one of the largest cohort studies of patients with MPN-SVT
to date with over 70% diagnosed in the last 13 years. Similar to previous published
studies, our evidence corroborates the female predominance and relatively young age
at diagnosis.
Our data highlight the significant morbidity and mortality burden incurred by patients
with MPN-SVT, including a substantially lower rate of re-canalisation compared to
a recent meta-analysis of all patients with SVT and high rates of thrombosis recurrence.
We have highlighted variations in clinical practice including choice of anticoagulant,
preferred cytoreductive agent and use of thrombolysis and TIPSS.
Future studies to further understand this patient population should be focused on
access to optimal care, the aetiology of thrombosis, optimum anticoagulation management
and strategies to minimise bleeding.
S200: IMPROVED OVERALL SURVIVAL WITH FIRST-LINE BRENTUXIMAB VEDOTIN PLUS CHEMOTHERAPY
IN PATIENTS WITH STAGE III/IV CLASSICAL HODGKIN LYMPHOMA: 6-YEAR ANALYSIS OF ECHELON-1
M. Hutchings1,*, S. M. Ansell2, D. J. Straus3, J. M. Connors4, W. S. Kim5, A. Gallamini6,
R. Ramchandren7, J. W. Friedberg8, R. Advani9, A. M. Evens10, P. Smolewski11, K. J.
Savage4, N. L. Bartlett12, H.-S. Eom13, J. S. Abramson14, C. Dong15, F. Campana15,
K. Fenton16, M. Puhlmann16, J. Radford17
1Department of Haematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen,
Denmark; 2Division of Hematology, Mayo Clinic, Rochester, MN; 3Department of Medicine,
Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
of America; 4BC Cancer Centre for Lymphoid Cancer and Department of Medical Oncology,
Vancouver, Canada; 5Division of Hematology-Oncology, Department of Internal Medicine,
Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea;
6Research and Innovation, Antoine-Lacassagne Cancer Centre, Nice, France; 7The University
of Tennessee Graduate School of Medicine, Knoxville, TN; 8James P. Wilmot Cancer Institute,
University of Rochester Medical Center, Rochester, NY; 9Department of Medicine, Division
of Oncology, Stanford University, Stanford, CA; 10Division of Blood Disorders, Rutgers
Cancer Institute of New Jersey, New Brunswick, NJ, United States of America; 11Department
of Experimental Hematology, Medical University of Lodz, Lodz, Poland; 12Washington
University School of Medicine Siteman Cancer Center, St Louis, MO, United States of
America; 13Center for Hematologic Malignancy, National Cancer Center, Goyang, South
Korea; 14Massachusetts General Hospital, Boston, MA; 15Takeda Development Center Americas,
Inc. (TDCA), Lexington MA; 16Seagen Inc., Bothell, WA, United States of America; 17The
University of Manchester and the Christie NHS Foundation Trust, Manchester Academic
Health Science Centre, Manchester, United Kingdom
Background: In classical Hodgkin lymphoma (cHL), improved overall survival (OS) with
first-line treatment using new combinations has seldom been observed compared with
existing approaches. Five-year data from the randomized phase 3 ECHELON-1 study (NCT01712490)
supported long-term progression-free survival (PFS) with brentuximab vedotin, doxorubicin,
vinblastine, and dacarbazine (A+AVD) versus doxorubicin, bleomycin, vinblastine, and
dacarbazine (ABVD), in patients (pts) with previously untreated stage III/IV cHL,
regardless of interim positron emission tomography (PET) status. The long-term safety
profile of A+AVD was manageable, and numerically fewer secondary malignancies were
reported versus ABVD, as well as a greater number of pregnancies (Straus et al, Lancet
Haematol 2021).
Aims: To report the pre-specified OS analysis from ECHELON-1, as well as relevant
long-term safety data, after approximately 6 years of follow-up (cut-off June 1, 2021).
Methods: Pts were randomized 1:1 to receive up to 6 cycles of A+AVD (n=664) or ABVD
(n=670) on day 1 and 15, every 28 days. PFS per investigator was reported for long-term
follow-up. The key secondary end point was OS, which was an event-driven, pre-specified,
alpha-controlled analysis in the intention-to-treat population, with a prespecified
interim analysis after 103 deaths. Analysis of OS in prespecified subgroups was exploratory
and was not adjusted for multiplicity. Informed consent was obtained for all pts.
Results: In total, 39 OS events in the A+AVD arm and 64 in the ABVD arm had occurred
at a median follow-up of 73 months, significantly favoring A+AVD (hazard ratio [HR]
0.590; 95% CI 0.396–0.879; p=0.009). There was a consistent OS benefit for A+AVD vs
ABVD across prespecified subgroups, including: stage III (HR: 0.863; 95% CI 0.452–1.648)
and stage IV disease (HR: 0.478; 95% CI 0.286–0.799) at diagnosis, pts who were PET-negative
at cycle 2 (PET2)-negative (HR: 0.583; 95% CI 0.338–0.856) and PET2-positive (HR:
0.163; 95% CI 0.037–0.717), pts aged <60 years (HR: 0.509; 95% CI 0.291–0.890), pts
aged ≥60 years (HR: 0.829; 95% CI 0.469–1.466), and across all geographies, including
Europe (HR: 0.783; 95% CI 0.467–1.315) and North America (HR: 0.327; 95% CI 0.153–0.699).
The 6-year PFS estimate was 82.3% (79.1–85.0) vs 74.5% (70.8–77.7) with A+AVD vs ABVD,
respectively (HR: 0.678; 95% CI: 0.532–0.863). The long-term safety profile of A+AVD
was comparable to that of ABVD. In both A+AVD and ABVD groups, treatment-emergent
peripheral neuropathy continued to resolve or improve, with 86% (379/443) and 87%
(249/286) of cases in the A+AVD and ABVD arms either completely resolving (72% vs
79%) or improving (14% vs 8%) by last follow-up. Overall, 23 secondary malignancies
in the A+AVD arm and 32 in the ABVD arm were reported. Pregnancies and live births
were reported by a greater number of female pts in the A+AVD group vs the ABVD group
(49 vs 28 and 42 vs 19, respectively) and there were no stillbirths reported during
the study. There were no new safety signals.
Summary/Conclusion: There was a statistically significant 41% reduction in the risk
of death with A+AVD vs ABVD, with a consistent OS benefit across prespecified subgroups.
The long-term safety profile was manageable, consistent with prior reports. These
data support A+AVD as a preferred option for previously untreated stage III and IV
cHL.
S201: CAMIDANLUMAB TESIRINE: UPDATED EFFICACY AND SAFETY IN AN OPEN-LABEL, MULTICENTER,
PHASE 2 STUDY OF PATIENTS WITH RELAPSED OR REFRACTORY CLASSICAL HODGKIN LYMPHOMA (R/R
CHL)
C. Carlo-Stella1,*, S. Ansell2, P. L Zinzani3, J. Radford4, K. Maddocks5, A. Pinto6,
G. P. Collins7, V. Bachanova8, N. Bartlett9, I. Bence-Bruckler10, M. Hamadani11, J.
Kline12, J. Mayer13, K. J. Savage14, R. Advani15, P. Caimi16, R.-O. Casasnovas17,
T. Feldman18, B. Hess19, M. Bastos-Oreiro20, S. Iyengar21, S. Eisen22, Y. Negievich22,
L. Wang23, J. Wuerthner22, A. F. Herrera24
1Department of Oncology and Hematology, Humanitas Clinical and Research Center - IRCCS
and Humanitas University, Rozzano, Milano, Italy; 2Division of Hematology, Mayo Clinic,
Division of Hematology, Minnesota, United States of America; 3Hematology, Institute
of Hematology Seràgnoli University of Bologna, Bologna, Italy; 4NIHR Clinical Research
Facility, the Christie NHS Foundation Trust and University of Manchester, Manchester
Academic Health Science Centre, Manchester, United Kingdom; 5Division of Hematology,
Ohio State University Medical Center, Columbus, United States of America; 6Fondazione
G. Pascale, IRCCS, Istituto Nazionale Tumori, Naples, Italy; 7Oxford Cancer and Haematology
Centre, NIHR Oxford Biomedical Research Centre, Churchill Hospital, Oxford, United
Kingdom; 8Division of Hematology, Oncology, and Transplantation, University of Minnesota,
Minneapolis; 9Division of Oncology, Washington University School of Medicine in St.
Louis, St. Louis, United States of America; 10Ottawa-Hospital General Campus, The
Ottawa Hospital-General Campus, Ottawa, Canada; 11Medical College of Wisconsin, BMT
& Cellular Therapy Program, Department of Medicine, Milwaukee; 12Department of Medicine,
The University of Chicago, Chicago, United States of America; 13Internal Medicine,
University Hospital and Masaryk University, Brno, Czechia; 14Department of Medical
Oncology, BC Cancer and University of British Columbia, Vancouver, Canada; 15Division
of Oncology, Department of Medicine, Stanford University, Stanford; 16Cancer Center,
Cleveland Clinic/Case Comprehensive Cancer Center, Cleveland, United States of America;
17Department of Hematology, University Hospital F. Mitterrand and Inserm UMR 1231,
Dijon, France; 18John Theurer Cancer Center, Hackensack Meridian Health, Hackensack;
19Division of Hematology and Medical Oncology, Department of Medicine, Medical University
of South Carolina, Charleston, United States of America; 20Haematology Department,
Gregorio Marañón Health Research Institute, Hospital General Universitario Gregorio
Marañón, Madrid, Spain; 21Royal Marsden Hospital, London, United Kingdom; 22ADC Therapeutics
SA, Epalinges, Switzerland; 23ADC Therapeutics America, Inc, Murray Hill; 24Department
of Hematology & Hematology & Hematopoietic Cell Transplantation, City of Hope Comprehensive
Cancer Center, Duarte, United States of America
Background: Camidanlumab tesirine (Cami), an antibody-drug conjugate comprising a
human IgG1 anti-CD25 monoclonal antibody conjugated to a pyrrolobenzodiazepine (PBD)
dimer, displayed antitumor activity and manageable toxicity in a phase 1 trial in
lymphoma, including R/R cHL (Hamadani et al. 2021).
Aims: Present updated efficacy and safety data from a phase 2 study of Cami monotherapy
in patients (pts) with R/R cHL (NCT04052997).
Methods: Pts with R/R cHL and ≥3 prior systemic therapies, including brentuximab vedotin
(BV) and anti-PD-1 (≥2 therapies if hematopoietic cell transplantation [HCT] ineligible),
were enrolled. Interim data was reported (Herrera et al. 2020, Zinzani et al. 2021).
The primary endpoint is overall response rate (ORR by 2014 Lugano criteria; central
review). Secondary endpoints include duration of response (DOR), progression-free
survival (PFS), and frequency and severity of adverse events (AEs). Pts received Cami
45 µg/kg (30-min infusion) on Day 1 of each 3-week cycle (2 cycles), then 30 µg/kg
(subsequent cycles) for up to 1 year.
Results: As of Nov 1, 2021, enrollment was complete (N=117). Median age (range) was
37 years (19-87), 62% of pts were male, and 95% had an ECOG score of 0-1. Pts had
received a median (range) of 6 (3-19) prior therapies and received a median (range)
of 5.0 (1-15) Cami cycles. Fourteen pts (12.0%) withdrew to undergo HCT (of which
12 [10.3%] received HCT and were censored). In the all-treated population (N=117),
ORR was 70.1% (82/117; 95% CI: 60.9-78.2), and 33.3% (39/117) had complete response
(CR) (Fig 1A). At median (range) follow-up of 10.7 (1.2-25.2+) months (mos), the median
(95% CI) DOR was 13.7 mos (7.4-14.7) for all responders, 14.5 (7.4-not reached, NR)
mos and 7.9 (3.8-NR) mos for pts with CR or PR, respectively (Fig 1B). Median (95%
CI) PFS was 9.1 (5.1-15.0) mos.
All-grade treatment-emergent AEs (TEAEs) in ≥25% of 117 pts were fatigue (45, 38.5%),
maculopapular rash (MR, 38, 32.5%), pyrexia (35, 29.9%), nausea (32, 27.4%), and rash
(31, 26.5%). Grade ≥3 TEAEs in ≥5% of pts were thrombocytopenia (11, 9.4%), anemia
(10, 8.5%), hypophosphatemia (9, 7.7%), neutropenia (9, 7.7%), MR (8, 6.8%), and lymphopenia
(6, 5.1%). TEAEs leading to Cami dose delay/reduction or discontinuation occurred
in 66 (56.4%) and 32 (27.4%) pts, respectively. TEAEs considered PBD-associated were
skin/nail reactions (any grade, 87, 74.4%; grade ≥3, 24, 20.5%), hepatobiliary test
abnormalities (any grade, 34, 29.1%; grade ≥3, 8, 6.8%), and edema/effusion (any grade,
20, 17.1%; grade ≥3, 0).
TEAEs considered immune-related (IR) occurred in 38 (32.5%) pts. Grade ≥3 IR AEs (TEAEs
and non-TEAEs; 10, 8.5%) included diabetic ketoacidosis/type 1 diabetes (2, 1.7%);
drug-induced liver injury (2, 1.7%); tubulointerstitial nephritis (2, 1.7%); and aplastic
anemia, autoimmune hemolytic anemia or hepatitis, and lichenoid keratosis (1 each,
0.9%).
Guillain–Barré syndrome (GBS)/polyradiculopathy was seen in 8 (6.8%) pts (Grade 2,
n=2; grade 3, n=3; grade 4, n=3). In 4 pts, onset was after 2 Cami cycles (range:
2-7) and median (range) duration was 115 days (43-523). At data cutoff, 4 cases had
recovered, and 4 had not recovered.
Image:
Summary/Conclusion: With median follow-up of 10.7 mos, Cami demonstrated an ORR of
70.1% (CR: 33.3%) in heavily pretreated R/R cHL after BV and PD-1 blockade failure,
with an encouraging median DOR of 13.7 mos and median PFS of 9.1 mos. Safety is consistent
with prior findings, including similar incidence rates of GBS/polyradiculopathy.
S202: CHILDBEARING AMONG CLASSICAL HODGKIN LYMPHOMA SURVIVORS TREATED WITH BEACOPP
AND ABVD IN SWEDEN, DENMARK, AND NORWAY
J. P. Entrop1,*, C. E. Weibull1, K. E. Smedby1, L. H. Jakobsen2 3, A. K. Øvlisen2,
D. Molin4, I. Glimelius4, A. Marklund5, K. B. Smeland6, T. C. El-Galaly2, S. Eloranta1
1Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet,
Stockholm, Sweden; 2Department of Hematology, Clinical Cancer Research Center, Aalborg
University Hospital; 3Department of Mathematical Sciences, Aalborg University, Aalborg,
Denmark; 4Department of Immunology, Genetics and Pathology, Unit of Experimental and
Clinical Oncology, Uppsala University, Uppsala; 5Department of Reproductive Medicine,
Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm,
Sweden; 6Department of Oncology, Oslo University Hospital, Oslo, Norway
Background: Recent studies have shown decreased childbearing rates in some classical
Hodgkin lymphoma (cHL) survivors compared to the general population. This decrease
is hypothesized to be caused by administered intensive chemotherapy. Understanding
if currently used treatment protocols lead to reduced post-treatment childbirth potential
is important as differential impact on childbearing may influence treatment decisions
for younger patients. Firm conclusions from previous population-based studies have
been hampered by low power for specific treatment contrasts.
Aims: To increase understanding of differences in childbearing patterns among male
and female HL survivors treated with ABVD and BEACOPP in a study combining population-based
register data from Sweden, Denmark, and Norway.
Methods: In this cohort study 2,937 individuals aged 18-40 years with a recorded diagnosis
of cHL in the Swedish lymphoma register (SLR), the Danish lymphoma registry (LYFO),
or the clinical database at Oslo University Hospital (OUH) between 1995 and 2019 were
included. Information on first-line chemotherapy treatment was available in the registers.
Treatment regimens were classified as ABVD, 6-8 BEACOPP, or other. The SLR, LYFO,
and OUH databases were linked to national medical birth registers to obtain information
on childbirths. Follow-up started 9 months after cHL diagnosis and was accrued until
date of first childbirth, death, or administrative censoring (December 2017, 2018,
or 2019 for Norway, Sweden, and Denmark, respectively) or after 10 years. Females
were additionally censored at date of relapse or stem cell transplantation due to
the limited accessibility of assisted reproductive techniques for female patients
experiencing one of those events. Stratified Cox models allowing for effect modification
between sex and treatment and adjusted for stage, age at diagnosis, year of diagnosis,
parity, and performance status were used to estimate hazard ratios (HRs). Flexible
parametric survival models were used to estimate marginal childbirth rates and standardized
cumulative incidence of childbirth across treatment groups. A comprehensive study
analysis plan been published and pre-registered on the Open Science Framework (https://osf.io/eumy5/).
Results: In summary, 75% of HL patients were treated with ABVD and 10.8% with 6-8
BEACOPP. An additional 14.2% received other treatments or had missing treatment information
and were excluded from further analyses. The rate of first childbirth per 1,000 person-years
for males and females was 45.5 and 50.0 respectively in the ABVD group, and 23.8 and
43.5 in the 6-8 BEACOPP group. The adjusted HR of first childbirths for males and
females, comparing patients treated with 6-8 BEACOPP to ABVD were 0.54 (95% CI 0.37-0.79)
and 0.92 (95% CI 0.61-1.39) respectively. Plots of the cause-specific cumulative incidence
functions (CIF) of childbirth show a constantly lower proportion of male patients
with childbirths over time since HL diagnosis comparing patients treated with 6-8
BEACOPP and ABVD. However, this difference was not present among female patients.
Adjusted marginal estimates of the cause-specific CIF of childbirth showed the same
pattern.
Image:
Summary/Conclusion: BEACOPP treatment is associated with decreased childbearing rates
in male but not female HL patients. Infertility counselling for this group is advisable.
S203: ABSCOPAL EFFECT OF RADIOTHERAPY AND NIVOLUMAB IN RELAPSED OR REFRACTORY HODGKIN
LYMPHOMA: PRE-PLANNED INTERIM ANALYSIS OF THE INTERNATIONAL GHSG PHASE II AERN TRIAL
P. Bröckelmann1 2 3 4,*, I. Bühnen3, J. Zijlstra5, A. Fossa6, J. Meissner7, S. Mathas8,
J. Rosenbrock9, C. Kobe10, M. Fuchs3, A. Plütschow3, S. Marnitz9, P. Borchmann11,
A. Engert11, C. Baues9
1Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne
Duesseldorf (CIO ABCD), University Hospital of Cologne; 2Max Planck Institute for
the Biology of Ageing; 3German Hodgkin Study Group (GHSG), University of Cologne;
4Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf (MSSO ABCD), Cologne,
Germany; 5Amsterdam UMC, Department of Hematology, Cancer Center Amsterdam, Vrije
Universiteit Amsterdam, Amsterdam, Netherlands; 6Department of Oncology, Oslo University
Hospital, Oslo, Norway; 7Faculty of Medicine and University Hospital Heidelberg, Department
V of Internal Medicine, University of Heidelberg, Heidelberg; 8Department of Hematology,
Oncology and Tumor Immunology, Charité – Universitätsmedizin Berlin, and Experimental
and Clinical Research Center (ECRC), Berlin; 9University Hospital Cologne, Department
of Radiation Oncology and Cyberknife Center; 10Department of Nuclear Medicine; 11Department
I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf
(CIO ABCD), University of Cologne, Faculty of Medicine, Cologne, Germany
Background: Failure of anti-PD1 checkpoint blockade in relapsed or refractory Hodgkin
lymphoma (r/r HL) constitutes an unmet medical need. It has been postulated that anti-PD1
and localized radiotherapy (RT) may work synergistically to induce a systemic anti-lymphoma
effect resulting in distant and systemic (“abscopal”) response outside the RT field.
Aims: We aimed to formally investigate safety and efficacy of nivolumab and RT in
r/r HL failing anti-PD1 treatment in a proof-of-concept study.
Methods: AERN is an ongoing international investigator-sponsored two stage phase II
trial enrolling adult patients with histologically confirmed r/r HL (NCT03480334).
All patients must have failed anti-PD1 treatment by either showing progressive disease
(PD) or stable disease (SD) >6 months as best response at the time of enrollment.
Presence of two distinct FDG-avid lesions with one eligible for 20Gy localized RT
and the other >5cm apart and outside the prescribed 95%isodose for measurement of
an abscopal response must be confirmed by the GHSG reference panel. After enrollment
patients receive 240mg nivolumab 2-weekly and 20Gy RT in 2Gy fractions is started
on day 6 after the first infusion on trial. Treatment is continued until PD or unacceptable
toxicity or for a maximum of 1.5 years. The primary endpoint is abscopal response
rate (ARR) at first interim restaging after six infusions, secondary endpoints include
objective response rate (ORR), progression-free survival (PFS), overall survival and
safety. Target enrollment are nine and 20 patients in a Simon’s two-stage desig. A
pre-planned interim analysis was conducted after completion of stage I and is reported
herein.
Results: Of 11 patients enrolled into stage I of the trial, two terminated trial participation
early (screening failure, withdrawal of informed consent) and nine patients at a median
age of 34 (range 25-82) years were evaluable for stage I. All patients presented with
stage III/IV and a median of 4 (3-15) prior lines of treatment. While all patients
had received prior anti-PD1 antibodies (100%), the majority also received brentuximab
vedotin (89%) and high-dose chemotherapy plus autologous stem-cell transplantation
(ASCT, 56%). Trial treatment was well tolerated. The most frequently reported °1/2
toxicities were those affecting the skin (55%), one case of °3 gastrointestinal and
no °4/5 toxicities. Patients received a median of 19 (7-36) nivolumab infusions with
a median treatment duration of 9 (3-18) months. At first interim restaging, five patients
had an abscopal response (ARR 56%, 95%CI 16.9-74.9%) by central response assessment
and 56% achieved a PR (ORR 56%). Most patients experienced a reduction in disease
burden at first interim restaging with a mean decrease of SPD -36.3% (0 to -81.2%).
Four out of five patients discontinuing treatment experienced PD at the last restaging.
With a median observation time of 15.5 months, the 1-year PFS was 42.3% and all patients
were alive at data cut off.
Summary/Conclusion: Treatment with nivolumab and 20Gy RT at a single lesion was well
tolerated and led to an abscopal response in the majority of patients (56%) previously
failing anti-PD1 treatment for r/r HL. An ORR of 56% and 1-year PFS of 42.3% further
highlight the potential of this innovative treatment strategy. After meeting the primary
efficacy endpoint in this pre-planned interim analysis, AERN trial enrollment is currently
continuing internationally.
S204: PEMBROLIZUMAB IN CHILDREN AND YOUNG ADULTS WITH NEWLY DIAGNOSED CLASSICAL HODGKIN
LYMPHOMA WITH SLOW EARLY RESPONSE TO FRONTLINE CHEMOTHERAPY: THE PHASE 2, OPEN-LABEL,
KEYNOTE-667 STUDY
L. Vinti1,*, S. Daw2, C. Sabado Alvarez3, F. Fagioli4, A. Beishuizen5, G. Michel6,
M. L. Moleti7, M. Cepelova8, A. Thorwarth9, C. Rigaud10, D. Plaza Lopez de Sabando11,
J. Landman Parker12, Y. Zhu13, P. Pillai13, A. Nahar13, C. Mauz-Koerholz14 15
1IRCCS Ospedale Pediatrico Bambino, Gesu, Italy; 2University College London Hospitals
NHS Foundation Trust, London, United Kingdom; 3Hospital Universitari Vall d Hebron,
Barcelona, Spain; 4Ospedale Infantile Regina Margherita, Turin, Italy; 5Princess Máxima
Centrum, Utrecht, Netherlands; 6CHU de Marseille Hopital de la Timone Enfants, Marseille,
France; 7Universita degli Studi di Roma La Sapienza, Rome, Italy; 8Fakultni nemocnice
v Motole, Prague, Czechia; 9Charite-Universitaetsmedizin Berlin Campus Virchow-Klinikum,
Berlin, Germany; 10Gustave Roussy Cancer Campus, Villejuif, France; 11Hospital Universitario
La Paz, Madrid, Spain; 12Hopital d’Enfants Armand Trousseau, Paris, France; 13Merck
Sharp & Dohme, Corp., a subsidiary of Merck & Co., Inc., Kenilworth, United States
of America; 14Justus-Liebig University of Giessen, Giessen; 15Medical Faculty, Martin-Luther-University
of Halle-Wittenberg, Halle, Germany
Background: Patients with classical Hodgkin lymphoma (cHL) with slow early response
(SER) to initial chemotherapy (chemo) are at higher risk of relapse, and the burden
of late organ toxicities may be higher after dose intensification and radiotherapy
(RT). The phase 2, open-label KEYNOTE-667 (NCT03407144) study is evaluating the efficacy
and safety of pembrolizumab (pembro) plus chemo in patients with cHL and SER to frontline
chemo.
Aims: To present results of an interim analysis of efficacy and safety in patients
with high risk cHL (Group 2) who were SER.
Methods: In Group 2, eligible patients aged 3-17 (children) or 18-25 years (young
adults) with newly-diagnosed stage IIEB, IIIEA, IIIEB, IIIB, IVA, or IVB cHL were
enrolled to receive induction with vincristine, etoposide, prednisone/prednisolone,
doxorubicin (OEPA) for 2 cycles. Response assessment after induction (early) and consolidation
(late) therapy was done by PET/MRI/CT. After induction treatment, patients with rapid
early response (RER) received non-study therapy and patients with SER received consolidation
with 4 cycles cyclophosphamide, vincristine, prednisone/prednisolone, dacarbazine
(COPDAC-28) plus pembro 2 mg/kg up to 200 mg IV Q3W (3-17 years of age) or 200 mg
IV Q3W (18-25 years of age). After consolidation therapy, patients with PET-positivity
(Deauville score 4-5) received modified involved-site RT to late PET-positive residua;
RT was omitted in patients with PET-negativity. All patients with SER received maintenance
pembro Q3W, for a total of 17 doses. Safety analyses included all patients with SER
treated with pembro, while efficacy analyses was based on all patients who had completed
late response assessment. All patients provided informed consent. The primary endpoint
was ORR by BICR per Cheson 2007 IWG criteria in patients with SER. Secondary endpoints
included rate of PET-negativity after consolidation, details of RT, and safety. Data
cut-off was Nov 22, 2021.
Results: At data cut-off, median (range) follow-up was 9.6 mo (2.5-21.2). A total
of 30 patients with high-risk cHL with SER were included. Median (range) age was 15y
(6-19), 13 (43%) had bulky disease, 19 (63%) had Ann Arbor stage IV disease. A total
of 6 (20%) patients had completed, and 24 (80%) were ongoing on treatment, with median
time on treatment of 3.3 mo (range, 0-11.8). Of 30 patients, 25 (83%) had a late response
assessment, of which 17 (68%) were PET-negative by BICR; 18 (72%) PET-negative by
investigator. All cause AEs occurred in 23 (77%) patients, with 14 (47%) having a
treatment-related AE. Grade ≥3 AEs occurred in 6 (20%) patients, with 3 (10%) having
an SAE. Grade ≥3 treatment-related AEs occurred in 2 (7%) patients. One (3%) pt had
a grade 2 immune-mediated AE of hypothyroidism.
Summary/Conclusion: In pediatric patients with high-risk cHL and SER to standard OEPA
induction, pembro in combination with COPDAC 28 consolidation therapy was well tolerated
and resulted in 68% of patients having a PET-negative response at end of chemo, and
being spared RT. This early data suggests that addition of pembro potentially may
have the ability to augment responses in this high-risk subgroup of patients.
S205: ZANUBRUTINIB + OBINUTUZUMAB (ZO) VS OBINUTUZUMAB (O) MONOTHERAPY IN PATIENTS
(PTS) WITH RELAPSED OR REFRACTORY (R/R) FOLLICULAR LYMPHOMA (FL): PRIMARY ANALYSIS
OF THE PHASE 2 RANDOMIZED ROSEWOOD TRIAL
P. L. Zinzani1,*, J. Mayer2, R. Auer3, F. Bijou4, A. C. de Oliveira5, C. R. Flowers6,
M. Merli7, K. Bouabdallah8, P. S. Ganly9, Y. Song10, H. Zhang11, R. Johnson12, A.
M García-Sancho13, M. Provencio14, M. Trněný15, S. Yuen16, H. Tilly17, E. Kingsley18,
G. Tuyman19, S. E. Assouline20, E. Ivanova21, P. Kim22, J. Huang22, R. Delarue21,
J. Trotman23 24
1Institute of Hematology “Seràgnoli”, University of Bologna, Bologna, Italy; 2Department
of Internal Medicine-Hematology and Oncology, Masaryk University and University Hospital,
Brno, Czechia; 3St. Bartholomew’s Hospital, Barts Health NHS Trust, London, United
Kingdom; 4Institut Bergonié, Bordeaux, France; 5Institut Catala d’Oncologia (ICO)
Hospital Duran I Reynals, Hospital, Barcelona, Spain; 6Department of Lymphoma/Myeloma,
The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America;
7Hematology, University Hospital “Ospedale di Circolo e Fondazione Macchi” - ASST
Sette Laghi, University of Insubria, Varese, Italy; 8Hôpital Haut-Lévêque, CHU Bordeaux,
Pessac, France; 9Department of Haematology, Christchurch Hospital, Christchurch, New
Zealand; 10Beijing Cancer Hospital, Beijing; 11Tianjin Medical University Cancer Institute
& Hospital, Tianjin, China; 12St. James’s University Hospital Trust, Leeds, United
Kingdom; 13Hospital Universitario de Salamanca, Salamanca; 14Hospital Universitario
Puerta de Hierro — Majadahonda, Madrid, Spain; 15Vseobecna fakultní nemocnice v Praze,
Prague, Czechia; 16Calvary Mater Newcastle, Waratah, New South Wales, Australia; 17Centre
Henri-Becquerel, Rouen, France; 18Comprehensive Cancer Centers of Nevada, Las Vegas,
NV, United States of America; 19Department of Chemotherapy of Hemoblastosis, Blokhin
Russian Cancer Research Center, Moscow, Russia; 20Jewish General Hospital, Montreal,
Canada; 21BeiGene Switzerland GmbH, Basel, Switzerland; 22BeiGene (Beijing) Co., Ltd.,
Beijing, China and BeiGene USA, Inc., San Mateo, CA, United States of America; 23Concord
Repatriation General Hospital; 24Department of Haemotology, University of Sydney,
Concord, New South Wales, Australia
Background: FL is the most common type of indolent non-Hodgkin lymphoma. Approved
treatment options are limited for pts with R/R FL. In a phase 1b trial (Blood Adv.
2020;4(19):4802-4811), ZO was found to be tolerable and associated with early signal
of efficacy.
Aims: To present a primary analysis of ROSEWOOD (BGB-3111-212; NCT03332017), a phase
2, randomized study designed to assess efficacy and safety of ZO vs O in pts with
R/R FL.
Methods: Pts with R/R FL who received ≥2 lines of therapy, including an anti-CD20
antibody and an alkylating agent, were randomized 2:1 to receive either ZO or O. O
was given in both arms on Days 1, 8, and 15 of Cycle 1, Day 1 of Cycles 2-6, and then
every 8 weeks up to 20 doses maximum. Z (160 mg twice daily) was given until progressive
disease (PD) or unacceptable toxicity; pts with confirmed PD or no response within
12 months in the O arm were allowed to crossover to ZO. Primary endpoint was overall
response rate (ORR) by independent central review. Secondary endpoints included complete
response rate (CRR), duration of response (DOR), progression-free survival (PFS),
overall survival (OS), and safety. Exploratory endpoint included ORR by investigator
after crossover. Primary analysis cutoff was October 8, 2021. All pts gave informed
consent.
Results: A total of 217 pts were randomized to ZO (n=145) or O (n=72). Median study
follow-up was 12.5 mo; median age was 64 yrs. Incidence of high FL International Prognostic
Index score was 53% (ZO) and 51% (O). Pts received a median of 3 prior lines of therapy,
with 28% (ZO) and 25% (O) of pts receiving >3 lines. Proportion of pts refractory
to rituximab, refractory to the most recent line of therapy, or with PD within 24
mo of initiation of first-line therapy was 54%, 32%, and 35% with ZO and 50%, 40%,
and 42% with O, respectively. The study met its primary endpoint: ORR was 68.3% with
ZO vs 45.8% with O (p=0.0017). CRR was 37.2% (ZO) vs 19.4% (O); 18-mo DOR rate was
70.9% (ZO) vs 54.6% (O); and median PFS was 27.4 mo (ZO) vs 11.2 mo (O; hazard ratio
[HR], 0.51 [95% CI, 0.32-0.81], p=0.0040). Median time to new anti-lymphoma therapy
or crossover was not evaluable (ZO; NE) vs 12.1 mo (O; HR, 0.37 [95% CI, 0.23-0.60],
p<0.0001). ORR for 29 pts who crossed over to ZO was 24.1%. Median OS was NE; 18-mo
OS probability was 85.4% (ZO) vs 72.6% (O). Most common any-grade adverse events (AEs)
with incidence >10% in the ZO arm were thrombocytopenia (34.3%), neutropenia (27.3%),
diarrhea (16.1%), fatigue (14.0%), constipation (13.3%), cough (11.9%), pyrexia (11.2%),
and dyspnea (10.5%). Grade ≥3 AEs with incidence >5% with ZO were neutropenia (22.4%)
and thrombocytopenia (14.0%); incidence of atrial fibrillation was 0.7% and major
hemorrhage was 1.4%. Incidence of treatment-emergent AEs leading to death was 5.6%
(ZO) and 9.9% (O).
Summary/Conclusion: ZO demonstrated superior efficacy to O in treatment of pts with
R/R FL. ZO had a favorable benefit-risk profile and represents a potential combination
therapy for pts with R/R FL.
S206: OBINUTUZUMAB PLUS CHEMOTHERAPY DEMONSTRATES LONG-TERM BENEFIT OVER RITUXIMAB
PLUS CHEMOTHERAPY IN PATIENTS WITH PREVIOUSLY UNTREATED FOLLICULAR LYMPHOMA: FINAL
ANALYSIS OF THE GALLIUM STUDY
W. Townsend1,*, W. Hiddemann2, C. Buske3, G. Cartron4, D. Cunningham5, M. J. Dyer6,
J. Gribben7, E. Phillips8, M. Dreyling2, J. F. Seymour9, A. Grigg10, T.-Y. Lin11,
X.-N. Hong12, D. Kingbiel13, T. G. Nielsen13, A. Knapp13, M. Herold14, R. Marcus15
1Cancer Research UK and UCL Cancer Trials Centre, University College Hospitals London,
London, United Kingdom; 2Ludwig-Maximilians-University Hospital Munich, Munich; 3Universtitätsklinikum
Ulm, Ulm, Germany; 4CHU Montpellier, Montpellier, France; 5Royal Marsden Hospital,
Sutton; 6Ernest and Helen Scott Haematological Research Institute, University of Leicester,
Leicester; 7Barts Cancer Institute, Queen Mary University of London, London; 8University
of Manchester, The Christie Hospital and National Institutes of Health Research Manchester
Biomedical Research Centre, Manchester, United Kingdom; 9Peter MacCallum Cancer Centre
and the Royal Melbourne Hospital, Melbourne; 10Austin Hospital, Austin, Australia;
11Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South
China, and Collaborative Innovation Center for Cancer Medicine, Guangzhou; 12Fudan
University Shanghai Cancer Center, Shanghai, China; 13F. Hoffmann-La Roche Ltd, Basel,
Switzerland; 14HELIOS-Klinikum Erfurt, Erfurt, Germany; 15Kings College Hospital,
London, United Kingdom
Background: Immunochemotherapy with rituximab plus chemotherapy (R-chemo) has significantly
improved outcomes for patients (pts) with previously untreated follicular lymphoma
(FL). However, most pts still experience disease relapse, progression, or death. Obinutuzumab
(G) is a type II anti-CD20 monoclonal antibody with enhanced direct cell killing,
antibody-dependent cellular cytotoxicity and antibody-dependent phagocytosis versus
R. The safety and efficacy of G-chemo versus R-chemo was assessed in pts with previously
untreated, advanced stage FL in the randomized, Phase III GALLIUM (NCT01332968) study.
In the primary analysis, G-chemo demonstrated a significant improvement in progression-free
survival (PFS) versus R-chemo, with a manageable safety profile (Marcus, et al. 2017);
this efficacy benefit was maintained after 5 years of observation (Townsend, et al.
2020).
Aims: To report the final analysis of the GALLIUM study after a median observation
time of 8 years.
Methods: Pts aged ≥18 years with previously untreated histologic Grade 1–3a FL requiring
treatment were enrolled. Pts were randomized 1:1 to receive G 1000mg intravenously
(IV; Days [D]1, 8 and 15 of Cycle 1 and D1 of subsequent cycles) or R 375mg/m2 IV
(D1 of each cycle) plus chemo for 6 or 8 cycles depending on the chemo backbone selected
at each institution (cyclophosphamide, doxorubicin, vincristine, and prednisolone
[CHOP]; cyclophosphamide, vincristine, and prednisolone [CVP]; or bendamustine). Pts
attaining a complete or partial response received maintenance with the same antibody
every 2 months for 2 years or until disease progression (PD). The primary endpoint
was investigator-assessed PFS; secondary endpoints included time-to-next anti-lymphoma
treatment (TTNLT), overall survival (OS) and incidence of adverse events (AEs). All
pts provided written informed consent.
Results: 1202 pts with FL were enrolled (G-chemo, n=601; R-chemo, n=601). As of July
30, 2021, median observation time was 8 years. Seven-year PFS was improved with G-chemo
(63.4%) versus R-chemo (55.7%; hazard ratio [HR], 0.77; 95% confidence interval [CI]:
0.64–0.93; p=0.006; Figure). TTNLT was also improved with G-chemo versus R-chemo (HR,
0.71; 95% CI: 0.58–0.87; p=0.001); the proportion of pts who had not started their
next treatment at 7 years was 74.1% and 65.4%, respectively. Disease transformation
was observed in 4.2% of pts with G-chemo and 5.0% of pts with R-chemo. Seven-year
OS was similar in both arms, 88.5% with G-chemo versus 87.2% with R-chemo (HR, 0.86;
95% CI: 0.63–1.18; p=0.36). Seventy-five pts in the G-chemo arm and 86 pts in the
R-chemo arm had died, most commonly due to PD (4.2% and 6.0%, respectively). The incidence
of serious AEs (SAEs) was 48.9% with G-chemo (28.2% and 24.4% during induction and
maintenance, respectively) and 43.4% with R-chemo (24.6% and 21.7%, respectively).
Rates of fatal AEs were similar with G-chemo (4.4%) and R-chemo (4.5%); pneumonia
was the most common fatal AE (0.8% [n=5] and 0.2% [n=1], respectively) and SAE (5.9%
[n=35] and 6.2% [n=37], respectively). Second malignancies occurred in 13.1% of pts
in the G-chemo arm and 9.9% of pts in the R-chemo arm. After adjusting for observation
time, rates were similar between arms. These safety findings are consistent with previous
analyses.
Image:
Summary/Conclusion: After a median observation time of 8 years, a meaningful improvement
in PFS was maintained with G-chemo versus R-chemo in pts with previously untreated
FL, confirming the role of G-chemo as a standard of care for the first-line treatment
of pts with FL.
S207: EFFICACY AND SAFETY OF A THIRD GENERATION CD20 CART (MB-106) FOR TREATMENT OF
RELAPSED/REFRACTORY FOLLICULAR LYMPHOMA (FL)
M. Shadman1,*, C. Yeung1, M. Redman2, S. Y. Lee2, D. H. Lee2, S. Ra2, D. Qian2, C.
Ujjani1, B. Dezube3, C. Poh1, E. H. Warren1, A. Chapuis1, D. Green1, A. Cowan1, R.
Cassaday1, H.-P. Kiem1, V. Chow1, J. Gauthier1, C. Turtle1, R. Lynch1, S. Smith1,
A. Gopal1, D. Maloney1, B. Till1
1Fred Hutchinson Cancer Research Center / University of Washington; 2Fred Hutch, Seattle;
3Mustang Bio, Worcester, United States of America
Background: Follicular lymphoma (FL) is incurable and novel treatments with high efficacy
and low toxicity are needed. Chimeric antigen receptor T-cells (CAR-T) targeting CD19
are effective, and axicabtagene ciloleucel is currently approved by FDA for treatment
of patients (pts) with relapsed FL, but toxicities like cytokine-release syndrome
(CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) may limit
its use. MB-106 is a fully human 3rd-generation CD20-targeted CAR-T product with both
4-1BB and CD28 costimulatory domains.
Aims: We present the results of the FL cohort from our ongoing phase I/II clinical
trial investigating MB-106 for B-cell lymphoma/CLL.
Methods: Pts with R/R B-cell malignancies including FL were eligible after confirmation
of CD20 expression. Prior treatment with CD19 CAR-T is permitted. Lymphodepletion
(LD) consists of cyclophosphamide (Cy) ± fludarabine (Flu). CAR-T cells are administered
at one of 4 dose levels (DL): DL1: 3.3x105, DL2: 1x106, DL3: 3.3x106, DL4: 1x107 CAR
T cells/kg. A continual reassessment method dose escalation design was used to find
the maximally tolerated dose. CAR-T was infused in the outpatient setting except for
the first pt of each dose cohort (overnight observation). Initial treatment response
is assessed on day 28 and best response is reported here. CRS and ICANS are graded
per ASTCT.
Results: Between Dec 2019 and Dec 2021, 16 pts with FL were treated and had day 28
assessment. Median age was 61.5 years (range: 45 – 81). High-risk features included
pts with progression of disease within 24 months of first-line chemotherapy (POD24)
(n=11; 69%), history of histologic transformation (n=3; 19%), prior treatment with
a CD19 targeted CAR-T (n=1; 6%). 6 pts (37.5%) had a PI3 kinase inhibitor. Median
time between leukapheresis and LD was 15 days (range: 9-21) and 2 pts received bridging
therapy with lenalidomide (1) and high-dose corticosteroids (1). All DLs were reached
(DL0=1, DL1=1, DL2=4, DL3=7, DL4=2), with no dose-limiting toxicities. All CRS events
were grade 1 (n=4; 25%) or grade 2 (n=1; 6%), with no grade ≥ 3 CRS events. There
was no occurrence of ICANS of any grade. No pts had tumor lysis syndrome or Gr 3-4
infections. In the first 28 days, thrombocytopenia (Gr 3-4: 12.5%) and neutropenia
(Gr 3-4: 94%) were common but there were no bleeding complications, and the rate of
febrile neutropenia was 25%. Overall response (ORR) rate was 94% (15/16) and a complete
response (CR) rate was 75% (12/16). Pts who received DL3 or DL4, had an ORR 100% and
CR rate of 89%. The single patient who had previously been treated with a CD19 targeted
CAR-T achieved a CR. With median follow-up of 10 months, one patient died, from complications
of myelodysplastic syndrome (MDS) while still in remission; the MDS was attributed
to the prior treatments (7 lines of chemo-based therapy and radioimmunotherapy). Most
of the remissions are ongoing and 4 pts have relapsed during the follow-up median
7 months after treatment (range 3-10.5). CAR T cells were detectable by flow cytometry
at last available timepoint (up to 2 years) for 15 of 16 patients. One patient had
undetectable CAR T cells by day 201.
Summary/Conclusion: Treatment with MB-106, a third generation CD20 targeting CAR-T,
resulted in high ORR and CR rates and CAR-T persistence in FL pts and was associated
with favorable safety profile with no occurrence of Gr 3 or Gr 4 CRS and no ICANS
event of any grade. A multicenter trial for treatment of B-cell malignancies including
FL will start enrollment in 2022.
S208: EFFICACY AND SAFETY OF ZANDELISIB ADMINISTERED BY INTERMITTENT DOSING (ID) IN
PATIENTS WITH RELAPSED OR REFRACTORY (R/R) FOLLICULAR LYMPHOMA: PRIMARY ANALYSIS OF
THE GLOBAL PHASE 2 STUDY TIDAL
A. Zelenetz1,*, W. Jurczak2, V. Ribrag3, K. Linton4, G. Collins5, J. Lopéz-Jiménez6,
N. Reddy7, A. Mengarelli8, T. Phillips9, G. Musuraca10, O. Sheehy11, J. Li12, W. Xu12,
M. Azoulay13, R. Ghalie12, P. L. Zinzani14
1Memorial Sloan Kettering Cancer Institute, New York City, United States of America;
2Maria Sklodowska Curie National Research Institute of Oncology, Krakow, Poland; 3Institut
Gustave Roussy, Villejuif, France; 4Manchester Cancer Research Centre, Manchester;
5GenesisCare, Oxford, United Kingdom; 6Hospital Universitario Ramon y Cajal, Madrid,
Spain; 7Vanderbilt University, Nashville, United States of America; 8Regina Elena
National Cancer Institute, Roma, Italy; 9University of Michigan Health System, Ann
Arbor, United States of America; 10Istituto Scientifico Romagnolo per lo Studio e
la Cura dei Tumori I.R.S.T., Meldola, Italy; 11Belfast Health and Social Care Trust,
Belfast, United Kingdom; 12MEI Pharma, Inc., San Diego; 13Kyowa Kirin Co., Princeton,
United States of America; 14University of Bologna, Bologna, Italy
Background: Zandelisib, a PI3Kδ inhibitor with high target-binding affinity, is administered
by intermittent dosing (ID) on days 1 to 7 of 28-day cycles to potentially enable
regulatory T-cell repopulation and lower the risk of immune-related adverse events
(irAEs) associated with continuous PI3Kδ inhibition. In a phase 1b study in 37 patients
(pts) with R/R FL, zandelisib administered daily for two 28-day cycles for tumor debulking
then on ID achieved an overall response rate (ORR) of 87% (78% as single agent and
95% with rituximab), a median duration of response (DOR) not reached at median follow-up
of 16.9 months, and a low rate (<10%) of grade 3 irAEs (Pagel et al. ICML 2021; #113).
Aims: We conducted the TIDAL study (NCT03768505) to further evaluate in a global trial
the efficacy and safety of zandelisib in larger group of pts with R/R FL and marginal
zone lymphoma (MZL). The MZL cohort is still enrolling and not reported here.
Methods: Eligible pts ≥18 years with FL Grade I-IIIA, ECOG performance status 0-1,
progressive disease after ≥2 prior therapies including an anti-CD20 antibody and chemotherapy,
and no prior PI3Kδ inhibitor, provided consent and then received zandelisib 60 mg
daily for two 28-day cycles then on ID. Another study arm evaluating zandelisib 60 mg
daily continuously was closed to enrollment early and is not reported here. The planned
sample size in FL was 120 pts on ID, with the primary efficacy population (PEP) pre-defined
as the first 91 pts treated and the safety population consisting of all FL pts treated.
The primary efficacy endpoint was ORR as assessed by independent review using the
Lugano criteria and analyzed 6 months after completing enrollment in the PEP.
Results: 121 FL pts were enrolled. In the PEP (N=91 pts), the median number of prior
therapies was 3 (range 2-8), 21 pts (23%) have received prior stem cell transplant,
42 pts (46%) were refractory to last therapy, 31 pts (34%) had tumors ≥5 cm, and 51
pts (56%) were POD24. The ORR was 70.3% (N=64) (95% CI 59.8-79.5%), with 32 pts (35.2%)
achieving a complete response (CR). Responses occurred early, with 85.5% (N=56) achieved
in the first 2 cycles of therapy and 75% of CRs (N=24) achieved the first 4 cycles.
The data are still immature to estimate accurately the DOR. With a median follow-up
of 9.4 months (range 0.8-24) in the safety population of 121 pts, 12 pts (9.9%) discontinued
therapy due to any drug-related AE. Grade 3 AEs of special interest (AESI) were diarrhea
in 6 pts (5%), colitis in 2 (1.7%), cutaneous rash in 4 (3.3%), stomatitis in 3 (2.5%),
and 1 (0.8%) each for AST and ALT elevation, and non-infectious pneumonitis. Grade
3 AESIs primarily (15 of 18, 83%) occurred in cycles 1-3, during daily dosing, with
only 3 cases reported on ID in Cycles ≥4.
Summary/Conclusion: Zandelisib on ID achieved high ORR (70.3%) and CR rate (35.2%)
in heavily pretreated R/R FL pts, and was associated with a low rate (<10%) of grade
3 AESI and discontinuations due to AEs, results comparable to the Phase 1b study.
Longer follow-up is needed to estimate median DOR. This profile supports evaluation
of zandelisib as a single agent and in combination regimens in various B-cell malignancies,
both in R/R disease and in earlier lines of therapy. Zandelisib plus rituximab vs
chemoimmunotherapy is being evaluated in the phase 3 study COASTAL in R/R FL and MZL
(NCT04745832).
S209: PRIMARY RESULTS FROM THE PHASE 3 SHINE STUDY OF IBRUTINIB IN COMBINATION WITH
BENDAMUSTINE-RITUXIMAB (BR) AND R MAINTENANCE AS A FIRST-LINE TREATMENT FOR OLDER
PATIENTS WITH MANTLE-CELL LYMPHOMA
M. L. Wang1,*, W. Jurczak2, M. Jerkeman3, J. Trotman4, P. L. Zinani5, D. Belada6,
C. Boccomini7, I. W. Flinn8, P. Giri9, A. Goy10, P. A. Hamlin11, O. Hermine12, J.-Á.
Hernández-Rivas13, X. Hong14, S. J. Kim15, D. Lewis16, Y. Mishima17, M. Özcan18, G.
F. Perini19, C. Pocock20, Y. Song21, S. E. Spurgeon22, J. M. Storring23, J. Walewski24,
J. Zhu21, R. Qin25, T. Henninger25, S. Deshpande25, A. Howes25, S. Le Gouill26, M.
Dreyling27
1The University of Texas MD Anderson Cancer Center, Houston, United States of America;
2Maria Sklodowska-Curie National Research Institute of Oncology, Karkow, Poland; 3Skane
University Hospital and Lund University, Lund, Sweden; 4Concord Repatriation General
Hospital, University of Sydney, Sydney, Australia; 5IRCCS Azienda Ospedaliero-Universitaria
di Bologna, Istituto di Ematologia “Seràgnoli”, Dipartimento di Medicina Specialistica,
Diagnostica e Sperimentale Università di Bologna, Bologna, Italy; 64th Department
of Internal Medicine - Haematology, Charles University, Hospital and Faculty of Medicine,
Hradec Králové, Czechia; 7SC Ematologia, AOU Città della Salute e della Scienza di
Torino - Presidio Molinette, Torino, Italy; 8Sarah Cannon Research Institute and Tennessee
Oncology, Nashville, United States of America; 9Royal Adelaide Hospital, Adelaide,
Australia; 10John Theurer Cancer Center, Hackensack; 11Memorial Sloan Kettering Cancer
Center, New York, United States of America; 12Department of Hematology, Hôpital Necker,
Assistance Publique - Hôpitaux de Paris and Institut Imagine, Université de Paris,
INSERM UMR1183, Paris, France; 13Department of Hematology, Hospital Universitario
Infanta Leonor, Universidad Complutense, Madrid, Spain; 14Fudan University Shanghai
Cancer Center, Shanghai, China; 15Division of Hematology-Oncology, Department of Medicine,
Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea;
16University Hospitals Plymouth NHS Trust, Plymouth, United Kingdom; 17Department
of Hematology Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer
Research, Toyo, Japan; 18Ankara University School of Medicine, Ankara, Turkey; 19Hospital
Israelita Albert Einstein, Sao Paulo, Brazil; 20Kent and Canterbury Hospital, Canterbury,
United Kingdom; 21Key Laboratory of Carcinogenesis and Translational Research (Ministry
of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute
(Beijing Cancer Hospital), Beijing, China; 22Division of Hematology and Medical Oncology,
Oregon Health & Science University, Portland, United States of America; 23The Research
Institute of the McGill University Health Centre, McGill University, Montreal, Canada;
24Maria Sklodowska-Curie National Research Institute of Oncology, Warszawa, Poland;
25Janssen Research & Development, Raritan, United States of America; 26Institut Curie
comprehensive cancer center, Paris, France; Hospitalier Universitaire de Nantes, Centre
de Recherche en Cancérologie et Immunologie Nantes Angers, INSERM, Université de Nantes,
Nantes, France; 27Department of Medicine III, Klinikum der Universität München, LMU,
Munich, Germany
Background: Elderly patients (pts) with mantle-cell lymphoma (MCL) are unsuitable
for intensive chemotherapy or transplantation due to excessive toxicities. Single-agent
ibrutinib (Ibr), a first-in-class, oral Bruton’s tyrosine kinase inhibitor (BTKi),
has transformed the care of pts with relapsed or refractory MCL with durable activity.
Aims: We conducted a phase III trial (SHINE; NCT01776840) to evaluate combining Ibr
with a standard chemoimmunotherapy (BR) and R maintenance in older pts with untreated
MCL.
Methods: Pts aged ≥ 65 years, enrolled between May 2013 and November 2014 from 183
sites across all geographical regions, were stratified by simplified MIPI score (low
vs intermediate vs high risk) and were randomized 1:1 to Ibr (560 mg orally daily)
or placebo (Pbo), plus 6 cycles of B (90 mg/m2) and R (375 mg/m2). Pts who achieved
an objective response received R maintenance, administered every 8 weeks for up to
12 additional doses in both arms. Ibr and Pbo were administered until disease progression
or unacceptable toxicity. The primary endpoint was progression-free survival (PFS)
assessed by the investigators.
Results: A total of 523 pts were randomized to Ibr + BR (n = 261) or Pbo + BR (n =
262). Median age was 71 years (range, 65–87), 65.6% of pts had low/intermediate simplified
MIPI, and 8.6% had blastoid/pleiomorphic histology. At the primary analysis, median
follow up was 84.7 months. The primary endpoint was met as PFS was significantly improved
in the Ibr arm vs the Pbo arm (hazard ratio, 0.75; one-sided P = 0.011) (Figure).
Median PFS was 80.6 months with Ibr in combination with BR and R maintenance, a 50%
improvement over Pbo in combination with BR and R maintenance (median PFS of 52.9
months). The complete response rate was 65.5% in the Ibr arm and 57.6% in the Pbo
arm (P = 0.0567). There was no difference in overall survival between treatment arms
(P = 0.648). Time to next treatment was longer in the Ibr arm compared with the Pbo
arm (P < 0.001). Fifty-two (19.9%) and 106 (40.5%) pts received subsequent anti-lymphoma
therapy in the Ibr and Pbo arms, respectively; 41/106 (38.7%) received a second-line
BTKi in the Pbo arm.
Rates of grade 3 or 4 treatment-emergent adverse events were 81.5% and 77.3% in the
Ibr and Pbo arms, respectively. Of adverse events of clinical interest for BTKis,
atrial fibrillation was reported in 13.9% and 6.5% of pts in the Ibr and Pbo arms,
respectively. Rates of major hemorrhage, hypertension, arthralgia, and secondary primary
malignancies were similar in both arms. Quality of life was also similar in both arms.
Image:
Summary/Conclusion: This phase III study in untreated MCL demonstrated that Ibr combined
with BR and R maintenance significantly improved PFS compared with standard chemoimmunotherapy,
with a median PFS of 6.7 years. The safety profile was consistent with the known profiles
of the individual drugs.
S210: CAR T-CELLS ASSOCIATED ACUTE TOXICITY IN B-CELL NON-HODGKIN LYMPHOMA: REAL-WORLD
STUDY FROM THE DESCAR-T REGISTRY
P. SESQUES1,*, R. DI BLASI2, S. LE GOUILL3, G. CARTRON4, G. MANSON5, D. BEAUVAIS6,
F. LE BRAS7, F. X. GROS8, S. CHOQUET9, P. BORIES10, M. T. RUBIO11, R. O. CASASNOVAS12,
L. BOUNAIX13, M. MOHTY14, M. JORIS15, J. ABRAHAM16, C. CASTILLA LLORENTE17, M. LOSCHI18,
S. CARRAS19, A. CHAUCHET20, L. DRIEU LA ROCHELLE21, J. ZERBIT22, O. HERMINE23, S.
GUIDEZ24, T. GASTINNE25, J. J. TUDESQ26, P. FOGARTY27, F. BROUSSAIS28, F. MORSCHHAUSER29,
R. HOUOT30, C. THIEBLEMONT31, E. BACHY1
1CHU LYON SUD, LYON; 2Hematology, Assistance Publique Hôpitaux de Paris; 3Hematology,
INSTITUT CURIE, PARIS; 4HEMATOLOGY, Département d’Hématologie clinique, CHU de Montpellier,
MONTPELLIER; 5CHU de Rennes, RENNES; 6Hématologie clinique, CHU de Lille, LILLE; 7Lymphoid
Malignancies, CRETEIL; 8Hématologie Clinique et Thérapie cellulaire, MERIGNAC; 9APHP
la Pitié Salpêtriere, PARIS; 10Hematology Laboratory, Onco-occitanie Network, TOULOUSE;
11Service Hématologie, NANCY; 12Department of Hematology, DIJON; 13Department of Hematology,
Clermont-Ferrand University Hospital, CLEMRONT FERRAND; 14Hôpital St Antoine, PARIS;
15Hematology department, CHU Amiens, AMIENS; 16CHU DE LIMOGES - HOPITAL DUPUYTREN,
LIMOGES; 17GUSTAVE ROUSSY CANCER CAMPUS GRAND PARIS, VILLEJUIF; 18CHU NICE, NICE;
19CHU DE GRENOBLE, GRENOBLE; 20CHU JEAN MINJOZ, BESANCON; 21CHU BRETONNEAU, TOURS;
22APHP HOPITAL COCHIN; 23APHP HOPITAL NECKER, PARIS; 24CHU DE POITIERS - HOPITAL DE
LA MILETRIE, POITIERS; 25Nantes University Hospital Clinical Hematology, NANTES; 26Hematology
Department, Montpellier University Hospital, Univ Montpellier, MONTPELLIER; 27biostatistics;
28LYSARC, LYON; 29Maladies du Sang CHRU Lille, LILLE; 30CHU de Rennes, Université
de Rennes, INSERM U1236, EFS, RENNES; 31Saint Louis Hospital, PARIS, France
Background: Cytokine release syndrome (CRS) and immune effector cell–associated neurotoxicity
syndrome (ICANS) are common immune-related toxicities associated with chimeric antigen
receptor (CAR)–T-cell therapy. Their clinical manifestations can be severe and potentially
life threatening.
Aims: Here, we report the French experience of CAR-T toxicity, we specifically addressed
the modifiable risk factors for toxicity and we assessed management toxicity in real-world
population.
Methods: We conducted a study in a large cohort of R/R aggressive B-cell lymphoma
patients (pts) treated with commercial products. All data were collected through the
French DESCAR-T registry. CRS/ICANS were graded prospectively (ASTCT grade scale).
Regarding previously validated predictive score of CRS and ICANS in the literature,
EASIX score (LDHxCreatinine/Platelets), modified-EASIX score (m-EASIX: CRPxCreatinine/Platelets)
and simplified-EASIX score (s-EASIX: LDH/Platelets) (Pennisi et al., 2021) were assessed
in our cohort.
Results: A total of 705 pts were included for the analysis of toxicity with a median
follow-up of 12 months (range: 0.2-39). Notably, 74 pts (11%) pts had an ECOG/PS ≥
2 before lymphodepletion. CRS of any grade occurred in 587 pts (83.3%) including 62
(10.5%) with grade 3 or higher (grade 3+). The median time from the infusion to the
onset of CRS was 2 days (range: 0-34); the median time to resolution was 6 days (range:
1-30). ICANS of any grade occurred in 289 pts (41%) including 78 (27%) with grade
3+. The median time from the infusion to the onset was 6 days (range: 0-379); the
median time to resolution was 7 days (range: 1-100). Most patients (94.5%) with ICANS
had previously experienced CRS. Only 89 pts (29.9%) required admission to intensive
care unit. Tocilizumab was the most common treatment for toxicity (411 pts, 68%) and
272 pts (45%) received corticosteroids. Interestingly, 38% of patients with CRS grade
1, 53 % with grade 2 and 65% with grade 3+ developed ICANS subsequently.
In multivariate analysis, the parameters which retained statistical significance to
predict any CRS were Axi-cel ([OR] = 2.79, 95% CI 1.75-4.45; p <0.0001) and elevated
LDH (>upper limit normal; [OR] = 1.69, 95% CI 1.03-1.63; p = 0.03). Bulky mass > 5 cm
([OR] = 2.95, 95% CI 1.60-5.45; p = 0.0005), age <65 years ([OR] = 0.42, 95% CI 0.20-0.86;
p = 0.01) and higher s-EASIX score ([OR] = 1.38, 95% CI 1.15-1.67; p = 0.0005) increased
the risk of developing grade 3+ CRS. Parameters which retained statistical significance
to predict any ICANS were Axi-cel ([OR] = 3.95, 95% CI 2.57-6.06; p <0.0001), ECOG
≥2 ([OR] = 2.10, 95% CI 1.26-3.51; p= 0.004), age ≥65 ([OR] = 3.17, 95% CI 2.10-4.79;
p <0.0001) and higher s-EASIX score ([OR] = 1.20, 95% CI 1.03-1.40; p= 0.01). Axi-cel
([OR] = 13.1, 95% CI 3.98-43.3; p <0.001), ECOG ≥2 ([OR] = 0.04, 95% CI 1.007-4.38;
p= 0.04) and higher s-EASIX score ([OR] = 1.35, 95% CI 1.08-1.67; p= 0.0067) increased
the risk of developing grade 3+ ICANS. No survival impact (OS and PFS) was detected
for pts who experimented any grade or grade 3+ of ICANS or CRS (Figure 1). Importantly,
cumulative dose and duration of tocilizumab or corticosteroids were not significantly
associated with adverse PFS or OS.
Image:
Summary/Conclusion: In this large study, robust biological and clinical predictive
factors of CRS and ICANS were identified. A propensity-score matched comparison of
axi-cel and tisa-cel for toxicities will be performed for the EHA congress. Neither
grade 3+ CRS, ICANS, nor Tocilizumab or corticosteroids use for CAR-T toxicity had
negative impact on survival.
S211: CLINICAL AND PATIENT-REPORTED OUTCOMES IN A PHASE 3 STUDY OF AXICABTAGENE CILOLEUCEL
(AXI-CEL) VS STANDARD-OF-CARE IN ELDERLY PATIENTS WITH RELAPSED/REFRACTORY LARGE B-CELL
LYMPHOMA (ZUMA-7)
A. Sureda1,*, J. Westin2, F. L. Locke3, M. Dickinson4, A. Ghobadi5, M. Elsawy6, T.
van Meerten7, D. B. Miklos8, M. Ulrickson9, M.-A. Perales10, U. Farooq11, L. Wannesson12,
L. Leslie13, M. J. Kersten14, C. A. Jacobson15, J. M. Pagel16, G. Wulf17, P. Johnston18,
A. P. Rapoport19, L. I. Gordon20, Y. Yang21, A. Peng21, L. Du21, J. T. Snider21, J.
Shah21, M. Schupp21, P. Cheng21, C. To21, O. O. Oluwole22
1Hematology Department, Institut Català d’Oncologia-Hospitalet, IDIBELL, Universitat
de Barcelona, Barcelona, Spain; 2The University of Texas MD Anderson Cancer Center,
Houston; 3Moffitt Cancer Center, Tampa, United States of America; 4Peter MacCallum
Cancer Centre, Royal Melbourne Hospital and the University of Melbourne, Melbourne,
Victoria, Australia; 5Washington University School of Medicine, St Louis, United States
of America; 6Division of Hematology, Department of Medicine, Dalhousie University,
Halifax, Nova Scotia, Canada; 7University Medical Center Groningen, on behalf of HOVON/LLPC,
Groningen, Netherlands; 8Stanford University School of Medicine, Stanford; 9Banner
MD Anderson Cancer Center, Gilbert; 10Memorial Sloan-Kettering Cancer Center, New
York; 11University of Iowa, Iowa City, United States of America; 12Istituto Oncologico
della Svizzera Italiana, Bellinzona, Switzerland; 13John Theurer Cancer Center, Hackensack,
United States of America; 14Amsterdam UMC, University of Amsterdam, on behalf of HOVON/LLPC,
Amsterdam, Netherlands; 15Dana-Farber Cancer Institute, Boston; 16Swedish Cancer Institute,
Seattle, United States of America; 17University Medicine Göttingen, Göttingen, Germany;
18Mayo Clinic, Rochester; 19University of Maryland School of Medicine and Marlene
and Stewart Greenebaum Comprehensive Cancer Center, Baltimore; 20Northwestern University
Feinberg School of Medicine, Chicago; 21Kite, a Gilead Company, Santa Monica; 22Vanderbilt
University Cancer Center, Nashville, United States of America
Background: The median age at large B-cell lymphoma (LBCL) diagnosis is 66 years,
and older patients with relapsed/refractory (R/R) LBCL are at risk of inferior outcomes,
increased toxicity, and inability to tolerate second-line standard-of-care (SOC) treatment
(Di M, et al. Oncologist. 2021). Further, second-line SOC treatment is often associated
with poor health-related quality of life (Lin V, et al. J Clin Oncol. 2020;38:e20070).
In the global Phase 3, randomized ZUMA-7 study, axi-cel, an autologous anti-CD19 CAR
T-cell therapy, significantly improved event-free survival (EFS; hazard ratio [HR],
0.398, P<0.0001; median 8.3 vs 2 months, respectively) compared with second-line SOC
in R/R LBCL (Locke FL, et al. N Engl J Med. 2022;386:640-654).
Aims: Here we report results of a planned subgroup analysis of the ZUMA-7 study assessing
outcomes, including patient-reported outcomes (PROs), of second-line axi-cel vs SOC
in patients aged ≥65 years.
Methods: Patients with ECOG performance status 0-1 and R/R LBCL ≤12 months after first-line
chemoimmunotherapy were randomized 1:1 to axi-cel or SOC (2-3 cycles of platinum-based
chemoimmunotherapy; patients with partial or complete response [CR] proceeded to high-dose
therapy with autologous stem cell transplantation). PRO instruments, including the
EORTC QLQ-C30 (Global Health and Physical Functioning) and the EQ-5D-5L visual analog
scale (VAS), were administered at baseline (prior to treatment), Day 50, Day 100,
Day 150, and Month 9, then every 3 months up to 24 months or time of EFS event, whichever
occurred first. The quality-of-life analysis set included all patients who had a baseline
PRO and ≥1 completed measure at Day 50, 100, or 150. A clinically meaningful change
was defined as 10 points for each EORTC QLQ-C30 score and 7 points for EQ-5D-5L VAS
score.
Results: The data cutoff for this analysis was March 18, 2021 and included 51 axi-cel
and 58 SOC patients with median ages of 70 years (range, 65-80) and 69 years (range,
65-81), respectively. At baseline, more axi-cel vs SOC patients had high-risk features,
including second-line age-adjusted International Prognostic Index 2-3 (53% vs 31%)
and elevated lactate dehydrogenase (61% vs 41%). EFS was superior with axi-cel vs
SOC (HR, 0.276, P<0.0001), with higher CR rates (75% vs 33%). Grade ≥3 treatment-emergent
adverse events (AEs) occurred in 94% and 82% of axi-cel and SOC patients, respectively,
and Grade 5 treatment-related AEs occurred in 0 and 1 patient. In the quality-of-life
analysis set comprising 46 axi-cel and 42 SOC patients, there were statistically significant
and clinically meaningful differences in mean change of scores from baseline at Day
100 favoring axi-cel for EORTC QLQ-C30 Global Health (P<0.0001) and Physical Functioning
(P=0.0019) and EQ-5D-5L VAS (P<0.0001). For all 3 domains, scores also favored (P<0.05)
axi-cel over SOC at Day 150. The mean estimated scores numerically returned to or
exceeded baseline scores earlier in the axi-cel arm (by Day 150) but never equaled
or exceeded baseline scores by Month 15 in the SOC arm.
Summary/Conclusion: Axi-cel demonstrated superiority over second-line SOC in patients
≥65 years with significantly improved EFS and a manageable safety profile. Axi-cel
also showed meaningful improvement in quality of life over SOC, measured by multiple
validated PRO instruments, with suggested faster recovery to pretreatment quality
of life. The superior clinical outcomes and patient experience with axi-cel over SOC
should help inform treatment choices in second-line R/R LBCL for patients ≥65 years.
S212: PHASE I STUDY OF YTB323, A CHIMERIC ANTIGEN RECEPTOR (CAR)-T CELL THERAPY MANUFACTURED
USING T-CHARGE™, IN PATIENTS WITH RELAPSED/REFRACTORY DIFFUSE LARGE B-CELL LYMPHOMA
M. Dickinson1,*, M. Kwon2, J. Briones3, U. Jäger4, K. Kato5, E. Bachy6, D. Blaise7,
N. Boissel8, N. Shah9, M Frigault10, P. Riedell11, L. Shune12, T. Teshima13, X. Zhu14,
E. Orlando14, L. Yi15, J. Davis16, E. Bleickardt16, I. Flinn17, P. Barba18, P. Barba18
1Peter MacCallum Cancer Centre and Royal Melbourne Hospital and the University of
Melbourne, Melbourne, Australia; 2Department of Hematology, Hospital General Universitario
Gregorio Marañón, Institute of Health Research Gregorio Marañón, Madrid; 3Hematology
Department, Hospital Santa Creu i Sant Pau, Barcelona, Spain; 4Clinical Division of
Hematology and Hemostaseology, Department of Medicine I, Vienna General Hospital –
Medical University of Vienna, Vienna, Austria; 5Department of Hematology, Oncology
and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan; 6Hematology
department, Hospices Civils de Lyon and Université Claude Bernard Lyon; 1, Lyon; 7Département
d’Hématologie, Programme de Transplantation et de Thérapie Cellulaire, Centre de Recherche
en Cancérologie de Marseille, Aix-Marseille University, Institut Paoli Calmettes,
Marseille; 8Hematology Adolescent and Young Adult Unit, Saint-Louis Hospital, Paris,
France; 9Medical College of Wisconsin, Milwaukee; 10Massachusetts General Hospital,
Massachusetts; 11University of Chicago, Chicago; 12Division of Hematologic Malignancies
and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, United
States of America; 13Department of Hematology, Hokkaido University Hospital, Sapporo,
Japan; 14Novartis Institutes for BioMedical Research; 15Novartis Pharmaceuticals Corporation,
Cambridge; 16Novartis Pharmaceuticals Corporation, East Hanover; 17Sarah Cannon Research
Institute and Tennessee Oncology, Nashville, United States of America; 18Hematology
Department, Hospital Universitari Vall d’Hebrón, Universitat Autònoma de Barcelona,
Barcelona, Spain
Background: CD19-directed CAR-T cell therapies (tx) demonstrate efficacy in patients
(pts) with B-cell malignancies. However, despite positive outcomes with currently
approved CAR-T cell tx, many patients fail to respond or relapse after initial response.
YTB323 is an autologous CD19-directed CAR-T cell tx generated by the innovative T-Charge™
platform, which demonstrates high potency, preserves T-cell stemness in the final
product, and takes <2 d to manufacture.
Aims: T-Charge™ is expected to prolong CAR-T cell persistence and yield higher response
rates and durability.
Methods: These data from the ongoing Phase I, multicenter, dose-escalation study (NCT03960840)
focus on safety and efficacy of YTB323 in adults with relapsed/refractory diffuse
large B-cell lymphoma (r/r DLBCL). Eligible pts had measurable disease at enrollment,
ECOG 0-1, and r/r DLBCL after ≥2 lines of prior tx. Pts received single-dose YTB323
at targeted dose levels (DL) 1 (2.5×106 CAR+ cells), DL2 (12.5×106 CAR+ cells), DL3
(25×106 CAR+ cells), or DL4 (40×106 CAR+ cells). Results presented here focus on DL1
and DL2. The primary endpoints are to identify a recommended dose for subsequent trials
and characterize the safety and dose-limiting toxicities. Secondary endpoints include
overall response rate (ORR) by local investigator assessment and cellular kinetics.
Results: As of August 20, 2021, 20 pts with r/r DLBCL received YTB323: 4 at DL1 and
16 at DL2. Median age was 65 y, 65% received 2 prior lines of tx, and 35% had prior
SCT. The median follow-up for pts in DL1 and DL2 was 20 and 7 months, respectively.
Of 20 pts evaluable for safety, all experienced at least 1 adverse event (AE) of any
Grade (Gr) and 80% at least 1 AE Gr ≥3. One pt (25%) experienced Gr 1 cytokine release
syndrome (CRS) at DL1, and 5 (31%) pts experienced CRS at DL2, including 4 (25%) Gr
1 or 2 and 1 (6%) Gr 4 (Lee et al, 2014), which met protocol defined DLT criteria
at DL2. Tocilizumab and corticosteroids were used for CRS management in 4 (80%) and
2 (40%) pts at DL2, respectively. Median time to CRS onset was 10 d (range, 1-17 d).
Five pts (25%) had neurological events (NE), of which the majority were Gr 1. Median
time to onset and resolution of NE was 7 and 12 d, respectively. Five pts died on
trial (beyond 30 d); 3 pts died due to disease progression (1 at DL1, 2 at DL2), and
1 pt each due to sepsis (1 at DL1) and intestinal hemorrhage (1 at DL2). At DL1, ORR
and complete response (CR) rate were both 75%. At DL2, ORR and CR rates were 81% and
75%, respectively. Of the 15 pts who received YTB323 at DL2 at least 3 mo prior to
the data cutoff, CR rate at mo 3 was 73% (95% CI, 44.9%-92.2%) (Figure). Median time
to peak YTB323 expansion was ~16 d and coincided with cytokine peak. YTB323 expansion
(Cmax and AUC0-28d) with 12.5×106 CAR+ cells (DL2) was comparable to a median tisagenlecleucel
dose of 312×106 CAR+ cells in pts with DLBCL, a 25-fold lower median dose (Awasthi
R, et al. 2020). Limited data indicate that CAR expression was detectable by flow
cytometry for at least 9 mo at DL2. T-Charge™ allowed preservation of CD4 and CD8
naive/stem memory T cells in the final product according to flow cytometry. Bulk RNAseq
analysis demonstrated that YTB323 retained a naive stem-like gene signature.
Image:
Summary/Conclusion: YTB323 is a potent new CAR-T cell tx with distinct cellular kinetics,
encouraging early efficacy results across DL1 and DL2, and a manageable safety profile.
Updated results will be presented at the meeting along with a recommended dose for
subsequent trials.
S213: EFFICACY AND SAFETY OF HUMANIZED VERSUS MURINIZED CD19 AND CD22 CAR-T CELL COCKTAIL
THERAPY FOR REFRACTORY/RELAPSED B-CELL LYMPHOMA
L. Huang1 2, J. Li2, J. Yang2, X. Zhang2, M. Zhang2, J. He2, G. Zhang2, Y. Su2, W.
Li2, H. Wang2, P. Lu1 2,*
1Tsinghua University School of Medicine-Lu Daopei Institute of Hematology, Beijing;
2Hebei Yanda Lu Daopei Hospital,., langfang, China
Background: CD19-targeted chimeric antigen receptor (CAR) T-cell therapy has demonstrated
about 50~60% of response rate in relapsed or refractory (R/R) B-cell non-Hodgkin lymphoma.
However, antigen-escape relapse has emerged as a major challenge for long-term disease
control after CD19-targeted therapies. In our previous study, we described a dual-targeted
of CD19 and CD22 CAR-T cell cocktail therapy, whichyielded 93.3% of complete remission
(CR) rate in R/R B-cell acute lymphoblastic leukemia and could reduce the risk of
CD19-negative relapse.
Aims: A phase I/II clinical trial (NCT05206071) was designed to explore the safety
and efficacy of CAR19/22 T-cell cocktail therapy for patients with R/R aggressive
B-cell lymphoma.
Methods: Eligible R/R aggressive B-cell lymphoma patients were enrolled from July
2020 toDecember 2021. All patients received fludarabine 30 mg/m2/d and cyclophosphamide
250 mg/m2/d for 3 consecutive days (day −5 to day −3) followed by cocktail CAR-T cell
infusion. CAR-T-cell-related cytokine release syndrome (CRS) was graded to assess
the safety. PET-CT was performed on day 28 and 3rd month to evaluate the response
rate.
Results: A total of 26 eligible patients received CAR19/22 cocktail infusion with
a median age of 46 years old (4-75) and a median of 3 prior lines of therapies (2-5
lines). Of 26 patients, 4 were Burkitt lymphoma, 2 were follicular lymphoma (grade
3a), and the rest were diffuse large B-cell lymphoma. Besides, 7 patients were diagnosed
with double-hit lymphoma and 4 patients were relapsed after previous CD19 CAR-T cell
therapy.
Fourteen patients received the murinized CAR-T cells at a median dose of 2.18×106/kg
(1-3×106/kg) CAR19 and 1.75×106/kg (1-3×106/kg) CAR22, while 12 patients received
the does of the humanized CAR-T cells consisting of 2×106/kg CAR19 and 5×105/kg CAR22.
At the day 28 assessment, 12/12 (100%) patients received the humanized CAR-T cells
achieved overall response, including 9/12 (75%) CR and 3/12 (25%) partial remission
(PR), while the overall response rate (ORR) and CR rate of the murinized group was
85.71% (12/14) and 42.86% (6/14) respectively. Among of them, all 4 Burkitt lymphoma
patients in both groups achieved CR. In 3 months evaluation, 7/10 (70%,2 cases received
infusion for less than 3 months) patients in the humanized group maintained in CR,
which was higher than the murinized group with CR rate of 5/14 (35.71%). (Figure 1A-B).
At a median follow-up of 291 days (range, 31 to 544), patients in the humanized group
had a favorable progression-free survival (PFS) than those in the murinized (1-year
PFS of 67.9% vs. 26.8%), although there was no statistical significance (p=0.08) due
to the limited number of patients enrolled (Figure 1E-F). Achieving CR on day 28 (HR:
0.21, 95% CI: 0.06-0.72; P=0.012) and maintaining CR till the 3rd month (HR: 0.18,
95% CI: 0.06-0.59; P=0.004) were found as independent prognostic factors associated
with favorable PFS. Maintaining CR till the 3rd month (HR: 0.10, 95% CI: 0.01-0.61;
P=0.012) and non-double hit (HR: 0.11, 95% CI: 0.02-0.76, P=0.024) predicted a longer
overall survival (OS) (Figure 1G-L). Despite impressive CR rates wereachieved, majority
of patients had mild CRS (grade 1-2) in both group. Only 2patients experienced grade
3 CRS, and 2 patients developed grade 3 neurotoxicity.
Image:
Summary/Conclusion: This clinical trial demonstrates promising efficacy and safety
of CD19/CD22 CAR-T cocktail therapy for R/R aggressive B-cell lymphoma, and patients
in the humanized group showed better results than those with murinized, although this
not a randomized trial.
S214: PHASE 1/2 STUDY OF ANBAL-CEL, NOVEL ANTI-CD19 CAR-T THERAPY WITH DUAL SILENCING
OF PD-1 AND TIGIT IN RELAPSED OR REFRACTORY LARGE B CELL LYMPHOMA
W. S. Kim1,*, S. J. Kim1, S. E. Yoon1, J. R. Kim2
1Hematology and Oncology, Samsung Medical Center, Seoul; 2Clinical Development, Curocell
Therapeutics, DaeJeon, South Korea
Background: Anbal-cel is a novel 2nd generation autologous CD19 CAR-T cell therapy
which has been knock-downed for PD-1 and TIGIT using OVIS platform. Anbal-cel demonstrated
the eradication of CD19 positive tumor cells in vitro and in vivo better than conventional
CD19 CAR-T cells. The knock-down of PD-1 and TIGIT at CD19 CAR-T cells exerts the
superior T-cell functionality by delaying the exhaustion of CAR-T cells.
Aims: This phase 1 dose escalation part (NCT04836507) was to evaluate the safety (DLT,
MTD), PK and preliminary efficacy (objective response rate and duration of response)
in patients with r/r LBCL. Anbal-cel was manufactured at GMP facility with fresh leukapheresis
product.
Methods: Patient was administered as a single intravenous dose at dose level 1 (2x105
cells/kg), dose level 2 (7x105 cells/kg) or dose level 3 (2x106 cells/kg). Lymphodepletion
with cyclophosphamide (500mg/m2) and fludarabine (30mg/m2) was performed for 3 days
prior to Anbal-cel infusion.
Results: As of Jan 17 2022, 9 patients with r/r DLBCL were infused with Anbal-cel;
4pts at DL1, 3pts at DL2 and 2pts at DL2. Median age was 54 (range 26-71); all patients
received 2 or more prior lines of therapy and 44% (4/9) received ≥4 prior line of
treatment before the study. 78% (7/9) patients were refractory to their last treatment.
67% (6/9) of patients were at IPI 3-4 and 44% (4/9) of patients had bulky disease.
No patient experienced DLT during the study. Of the 9 patients, 5 (56%) experienced
CRS; 4 (44%) were grade 1 or 2 and one patient experienced grade 3 CRS. Median time
to onset of CRS was 7 days (range, 1-16) with median duration of 4 days (range, 0-18).
One patient dosed at DL3 experienced grade 2 ICANS, time to onset of ICANS was 7 days
and lasted for 13 days. This patient had prior CNS involvement history before the
study. Most commonly reported grade 3/4 AEs were neutrophil count decrease (6/9, 67%),
anemia (5/9, 56%), thrombocytopenia (2/9, 22%), platelet count decrease (2/9, 22%)
and no infection was reported. Complete response rate (CRR) was 78% and complete responses
were observed at the lowest dose level and from patients expressing less than 10%
CD19 at IHC; 3 complete responses (CR) at DL1, 2 CRs at DL2 & DL3 respectively. Dose-dependent
CRC01 expansion was observed; median Tmax was 15.4, 15.8, 11.0 days at DL1, DL2 &
DL3 each; median Cmax was 18,003, 30,103, 53,688 copies/ug gDNA at DL1, DL2 & DL3
each; median AUC0-28day was 679,125, 1,110,108, 2,852,235 copies/ug gDNA at DL1, DL2
& DL3 respectively.
Summary/Conclusion: Anbal-cel demonstrated promising efficacy and tolerable safety
profile in this dose escalation study. Based on this phase 1 study, phase 2 patient
enrollment will be commenced in Mar 2022 to evaluate the response rate, duration of
response of CRC01 as well as safety. In addition, various biomarker studies are planned
to investigate the differential mode of action of Anbal-cel during phase 2 study.
S215: GEMSTONE-201: PRE-PLANNED PRIMARY ANALYSIS OF A MULTICENTER, SINGLE-ARM, PHASE
2 STUDY OF SUGEMALIMAB IN PATIENTS WITH RELAPSED OR REFRACTORY EXTRANODAL NATURAL
KILLER/T CELL LYMPHOMA (R/R ENKTL)
H. Huang1,*, R. Tao2, Y. Yang3, H. Cen4, H. Zhou5, Y. Guo6, L Zou7, J. Cao8, Y. Huang9,
J. Jin10, L. Zhang11, H. Yang12, X. Xing13, H. Zhang14, Y. Liu15, K. Ding16, X. Zhu17,
T. Fang17, H. Dai17, Q. Qi17, J. Yang17
1Sun Yat-sen University Cancer Center, Guangzhou; 2Xinhua Hospital, Shanghai Jiao
Tong University School of Medicine, Shanghai; 3Fujian Cancer Hospital & Fujian Medical
University Cancer Hospital, fuzhou; 4Guangxi Cancer Hospital and of Guangxi Medical
University Affiliated Cancer Hospital, nanning; 5Hunan Cancer Hospital, changsha;
6Shanghai East Hospital, Tongji University School of Medicine, Shanghai; 7State Key
Laboratory, Cancer Center, West China Hospital of Sichuan University, chengdu; 8Fudan
University Shanghai Cancer Center, Shanghai; 9The Affiliated Cancer Hospital of Guiyang
Medical University, guiyang; 10First Affiliated Hospital, College of Medicine, Zhejiang
University, hangzhou; 11Union Hospital, Tongji Medical College, Huazhong University
of Science and Technology, wuhan; 12Zhejiang Cancer Hospital, hangzhou; 13Liaoning
Cancer Hospital & Institute, Shenyang; 14Tianjin Medical University Cancer Institute
& Hospital, tianjin; 15Henan Provincial Cancer Hospital, zhengzhou; 16Anhui Provincial
Cancer Hospital, Hefei; 17CStone Pharmaceuticals (Su Zhou) Co., Ltd., Shanghai, China
Background: R/R ENKTL is a rare and aggressive type of non-Hodgkin’s lymphoma. Responses
to chemotherapy after failure of prior asparaginase-based regimen were not durable
with a median OS of < 7 months (mos) and 1-year OS rate of < 20% (Lim et al, Ann Oncol
2017; Bellei et al, Haematologica 2018). The only targeted therapy approved in China
for R/R peripheral T cell lymphoma (ENKTL included) showed an ORR of 18.8% and a CR
rate of 6.3% (Shi et al, Ann Oncol 2015).
Aims: Here, we present the primary analysis from GEMSTONE-201, the largest registrational
study reported to date to evaluate an anti-PD-L1 mAb in R/R ENKTL. Sugemalimab (Suge)
received breakthrough therapy designation from US FDA in 2021 for adult R/R ENKTL
patients (pts) based on preliminary data of this study.
Methods: Pts with ECOG PS of 0 or 1 and histologically confirmed ENKTL who failed
prior asparaginase-based regimen were enrolled. Pts accepted suge at 1200 mg intravenously
every 3 weeks, for up to 24 mos until progression, death, or withdrawal from study.
The primary endpoint was ORR (CR+PR) assessed by independent radiological review committee
(IRRC) per Lugano 2014 criteria. Key secondary endpoints included investigator-assessed
ORR, CR and PR rate, DoR assessed by IRRC and investigators, and safety.
Results: As of the data cutoff date, 10 November 2021, 80 pts were enrolled and treated
(median follow-up of 13.4 mos). Median age was 48 years (range 29-74); 64% were males;
74% had ECOG PS of 1 at baseline; 68% had stage IV disease; about half (49%) received
≥ 2 lines of prior systemic therapy. The median duration of treatment was 5.2 mos
(range 0.7-37.4); 23 pts remained on treatment. Among the 78 evaluable pts as per
IRRC, ORR was 46.2% (95% CI: 34.8%, 57.8%); 29 (37.2%) pts achieved CR; median DoR
was not reached (NR); 12-mo DoR rate was 86%. Investigator’s assessments in 79 evaluable
pts were consistent with IRRC results, i.e., ORR of 45.6% (95% CI: 34.3%, 57.2%),
24 (30.4%) pts with CR, and median DoR of NR. The 1- and 2-year OS rates were 68.6%
and 54.6%, respectively; median OS was NR (range 0.9-37.2+ mos).
Of all pts, 96% (n = 77) had at least one AE. The most common AEs were pyrexia and
WBC decreased (n = 24 each, 30%). Grade ≥ 3 AEs occurred in 31 (38.8%) pts. Suge-related
AEs occurred in 61 (76%) pts and were mostly (60%) Grade 1 or 2. The most common immune-related
AE assessed by sponsor was hypothyroidism (n = 13, 16%). SAEs occurred in 18 (23%)
pts; 5 (6%) pts had suge-related SAEs which had all been resolved (1 with sequelae).
Fatal AEs occurred in 5 (6%) pts and none were suge-related as assessed by investigators.
Summary/Conclusion: Suge has demonstrated deep and durable anti-tumor activity in
R/R ENKTL pts, with a high CR rate and a promising OS benefit trend comparing to historical
data. Suge had a well-tolerated safety profile and no new safety signals were detected.
Primary analysis indicates that suge could provide a new treatment option to R/R ENKTL
pts.
S216: CLINICAL ACTIVITY OF CC-99282, A CEREBLON E3 LIGASE MODULATOR (CELMOD) AGENT,
IN PATIENTS (PTS) WITH RELAPSED/REFRACTORY NON-HODGKIN LYMPHOMA (R/R NHL) – RESULTS
FROM A PHASE 1, OPEN-LABEL STUDY
J.-M. Michot1,*, J. C. Chavez2, C Carpio3, S. Ferrari4, T. A. Feldman5, D. Morillo6,
J. Kuruvilla7, A. Pinto8, V. Ribrag1, E Bachy9, T. J. Buchholz10, S. Carrancio11,
W.-C. Chou12, C. Guarinos13, F. Wu14, S. Li15, P. Patah16, M. Pourdehnad17, L. Nastoupil18
1Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy
Institute of Cancer, Villejuif, France; 2Department of Malignant Hematology, Moffitt
Cancer Center, University of South Florida, Tampa, United States of America; 3Department
of Hematology, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall
d’Hebron, Autonomous University of Barcelona (UAB), Barcelona, Spain; 4Hematology
and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy; 5Lymphoma
Division, John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, United
States of America; 6Department of Hematology, University Hospital Fundación Jiménez
Díaz, Madrid, Spain; 7Division of Medical Oncology and Hematology, Princess Margaret
Cancer Centre, Toronto, Canada; 8Hematology-Oncology & Stem Cell Transplantation Unit,
National Cancer Institute, Fondazione G. Pascale, IRCCS, Naples, Italy; 9Department
of Hematology, Hospices Civils de Lyon, Lyon, France; 10Early Clinical Development,
Oncology, Bristol Myers Squibb, San Francisco; 11Oncogenesis (ONC) Thematic Research
Center (TRC), Bristol Myers Squibb, San Diego; 12Early Development Predictive Sciences,
Bristol Myers Squibb, Cambridge, United States of America; 13ONC TRC-Celgene Institute
for Translational Research Europe (CITRE), Bristol Myers Squibb, Seville, Spain; 14Clinical
Pharmacology & Pharmacometrics, Bristol Myers Squibb, Summit; 15Global Biometric Sciences,
Bristol Myers Squibb, Berkley Heights; 16Early Clinical Development, Bristol Myers
Squibb, Lawrenceville; 17Early Clinical Development, Hematology/Oncology and Cell
Therapy, Bristol Myers Squibb, San Francisco; 18Department of Lymphoma & Myeloma,
The University of Texas MD Anderson Cancer Center, Houston, United States of America
Background: CC-99282 is a novel, oral small molecule CELMoD® agent that co-opts cereblon
to induce targeted degradation of Ikaros/Aiolos, transcription factors critical for
development of B-cell malignancies. Compared with immunomodulatory drugs (IMiD®) and
other CELMoD agents, CC-99282 had similar immunostimulatory effects and 10- to 100-fold
stronger antiproliferative and apoptotic activity in preclinical models of diffuse
large B-cell lymphoma (DLBCL).
Aims: To evaluate the safety, tolerability, maximum tolerated dose, pharmacokinetics
(PK), and preliminary efficacy of CC-99282 in pts with R/R NHL.
Methods: CC-99282-NHL-001 (NCT03930953) is a 2-part multicenter first-in-human study
comprising dose escalation of CC-99282 monotherapy (part A) and expansion with or
without combination partners (part B). Part A includes pts with R/R DLBCL or follicular
lymphoma (FL) who had progressed after ≥2 lines of therapy, including IMiD/CELMoD
agents and chimeric antigen receptor T-cell (CAR-T) therapy, and pts with R/R DLBCL
who received ≥1 line of standard therapy and are unfit for transplant. Pts receive
oral CC-99282 0.2, 0.4, 0.6, or 0.8 mg once daily on 3 intermittent dosing schedules
of 28-day cycles, with ≥3 pts per dosing cohort.
Results: As of Oct 6, 2021, 50 pts were treated in part A (38 DLBCL, 12 FL; median
age 66y; 58% male); 17 pts (34%) were ongoing and 26 pts (52%) discontinued due to
progressive disease. Grade 3/4 CC-99282–related adverse events were reported in 32
pts (64%); the most common were neutropenia (29 pts [58%]), thrombocytopenia (4 pts
[8%]), and anemia (4 pts [8%]). Correlation analysis showed that grade 4 neutropenia
in the first month was more strongly associated with number of prior lines of alkylating
agent and anti-CD20 therapy than with CC-99282 dose level or schedule. Neutropenia
was manageable with CC-99282 dose modifications and granulocyte colony–stimulating
factors. All dose-limiting toxicities were hematologic. The maximal recommended doses
for schedules of interest were 0.6 mg on 7/14 days and 0.4 mg on 14/28 days. For doses
≥0.4 mg on schedules of interest, the overall response rate was 42% (15/36 evaluable
pts; 6 FL, 9 DLBCL), with complete responses in 6 pts and partial responses in 9 pts.
Responders included pts previously treated with CAR-T therapy and/or IMiD/CELMoD agents.
Responses were durable, with median durations of 239 days (range 48–587; median follow-up
[mFUP] 247 days [range 21–690]) and 112 days (range 63–414; mFUP 121 days [range 22–464])
for 7/14- and 14/28-day schedules, respectively. CC-99282 was absorbed rapidly and
had a prolonged terminal half-life (median ~50 hours at doses ≥0.4 mg), with considerable
distribution into the peripheral compartment. Increase in plasma CC-99282 and degradation
of Ikaros/Aiolos in peripheral T cells occurred in a dose-dependent manner, and maximum
degradation (>90%) occurred by day 4 of treatment at doses ≥0.4 mg. Within 2 weeks
of initiating CC-99282, peripheral T-cell subsets showed a significant shift toward
a more activated phenotype (P<0.05). Analysis of mutant circulating tumor DNA levels
showed rapid reductions that correlated with response to CC-99282, suggesting strong
tumor cell–intrinsic activity.
Summary/Conclusion: CC-99282 monotherapy showed a manageable safety profile and demonstrated
promising efficacy in heavily pretreated pts with R/R NHL. PK and pharmacodynamic
data were consistent with robust and rapid CC-99282 antitumor activity. This study
is ongoing, with patients actively being enrolled in the monotherapy and CC-99282+rituximab
combination expansion cohorts.
S217: PRELIMINARY ANALYSIS OF THE PHASE II STUDY USING TOLINAPANT (ASTX660) MONOTHERAPY
IN 98 PERIPHERAL T-CELL LYMPHOMA AND 51 CUTANEOUS T-CELL LYMPHOMA SUBJECTS WITH RELAPSED
REFRACTORY DISEASE.
J.-M. Michot1, A. Mehta2, F. Samaniego3, E. Bachy4, P. L. Zinzani5, A. Prica6, G.
P. Colins7, V Ribrag1,*, N. Wagner-Johnston8, D. El-Sharkawi9, O. A. O’Connor10, R.
Wilcox11, L. Wang12, L. Wilson12, M. Sims13, J. A. Taylor12, H. N. Keer12, F. Foss14
1Department of Hematology, Institut Gustave Roussy, Villejuif, France; 2University
of Alabama at Birmingham, Birmingham; 3The University of Texas MD Anderson Cancer
Center, Houston, United States of America; 4Hematology Department, Lyon Sud Hospital,
Lyon, France; 5Institute of Hematology “L. e A. Seràgnoli”, Bologna, Italy; 6Princess
Margaret Cancer Centre, Toronto, Ontario, Canada; 7Oxford University Hospitals NHS
Foundation Trust, Oxford, United Kingdom; 8Johns Hopkins University, The Sidney Kimmel
Comprehensive Cancer Center, Baltimore, United States of America; 9The Royal Marsden
NHS Foundation Trust, Surrey, United Kingdom; 10University of Virginia, Charlottesville;
11University of Michigan Medical School, Ann Arbor; 12Astex Pharmaceuticals, Inc.,
Pleasanton, United States of America; 13Astex Pharmaceuticals, Cambridge, United Kingdom;
14Yale Cancer Center, New Haven, United States of America
Background: There are limited treatment options for patients with Peripheral T-cell
lymphoma (PTCL) and Cutaneous T-cell lymphoma (CTCL), especially when front line therapy
has failed. Tolinapant (ASTX660) is a novel oral non-peptidomimetic, small-molecule
antagonist of cellular/X-linked inhibitors of apoptosis proteins (cIAP1/2 and XIAP),
which also induces necroptosis in T-cell lymphoma models (Ferrari et al., Blood Advances,
2021). Tolinapant is being evaluated in a first-in-human ongoing Phase I/II study
in subjects with advanced solid tumors and lymphoma (ClinicalTrials.gov NCT02503423).
Phase I results were previously reported (Mita et al. Clin Cancer Res, 2020) and the
recommended phase 2 dosing (RP2D) was established. Initial results for Phase II were
previously reported at EHA 2019 (Mehta et al., EHA 2019, # PS1073).
Aims: Here we report the preliminary efficacy and safety analysis for the Phase 2
PTCL and CTCL cohorts.
Methods: This is a single-arm open-label Phase II study. To be eligible, subjects
must have evidence of documented progressive disease and received at least two prior
systemic therapies. Subjects received treatment with tolinapant at the RP2D 180 mg/day
on Days 1 to 7, and 15 to 22 in a 28-day cycle. The primary endpoint is best overall
response rate (ORR) as assessed by the investigator according to either the Lugano
criteria (PTCL) or Global Assessment (CTCL). Adverse events (AEs) are assessed per
CTCAE v4.03. The efficacy data set is based on subjects who had tumor evaluation at
baseline and at least 1 post-treatment tumor evaluation visit, unless they died or
stopped treatment earlier due to clinical progression or toxicity. The safety data
set is based on all subjects that received at least one dose of tolinapant.
Results: As of the data cut of January 5, 2022, there were 98 subjects with PTCL and
51 subjects with CTCL that received drug and 98 and 50 subjects that were evaluable
respectively. The study is currently closed to enrollment with a minimum of 6 months
follow-up on all subjects at the time of the data cut. Subject characteristics: median
(range) age PTCL 62.5 (27, 82) and CTCL 62 (24,87), median number of previous therapies
PTCL 3 (0-8) and CTCL 6 (1-10). Among all subjects, the most common related AEs of
any grade (≥ 15%) were: lipase elevation (35%), amylase elevation (25%), rash (combined
listings) (24%), ALT elevation (15%), and AST elevation (15%). Related AEs ≥ Grade
3 (≥ 5%) were: lipase elevation (15%), rash (combined listings) (9%), and amylase
elevation (7%). Pancreatitis was identified in 2 subjects (1%) (both Grade 4). There
were no related ≥ Grade 3 AEs for diarrhea, nausea or vomiting; for related Grade
2 AEs there was a 5% incidence of diarrhea and 1% incidence of nausea/vomiting.
The ORR for PTCL is 22%, including 9 complete responses (CRs) and 12 partial responses
(PRs). The ORR in CTCL is 26% including 2 CRs and 11 PRs. The median durability of
response for PTCL is 133 (Q1-Q3; 69 - 280) days and for CTCL is 148 (Q1-Q3; 103 -
294) days. Pharmacodynamic and correlative analysis is ongoing with preliminary analysis
suggesting an immunomodulatory antitumoral effect of tolinapant (Ferrari et al., Blood
Advances, 2021).
Summary/Conclusion: In this Phase II study, the novel oral agent tolinapant has shown
meaningful clinical activity against PTCL and CTCL with a manageable safety profile.
These results support the continued development of tolinapant for the treatment of
R/R PTCL and CTCL. A drug combination study using tolinapant in R/R PTCL is being
developed.
S218: A PHASE I/II STUDY OF GOLIDOCITINIB, A SELECTIVE JAK1 INHIBITOR, IN REFRACTORY
OR RELAPSED PERIPHERAL T CELL LYMPHOMA
W.-S. Kim1,*, D.-H. Yoon2, Y. Song3, H. Yang4, J. Cao5, D. Ji5, Y. Koh6, H. Jing7,
H.-S. Eom8, J.-Y. Kwak9, W.-S. Lee10, J.-S. Lee11, H.-J. Shin12, J. Jin13, M. Wang14,
J. Li14, X. Huang14, X. Deng14, Z. Yang14, J. Zhu3
1Department of Hematology and Oncology, Samsung Medical Center; 2Department of Oncology
- Hematologic Cancer & BMT center, Asan Medical Center, Seoul, South Korea; 3Department
of Lymphoma, Peking University Cancer Hospital, Beijing; 4Department of Lymphoma,
Zhejiang Cancer Hospital, Hangzhou; 5Department of Oncology, Fudan University Shanghai
Cancer Center, Shanghai, China; 6Department of Hemato-Oncology Center, Seoul National
University Hospital, Seoul, South Korea; 7Department of Hematology, Peking University
Third Hospital, Beijing, China; 8Center for Hematologic Malignancy, National Cancer
Center, Goyang; 9Department of Hemato-Oncology Center, Chonbuk National University
Hospital, Jeonju; 10Department of Hemato-Oncology Center, Inje University Busan Paik
Hospital, Busan; 11Department of Hematology & Medical Oncology, Seoul National University
Bundang Hospital, Seongnam; 12Department of Hemato-Oncology Center, Pusan National
University Hospital, Busan, South Korea; 13Department of Hematology, The First Affiliated
Hospital of Zhejiang University, Hangzhou; 14Clinical Development, Dizal Pharmaceutical,
Shanghai, China
Background: Peripheral T cell lymphoma (PTCL) is a group of heterogeneous T cell lymphomas.
Patients who relapse from or are refractory to 1st line therapy face dismal prognosis.
The response rates to commonly used 2nd line agents such as histone deacetylase inhibitors
are below 30%. Immunotherapies, such as anti-PD1 antibodies, may induce hyperprogression
in certain PTCL subtypes. Hence, r/r PTCL patients urgently need better therapies.
Aims: Preclinical data shows JAK/STAT pathway may mediate the pathogenesis of PTCL,
making it a promising target. Golidocitinib (DZD4205) is an orally available, potent,
JAK1 specific inhibitor, demonstrating profound anti-tumor activities in T lymphoma
cells in vitro and tumor xenograft in vivo. Here we report the preliminary data from
an ongoing phase I/II study (NCT04105010) of Golidocitinib in r/r PTCL.
Methods: The study included two parts: Part A (dose escalation) and Part B (dose expansion).
In Part A, patients with r/r PTCL were enrolled and received Golidocitinib at different
doses (150 mg or 250 mg, QD) to determine the recommended phase II dose (RP2D). Evaluation
of safety and efficacy were performed by investigators per CTCAE and Lugano criteria,
respectively. Part B is a single-arm, pivotal study, where patients with r/r PTCL
will receive Golidocitinib at the RP2D till disease progression or intolerance.
Results: As of May 31, 2021, a total of 51 patients enrolled in Part A and received
Golidocitinib at 150 mg (n = 35) or 250 mg (n = 16). Patient characteristics: median
age (range): 61.0 years (29-79); median prior systemic therapies (range): 2 lines
(1-8). Ten patients (19.6%) had undergone hematopoietic stem cell transplantation.
Fifteen patients (29.4%) had bone marrow involvement at baseline. Histological subtypes
included PTCL-NOS (41.2%), AITL (39.2%), ALCL ALK- (7.8%), NKTCL (7.8%), and MEITL
(3.9%).
At the data cut-off (DCO), 49 patients completed at least one post-treatment Lugano
assessment, of whom 21 (42.9%) achieved tumor response, including 11 complete responses
(CRs, 22.4%) and 10 partial responses (20.4%). Tumor response was observed in various
subtypes, including AITL (13/20), PTCL-NOS (5/19), ALCL ALK- (2/4) and NKTCL (1/4).
At the DCO, the median duration of response (DoR) was not reached, and the longest
DoR was > 14 months.
Forty-eight patients (94.1%) experienced treatment emergent adverse events (TEAEs),
of whom 30 (58.8%) experienced ≥ grade 3 TEAEs. Per investigators’ assessment, 20
patients (39.2%) experienced ≥ grade 3 TEAEs possibly related to the drug. The most
common (≥ 10%) ≥ grade 3 TEAEs were neutropenia (29.4%), thrombocytopenia (15.7%)
and pneumonia (11.8%). The majority of TEAEs were reversible or clinically manageable
with dose modifications.
Image:
Summary/Conclusion: Golidocitinib shows good safety and promising anti-tumor efficacy
in r/r PTCL, indicating its potential as a therapeutic option for this unmet medical
need.
S219: FIRST CLINICAL STUDY OF THE ANTI-SIGNAL REGULATORY PROTEIN-ALPHA (SIRPΑ) ANTIBODY
CC-95251 COMBINED WITH RITUXIMAB IN PATIENTS WITH RELAPSED/REFRACTORY (R/R) NON-HODGKIN
LYMPHOMA (NHL)
P. Strati1,*, E. Hawkes2, N. Ghosh3, J. Tuscano4, Q. Chu5, M. A. Anderson6, A. Patel7,
M. R. Burgess7, K. Hege7, S. Chhagan7, S. Boyanapalli7, T. Day7, F. Shen7, A. Mehta8
1The University of Texas MD Anderson Cancer Center, Houston, United States of America;
2Austin Health–Austin Hospital, Heidelberg, Australia; 3Levine Cancer Institute, Charlotte;
4University of California, Davis, Sacramento, United States of America; 5Cross Cancer
Institute, Edmonton, Canada; 6Peter MacCallum Cancer Centre, Melbourne, Australia;
7Bristol Myers Squibb, Princeton; 8University of Alabama at Birmingham, Birmingham,
United States of America
Background: CD47 is a cell-surface ligand overexpressed in various malignancies that
binds to SIRPα on effector macrophages to promote tumor cell evasion of phagocytosis.
Blockade of this CD47–SIRPα interaction enhances phagocytosis mediated by tumor-targeting
antibodies, such as rituximab (ritux). Agents targeting CD47 combined with ritux have
demonstrated promising clinical activity in R/R NHL; however, the broad expression
of CD47 potentially acts as an antigen sink, reducing anti-tumor activity, and leads
to on-target, off-tumor toxicities, including hemolytic anemia. CC-95251 is a novel,
fully human immunoglobulin G1 antibody (Ab) that binds to SIRPα on macrophages to
potently block the CD47–SIRPα interaction.
Aims: The primary objectives of this multicenter, open-label, phase 1, dose-escalation
and dose-expansion study were to evaluate the safety and tolerability of escalating
doses of CC-95251 + ritux and to define the maximum tolerated dose (MTD) and/or recommended
phase 2 dose for the combination in patients (pts) with CD20+ R/R NHL (NCT03783403).
Here, we report interim results.
Methods: Pts were treated in 28-day cycles (C) with CC-95251 administered intravenously
at doses of 3, 10, or 20 mg/kg every week (QW) and with ritux 375 mg/m2 on days (D)
1, 8, 15, and 22 of C1, D1 of C2–5, and D1 of every other cycle C6–24 until disease
progression or unacceptable toxicity.
Results: As of 5 August 2021, 17 pts had received ≥ 1 dose of CC-95251 + ritux. Median
age was 67 (range 30–84) years. Pts had received a median of 3 (range 1–7) prior systemic
therapies, with 100% of pts having a prior history of anti-CD20 Ab exposure. Enrolled
tumor types included R/R diffuse large B-cell lymphoma in 13 (77%) pts, follicular
lymphoma in 2 (12%), and mantle cell lymphoma and marginal zone lymphoma in 1 (6%)
pt each. Twelve (71%) pts had disease refractory to any prior line of therapy (LOT),
including 10 (59%) refractory to any anti-CD20 Ab-containing regimen, and 9 (53%)
to their last LOT. Pts received a median of 4 (range 1–14) cycles of CC-95251. Median
duration of treatment was 16.4 (range 2.1–53.1) weeks. There was 1 CC-95251 dose reduction,
and 6 (35%) pts experienced ≥ 1 treatment-emergent adverse event (TEAE) leading to
CC-95251 dose interruption. MTD has not been reached. Neutropenia and infection were
the most common TEAEs for any grade (71% and 59% respectively) and grade ≥ 3 (59%
and 29% respectively). Treatment-related adverse events of grade ≥ 3 included neutropenia
in 9 pts (53%) and infection in 2 pts (12%); no treatment-related anemia or deaths
were reported. Overall response rate in the efficacy evaluable population was 56%
(9/16), with 4 (25%) pts achieving a complete response (CR). Of 9 pts refractory to
any prior anti-CD20 Ab-containing regimen, 2 pts achieved a CR and 2 pts had stable
disease. Median time to response was 7.9 weeks. Duration of response ranged 7.4–28.1
weeks with a CR ongoing in 1 pt. In this dose-escalation study, CC-95251 demonstrated
dose-proportional increases in exposure at doses > 3 mg/kg QW and full receptor occupancy
at doses ≥ 3 mg/kg.
Summary/Conclusion: CC-95251 + ritux demonstrated a manageable safety profile and
promising efficacy in pts with heavily pretreated CD20+ R/R NHL. The study is currently
enrolling in the dose-expansion phase.
S220: GLOFITAMAB INDUCES DURABLE COMPLETE REMISSIONS AND HAS FAVORABLE SAFETY IN PATIENTS
WITH RELAPSED/REFRACTORY DIFFUSE LARGE B-CELL LYMPHOMA AND ≥2 PRIOR THERAPIES: PIVOTAL
PHASE II EXPANSION RESULTS
M. Dickinson1,*, C. Carlo-Stella2, F. Morschhauser3, E. Bachy4, P. Corradini5, G.
Iacoboni6, C. Khan7, T. Wróbel8, F. Offner9, M. Trněný10, S.-J. Wu11, G. Cartron12,
M. Hertzberg13, A. Sureda14, D. Perez-Callejo15, L. Lundberg15, J. Relf16, E. Clark16,
K. Humphrey16, M. Hutchings17
1Peter MacCallum Cancer Centre, Royal Melbourne Hospital and The University of Melbourne,
Melbourne, VIC, Australia; 2Humanitas University and IRCCS Humanitas Research Hospital,
Milan, Italy; 3Hôpital Claude Huriez and CHU de Lille, Lille; 4Centre Hospitalier
Lyon-Sud, Lyon, France; 5Università degli Studi di Milano and Fondazione Istituti
di Ricovero e Cura a Carattere Scientifico (IRCSS) Istituto Nazionale dei Tumori,
Milan, Italy; 6Vall d’Hebron University Hospital, Barcelona, Spain; 7Allegheny Health
Network, Pittsburgh, PA, United States of America; 8Uniwersytet Medyczny we Wrocławiu,
Wroclaw, Poland; 9Universitair Ziekenhuis Gent, Ghent, Belgium; 10Charles University
Hospital, Prague, Czechia; 11National Taiwan University Hospital, Taipei, Taiwan;
12CHU de Montpellier, Montpellier, France; 13Prince of Wales Hospital and University
of New South Wales, Sydney, NSW, Australia; 14Institut Català d’Oncologia Hospital,
Barcelona, Spain; 15F. Hoffmann-La Roche Ltd, Basel, Switzerland; 16Roche Products
Ltd, Welwyn Garden City, United Kingdom; 17Rigshospitalet, Copenhagen, Denmark
Background: Glofitamab is a T-cell engaging bispecific antibody (Ab) with a novel
2:1 configuration that confers bivalency for CD20 (B cells) and monovalency for CD3
(T cells). In a Phase I/II study (NCT03075696), escalating glofitamab doses were highly
active and well tolerated in patients with relapsed/refractory (R/R) B-cell lymphomas,
with obinutuzumab pretreatment (Gpt) and Cycle (C) 1 step-up dosing providing effective
cytokine release syndrome (CRS) mitigation.
Aims: We present pivotal Phase II expansion results in patients with R/R diffuse large
B-cell lymphoma (DLBCL) and ≥2 prior therapies.
Methods: All patients had DLBCL (DLBCL not otherwise specified [NOS], high-grade B-cell
lymphoma, primary mediastinal large B-cell lymphoma, or transformed follicular lymphoma)
and had received ≥2 prior regimens, including ≥1 anti-(a) CD20 Ab and ≥1 anthracycline.
Intravenous (IV) Gpt (1000mg) was given 7 days before the first glofitamab dose. IV
glofitamab was then given as step-up doses on Day (D) 1 (2.5mg) and D8 (10mg) of C1
and at the target dose (30mg) on D1 of C2–12 (21-day cycles). The primary endpoint
was complete response (CR) rate assessed by Independent Review Committee (IRC) using
Lugano 2014 criteria. CRS was assessed using ASTCT criteria. All patients provided
informed consent.
Results: As of September 14, 2021, 107 patients had received ≥1 dose of study treatment
(median age: 66 years [21–90]; Ann Arbor stage III–IV disease: 74%; IPI score ≥3:
54%; DLBCL NOS: 74%). Median prior therapies was 3 (2–7); 59% had ≥3 prior therapies
and 35% had received prior CAR T-cells (CAR-Ts). Most patients were refractory to
a prior aCD20 Ab-containing regimen (85%) and to their most recent regimen (85%).
Many were refractory to their initial therapy (59%) and to prior CAR-Ts (32%). After
a median follow-up of 9 months (0.1–16), overall response and CR rates by IRC were
50.0% and 35.2%, respectively. CR rates were consistent in patients with and without
prior CAR-Ts (32% vs 37%). Median time to CR was 42 days (95% CI: 41–48). The majority
of CRs (33/38; 87%) were ongoing at data cut. An estimated 84% of complete responders
and 61% of responders remained in response at 9 months. At data cut, the projected
12-month overall survival rate was 48%, and 92% of complete responders were alive.
These results are consistent with earlier Phase I data in 100 patients treated with
target glofitamab doses ≥10mg (CR rate: 34%; estimated 20-month CR rate in complete
responders: 72%). CRS occurred in 68% of patients, was primarily associated with the
initial doses, and was mostly Grade (Gr) 1 (51%) or Gr 2 (12%); Gr 3 (3%) and Gr 4
(2%) events were uncommon. All but 2 CRS events were resolved at data cut. Glofitamab-related
neurologic adverse events (AEs) potentially consistent with immune effector cell-associated
neurotoxicity syndrome (ICANS) occurred in 3 patients (all Gr 1–2). No glofitamab-related
Gr 5 (fatal) AEs occurred. Glofitamab-related AEs leading to discontinuation were
uncommon (3 patients, 3%).
Summary/Conclusion: Fixed-duration glofitamab induces durable complete remissions
and has favorable safety in patients with R/R DLBCL and ≥2 prior therapies, including
those with prior exposure to CAR-Ts. Glofitamab is a promising new therapy for patients
with heavily pretreated and/or highly refractory DLBCL.
S221: BORTEZOMIB TO R-DHAP COMPARED TO R-DHAP IN RELAPSED/REFRACTORY DIFFUSE LARGE
B-CELL LYMPHOMA ELIGIBLE TO STEM CELL TRANPLANTATION: FINAL RESULTS OF PHASE II RANDOMIZED
FIL-VERAL12
A. Chiappella1,*, M. Balzarotti2, B Botto3, A. Castiglione4, F. Cavallo5, S. V. Usai6,
M. Zanni7, C. Califano8, F. Re9, C. Ghiggi10, A. Olivieri11, P. Corradini12, A. M.
Liberati13, S. Volpetti14, M. G. Michieli15, A Tucci16, G. Gaidano17, M. Tani18, F.
Ciambelli19, G. Musuraca20, D. Vallisa21, F. Merli22, A. L. Molinari23, A. Tafuri24,
V. R. Zilioli25, D. Marino26, C. Stelitano27, S. A. Pileri28, G. Ciccone4, U. Vitolo29
1Hematology, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milano; 2Hematology,
Humanitas Cancer Center, Istituto Clinico Humanitas, Rozzano; 3Hematology, AOU Città
della Salute e della Scienza; 4Unit of Clinical Epidemiology, AOU Città della Salute
e della Scienza e CPO Piemonte; 5Hematology, Department of Molecular Biotechnologies
and Health Sciences, Università degli studi e AOU Città della Salute e della Scienza,
Torino; 6Hematology, Ospedale Businco, Cagliari; 7Hematology, AO SS. Antonio e Biagio
e Cesare Arrigo, Alessandria; 8Oncology and Hematology, Presidio ospedaliero “A. Tortora”,
Pagani; 9Hematology, Ospedale Policlinico San Martino S.S.R.L. - IRCCS per l Oncologia,
Parma; 10Hematology, Ospedale Policlinico San Martino S.S.R.L. - IRCCS per l Oncologia,
Genova; 11Hematology, AOU Ospedali Riuniti, Ancona; 12Chair of Hematology, Università
degli Studi di Milano e Hematology, Fondazione IRCCS, Istituto Nazionale dei Tumori,
Milano; 13Oncology and Hematology, AO. S. Maria, Terni; 14Hematology, Azienda Sanitaria
Universitaria Friuli Centrale, Udine; 15Oncology and Hematology, IRCCS Centro di Riferimento
Oncologico, Aviano; 16Hematology, ASST Spedali Civili, Brescia; 17Hematology and Department
of Translational Medicine, Università del Piemonte Orientale e AOU Maggiore della
Carità, Novara; 18Hematology, Ospedale delle Croci, Ravenna; 19Oncology, AO S. Antonio
Abate, Gallarate; 20Hematology, IRCCS Istituto Romagnolo per lo studio dei Tumori
“Dino Amadori” – IRST S.R.L., Meldola; 21Hematology, Ospedale Guglielmo da Saliceto,
Piacenza; 22Hematology, Azienda Unitа Sanitaria Locale-IRCCS - Arcispedale Santa Maria
Nuova, Reggio Emilia; 23Hematology, Ospedale degli Infermi, Rimini; 24Hematology,
AO Sant Andrea, Roma; 25Hematology, ASST Grande Ospedale Metropolitano Niguarda, Milano;
26Oncology, IRCCS Istituto Oncologico Veneto, Padova; 27Hematology, Grande Ospedale
Metropolitano Bianchi Melacrino Morelli, Reggio Calabria; 28Haematopathology, Istituto
Europeo di Oncologia IRCCS, Milano; 29Hematology, Candiolo Cancer Institute, FPO-IRCCS,
Candiolo, Italy
Background: The standard treatment in patients with diffuse large-B cell lymphoma
(DLBCL) relapsed/refractory (R/R) after first line therapy is a cisplatin-containing
regimen followed by consolidation with high-dose chemotherapy and autologous stem
cell transplantation (auto-SCT) in responsive ones after induction. Bortezomib had
proven activity in aggressive lymphomas.
Aims: On these bases, the Fondazione Italiana Linfomi designed the FIL-VERAL12 trial,
aimed at evaluating whether the addition of bortezomib to rituximab-cisplatin-cytarabine-dexametasone
(BR-DHAP) increases complete response rate (CR, according to Lugano 2007 criteria)
prior auto-SCT compared to standard R-DHAP.
Methods: FIL-VERAL12 was a prospective, multicenter, two-arm randomized phase II trial
(NCT01805557).The primary study endpoint was CR after 4 courses of R-DHAP or BR-DHAP,
assuming a 30% CR for the standard arm and a 50% CR in experimental arm. Inclusion
criteria were: patients aged 18-65 years eligible to high-dose therapy, with R/R DLBCL
after first line chemoimmunotherapy. Patients were stratified by relapsed or refractory
and randomized 1:1 to receive: a) the standard salvage therapy R-DHAP every 28 days
for 4 cycles and b) subcutaneous 1.5 mg/ms bortezomib on days 1 and 4 of each 4-week
cycle in addition to the same regimen.
Results: From January 2013 to November 2018, 114 patients were screened, and 107 patients
that fulfilled the inclusion criteria were enrolled into the trial and randomized
to receive R-DHAP or BR-DHAP (54 patients in R-DHAP, 53 in BR-DHAP). Principal clinical
characteristics were: median age 57 years (IQR: 48;62); stage III/IV 83 patients (78%);
International Prognostic Index (IPI) risk >2 37 (35%). All patients received rituximab
and anthracycline-based regimens as first line treatment. Considering the time at
relapse, 53 patients (50%) were registered as relapsed (median time at relapse 10.8
months, IQR: 6.9;20.9) and 54 (50%) as refractory (0.9 months, IQR 0.52;1.3). 52 (49%)
patients completed the planned 4 cycles of therapy; 55 did not, due to progressive
disease in 42, adverse events or clinician decision in 13. At the end of the 4 courses,
the pre-auto-SCT response was: CR 29 (27%), Partial Response (PR) 9 (17%); according
to arm of randomization, the primary end point was not met, with CR 28% for R-DHAP
and 26% for BR-DHAP (p-value 0.563). Fifty patients (44%) performed a consolidation
with SCT, 24 in R-DHAP arm and 26 in BR-DHAP arm; auto-SCT was performed in 39 patients
and allo-SCT in 11. The addition of bortezomib to standard R-DHAP did not impact the
mobilization, with a median number of CD34+ collected of 6.43 x 10^6 cells CD34/kg
(IQR: 4.40;9.11) in R-DHAP and 6.78 x 10^6 cells CD34/kg (IQR: 5.00;9.68) in BR-DHAP.
Sixty patients died: 49 (82%) due to lymphoma, 1 due to toxicity, 3 due to transplant
related mortality, 7 due to other causes. The incidence of adverse events was similar
in the two arms, with grade 3-4 haematological toxicities in 96 patients (90%), g3-4
infection in 5 (5%), g3-4 neurotoxicity in 4 (4%). At a median follow-up of 50 months,
2-years PFS was 29% (95%CI: 19.94;41.83) and 41% (27.67;53.84) for R-DHAP and BR-DHAP,
respectively; HR 0.65 (0.41;1.02) p 0.062; 2-years OS was 43% (28.98;56.30) and 52%
(37.80;64.56) for R-DHAP and BR-DHAP, respectively; HR 0.74 (0.44;1.23) p 0.244.
Summary/Conclusion: In the FIL-VERAL12 phase II randomized trial, the addition of
bortezomib to R-DHAP did not improve the CR rate pre-auto-SCT of R/R DLBCL patients
eligible to high-dose chemotherapy plus SCT; a numerically higher 2-year PFS rate
was observed in BR-DHAP arm.
S222: MATURE T AND NK CELL LYMPHOMAS CLASSIFIED ACCORDING TO 2016 WHO CLASSIFICATION.
A REPORT OF THE INTERNATIONAL PROSPECTIVE T-CELL PROJECT 2.0.
M. Federico1,*, Y. Stepanishyna2 3, T. Skrypets2 4, C. S. Chiattone5, M. H. Prince6,
A. Pavlovsky7, A. Lymboussakis1, M. Manni1, M. Civallero1, C. A. de Souza8, E. A.
Hawkes9, L. Fiad10, R. Nair11, I. Kriachok12, F. Hitz13, O. Kostina14, C. Tomuleasa15,
A. Guarini16, S. Luminari1 17
1Surgical, Medical and Dental Department of Morphological Sciences related to Transplant,
Oncology and Regenerative Medicine, University of Modena and Reggio Emilia; 2TCP2
Trial Office, TCP2 Trial Office, Modena, Italy; 3Department of Bone Marrow Transplant,
National Cancer Institute, Kiev, Ukraine; 4Clinical and Experimental Medicine (CEM),
University of Modena and Reggio Emilia, Modena, Italy; 5Santa Casa Medical School,
Santa Casa Medical School, Sao Paulo, Brazil; 6Sir Peter MacCallum Department of Oncology,
University of Melbourne, Melbourne, Australia; 7Haematology, Fundaleu, Buenos Aires,
Argentina; 8Universidade de Campinas (UNICAMP), Universidade de Campinas (UNICAMP),
Campinas, Brazil; 9School of Public Health and Preventive Medicine Monash University
and Olivia Newton John Cancer Research Institute, Austin Health, Melbourne, Australia;
10Hospital Italiano La Plata, Hospital Italiano La Plata, Buenos Aires, Argentina;
11TATA Medical Center, TATA Medical Center, Kolkata, India; 12Department of Oncohematology,
National Cancer Institute, Kiev, Ukraine; 13Department of Oncology/Haematology, Cantonal
Hospital, St Gallen, Switzerland; 14North Estonia Medical Centre, North Estonia Medical
Centre, Tallinn, Estonia; 15Ion Chiricuta Oncology Institute, Ion Chiricuta Oncology
Institute, Cluj Napoca, Romania; 16Haematology and Cell Therapy Unit, IRCCS-Istituto
Tumori ‘Giovanni Paolo II’, Bari; 17Hematology Unit, Azienda Unità Sanitaria Locale
IRCCS, Arcispedale Santa Maria Nuova IRCCS, Reggio Emilia, Italy
Background: Mature T and NK-cell lymphomas represent a heterogeneous group of rare
lymphoid disorders arising from mature T cells of post-thymic origin. In 2018 the
International T-cell non-Hodgkin’s Lymphoma Study Group launched the T-cell Project
2.0. Here we report the distribution of cases of PTCLs registered in the study, based
on the diagnosis made locally according to the WHO 2016 classification, and some preliminary
information on treatment and outcome.
Aims: The aim of the study is a better understanding of this group of rare disorders,
capturing a real-life snapshot of the evolving landscape of T-cell lymphoma biology,
treatment strategies, and outcome.
Methods: The T-cell Project 2.0 (ClinicalTrials.gov Identifier: NCT03964480) is a
prospective, longitudinal, international, observational study of patients with PTCLs.
Results: Between October 2018 and February 2022, 901 patients with newly diagnosed
PTCL were registered by 94 active centers across 17 countries.
Distribution of cases according to different subtypes is reported in Figure 1. Overall,
the most frequent 6 subtypes account for 93% of cases, and the remaining 7% was represented
by few cases of 12 different subtypes. Of note, only 9 cases have been classified
according to entities not considered in the previous WHO 2008 classification.
With respect to clinical presentation, the median age at diagnosis was 56 years (18-93),
57.4% of the patients were male, the presence of systemic symptoms was reported in
31% of cases, 8.9% had ECOG-PS 3-4, 71% advanced disease and 35.4% bone marrow involvement.
At time of data lock, 831 cases had information on initial therapy. Overall, 731 patients
(88%) were treated with combination chemotherapy and 125 (15%) were consolidated with
high dose therapy and stem cell transplantation.
After a median follow-up of 20 months, the 2-year PFS and OS were 49% and 57%, respectively.
For patients with PTCL-NOS, AITL, ENKTL, ALCL ALK- and ALCL ALK+, a comparison in
terms of PFS was performed for those enrolled in the TCP2 and in the previous TCP1.
Interestingly, a statistically significant difference (p=0.009) was observed, with
2-year PFS of 50% and 45% for patients enrolled in the TCP2 and TCP1, respectively.
Image:
Summary/Conclusion: Regardless the difficulties linked to the COVD-19 pandemic, the
TCP2 is recruiting very well and allow us in better understanding the outcome of patients
with PTCL classified according to the 2016 WHO in the real world. Moreover, our data
show that some improvement in the curability of patients affected by PTCL is emerging.
S223: LATE CARDIOVASCULAR TOXICITY AFTER HIGH-DOSE CHEMOTHERAPY FOR LYMPHOMA: A DANISH
POPULATION-BASED STUDY
S. Husby1,*, J. Baech2, T. Trab2, J. Gørløv3, J. M. Jørgensen4, S. Gudbrandsdottir5,
M. T. Severinsen2, K. Grønbæk3, T. S. Larsen6, P. Brown3, L. H. Jakobsen2, K Kragholm7,
T. C. El-Galaly2
1Hematology, Aalborg University hospital, Copenhagen N; 2Hematology, Aalborg University
hospital, Aalborg; 3Hematology, Rigshospitalet, Copenhagen N; 4Hematology, Aarhus
University Hospital, Aarhus; 5Hematology, Zealand University Hospital, Roskilde; 6Hematology,
Odense University Hospital, Odense; 7Cardiology, Aalborg University hospital, Aalborg,
Denmark
Background: Salvage chemotherapy followed by high-dose chemotherapy (HDT) and autologous
stem cell transplantation (ASCT) are standard treatments for younger patients with
relapsed/refractory aggressive diffuse large B-cell lymphoma (DLBCL) and Hodgkin’s
lymphoma (HL) as well as for consolidation in 1st line treatment for mantle cell lymphoma
(MCL) and peripheral T-cell lymphoma (PTCL). Congestive heart failure (CHF) is a known
late complication to lymphoma therapy, but it is unclear how much HDT adds to CHF
risk. Delineating late complications to HDT/ASCT is increasingly important due to
the availability of alternatives such as CAR-T therapy.
Aims: To investigate how HDT/ASCT treatment influences the risk of developing congestive
heart failure in a population-based nationwide setting.
Methods: Data were obtained from the nationwide Danish lymphoma registry and the Danish
National Patient Register. DLBCL, MCL, HL, and PTCL patients treated with HDT between
2001 and 2017 and without CHF prior to HDT were included. Patients were compared to
two populations: 1) A matched population of individuals from the general population
without prior CHF matched on age and sex in a 1:5 ratio, and 2) A population of lymphoma
patients (HL or DLBCL) in clinical remission two years after standard 1st line treatment
without HDT. The latter group was chosen due to very low lymphoma-specific mortality.
CHF was defined by ICD-10 outpatient clinic and hospital discharge diagnosis codes.
The Aalen-Johansen estimator was used to compute the cumulative risks treating death
as competing event. This analysis was conducted for CHF and cardiovascular diseases
in general, separately (CVD; ischemic heart disease, atrial fibrillation/flutter,
ventricular fibrillation/flutter, and CHF). Crude and adjusted cause-specific hazard
ratios (HR) were obtained using Cox regression and adjustments were made for age and
sex.
Results: A total of 958 HDT patients were identified (35% DLBCL, 28% MCL, 21% PTCL,
and 16% HL) with a median follow-up of 7.9 years. Almost all patients were previously
treated with anthracycline (97.8%).
The risk of both CHF and CVD was significantly increased in patients treated with
HDT compared to a matched background population (p<0.001; Fig. 1a, 1b). The risk of
CHF was more pronounced in HDT-treated males compared to females (10-year incidence
7.8% vs 3.8%, respectively, Fig. 1c and Fig. 1d); however, the risk was significantly
increased for both sexes (p<0.001 for both).
Multivariable Cox regression adjusted for sex and age confirmed the increased risk
of CHF for HDT patients. The risk was increased both when compared to the matched
background population (adjusted HR 6.40) and to lymphoma patients not treated with
HDT (adjusted HR 2.54). This is intriguing since HDT patients are generally considered
more fit than unselected lymphoma patients. Comparison of comorbidities between the
groups is ongoing.
In the HDT group, the major risk factors for CHF were older age at the time of HDT
(HR 1.20 per decade), and male sex (HR 2.15).
Image:
Summary/Conclusion: The risk of CHF and CVD in general is significantly increased
for HDT treated patients compared to both a matched background population and patients
treated with 1st line therapy without consolidating HDT, despite likely selection
of healthy patients to HDT. Major risk factors were male sex and age. These findings
are important when considering the possible use of new therapies such as CAR-T cell
therapy which have a different safety profile.
S224: PRELIMINARY RESULTS OF A PHASE II STUDY OF ORELABRUTINIB IN COMBINATION WITH
ANTI-PD-1 MONOCLONAL ANTIBODY IN REFRACTORY OR RELAPSED PRIMARY CNS LYMPHOMA
Y. Zhang1,*, W. Wang1, D. Zhao1, Y. Zhang2, W. Zhang1, D. Zhou1
1Department of Hematology, Peking Union Medical College Hospital, Chinese Academy
of Medical Sciences & Peking Union Medical College; 2Department of Hematology, Beijing
Longfu Hospital, Beijing, China
Background: The survival outcomes of patients with relapsed/refractory (R/R) primary
central nervous system lymphoma (PCNSL) remain extremely poor and there are no approved
therapies or widely accepted “standard-of-care” approaches. Previous studies showed
that Bruton tyrosine kinase (BTK) inhibitor and anti-programmed death protein-1(PD-1)
monoclonal antibody have significant activities in R/R PCNSL, respectively. The combination
of BTK inhibitor and anti-PD-1 monoclonal antibody demonstrated synergistic effects
both in vivo and in vitro in diffuse large B cell lymphoma, but no clinical data is
currently available for PCNSL. Orelabrutinib is a new-generation BTK inhibitor with
high CSF concentration and sintilimab is a new-generation anti-PD-1 monoclonal antibody.
Aims: To evaluate the safety and efficacy of orelabrutinib combined with sintilimab
in patients with R/R PCNSL.
Methods: The prospective, multicenter, single-arm phase II study (NCT04899427) enrolled
immunocompetent adult patients with R/R PCNSL and eligible organ functions. The patients
received once daily orelabrutinib (150mg) in combination with sintilimab (200mg on
day 1 of each cycle) every 3 weeks per cycle up to two years or until disease progression,
intolerable toxicity, or death. The primary objective was the overall response rate
(ORR; defined as partial response [PR] or better) after 4 cycles. Other endpoints
mainly included 1-year progression free survival (PFS) rate, time to response (TTR)
and safety.
Results: The data cut-off date was February 25, 2022. Thirteen patients were enrolled
from March 2021 to January 2022, with a median follow-up of 7.0 (1.5-10.5) months.
Median age was 61 yr (range 48 to 71) and 6 (46.1%) were male. The median lines of
prior treatment were 2 (range 1 to 4). All the patients were high-dose methotrexate
treated, seven of those (53.8%) were refractory to the last treatment. Ten patients
completed 4 cycles of the experimental regimen while 3 patients ended of treatment
in the first 2 cycles due to disease progression. The toxicities were quite mild,
with one grade 3 adverse event (AE) of interstitial pneumonitis related pneumocystis
carinii infection. No other Grade 3-4 hematological or non-hematological AE was reported.
All patients were evaluable for response. The ORR was 61.5%, and 4 patients achieved
complete remission (CR), 1 CRu, and 3 PR. The median TTR was 6 weeks (2 to 4 cycles).
No relapses were observed in patients who achieved ORR after 4 cycles. Only 1 patient
died of disease progression (Fig. 1). The estimated median1-year PFS rate was 67.7%
(Fig. 2). Plasma and CSF sample from 4 of 13 patients were collected at 2 h after
15 days orelabrutinib administration. The median CSF concentration of orelabrutinib
was 28.7ng/ml (rang 11.8 ng/ml to 52.7 ng/ml). The median CSF/plasma free ratio was
59.8%(rang 46.09% to 86.67%).
Image:
Summary/Conclusion: The combination of orelabrutinib and sintilimab showed a high
ORR and a rapid onset of response in patients with r/r PCNSL with well-tolerated toxicities.
Although preliminary, these results supported the use of BTK inhibitor plus anti-PD-1
monoclonal antibody. More clinical data will be updated from this ongoing study.
S225: CIRCULATING TUMOR DNA IS A PROGNOSTIC BIOMARKER AT BASELINE AND IMPROVES THE
ACCURACY OF INTERIM PET IN CLASSIC HODGKIN LYMPHOMA
M. C. Pirosa1,*, A. Bruscaggin1, L. Terzi di Bergamo1, M. Salehi1, K. Pini1, V. Spina1,
S. Bocchetta1, A. Condoluci1, G. Forestieri1, D. Piffaretti1, J. Marques De Almeida1,
S. Annunziata2, F. Bergesio3, E. Borsatti4, P. Bulian5, S. Chauvie3, C. Marco6, D.
T. Martina7, G. Bernhard8, M. Kurlapski9, A. Moccia10, R. Moia11, A. Rinaldi12, M.
Rodari13, G. Romanowicz14, G. M. Sacchetti15, A. Stasia8, A. Stathis10, G. Stüssi8,
I. Zangrilli16, L. M. Larocca17, A. Pinto18, A. Santoro7, F. Cavalli1, E. Zucca10,
V. Gattei19, J. M. Zaucha9, C. Carlo-Stella7, S. Hohaus16, G. Gaidano11, L. Ceriani6,
D. Rossi1
1Division of Experimental Hematology, Institute of Oncology Research, Bellinzona,
Switzerland; 2Institute of Nuclear Medicine, Policlinico Gemelli Foundation, Catholic
University of the Sacred Heart, Rome; 3Department of Medical Physics, Santa Croce
e Carle Hospital, Cuneo; 4Nuclear Medicine, IRCCS CRO; 5Clinical and Experimental
Onco Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano,
Italy; 6Clinic of Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern
Switzerland, Bellinzona, Switzerland; 7Humanitas Cancer Center, Humanitas Clinical
and Research Center, Milan, Italy; 8Division of Hematology, Oncology Institute of
Southern Switzerland, Bellinzona, Switzerland; 9Department of Hematology and Transplantology,
Medical University of Gdańsk, Gdańsk, Poland; 10Clinic of Medical Oncology, Oncology
Institute of Southern Switzerland, Bellinzona, Switzerland; 11Division of hematology,
University of Eastern Piedmont, Novara, Italy; 12Genomics Facility, Institute of Oncology
Research, Bellinzona, Switzerland; 13Unit of Nuclear Medicine, Humanitas Clinical
and Research Center, Milan, Italy; 14Department of Nuclear Medicine, Medical University
of Gdańsk, Gdańsk, Poland; 15Nuclear Medicine, “Maggiore della Carità ” Hospital,
Novara; 16Department of diagnostic imaging, oncological radiotherapy and hematology,
Fondazione Policlinico Universitario Agostino Gemelli IRCCS; 17Division of Pathology,
Policlinico Gemelli Foundation, Catholic University of the Sacred Heart, Rome; 18Hematology‐Oncology
and Stem Cell transplantation Unit, National Cancer Institute, Fondazione “G. Pascale”
IRCCS, Naples; 19Clinical and Experimental Onco Hematology Unit, IRCCS CRO, Aviano,
Italy
Background: Classic Hodgkin lymphoma (cHL) has few pre-treatment prognostic biomarkers.
Circulating tumor DNA (ctDNA) is a metrics of tumor volume and inflammation, which
are both prognostic in cHL, it improves the accuracy of treatment response assessment,
which is an unmet need at the interim timepoint in cHL, and it allows to accurately
genotype the disease, which has technical hurdles if done of the tumor biopsy in cHL.
Aims: The study aims at addressing the following questions: i) is pre-treatment ctDNA
load a metrics that captures both tumor and inflammation extents in a single, measurable
and radiation-free test? Can the integration of ctDNA with interim PET (iPET) improve
accuracy of the sole iPET in predicting treatment outcome? Can ctDNA identify molecular
groups with phenotype- and outcome-associated signatures?
Methods: IOSI-EMA003 (NCT03280394) is a prospective, observational, multi-center study.
The study recruited adult patients with previously untreated cHL. Blood samples were
collected during staging and disease response assessment at the same time of PET scan
acquisition. PET scans were centrally and blindly reviewed by a panel of nuclear medicine
physicians. ctDNA was genotyped and quantified by phased variant-enhanced-LyV4.0 ctDNA
CAPP-seq assay. Interim molecular response (iMR) was defined as lack tumor reporters
in cfDNA collected at the interim timepoint (sensitivity 10-4).
Results: A total of 215 patients were recruited. Median follow-up was 30 months. The
full analysis dataset was divided into training (N=135) and validation (N=80) cohorts
by using a random sample procedure. In the training cohort, ctDNA load directly associated
with inflammation (B symptoms, elevated ESR), but not with disease burden: total metabolic
tumor volume (TMTV), GHSG stage. The optimized threshold of pretreatment ctDNA load
to stratify PFS was 850 hGE/mL of plasma (Figure 1A). In multivariable analysis, pretreatment
ctDNA levels remained prognostic for PFS when controlling for either GHSG stage, TMTV,
B symptoms or ESR. The validation cohort confirmed that patients with higher pretreatment
ctDNA levels had inferior PFS (Figure 1B). Among intermediate and advanced stage patients,
the PFS of patients with positive iPET was significantly inferior (Figure 1C). iMR
was achieved in 54% patients. Patients who did not achieved iMR had a lower PFS. iMR
combined with iPET had a statistically significant superior accuracy for the anticipation
of progression or relapse compared to iMR or iPET alone (Figure 1D). To identify molecular
subgroups within cHL, we focused on fragmentation profiles of cfDNA. cfDNA fragmentome
can comprehensively represent both genomic and chromatin characteristics. Pre-treatment
cfDNA fragmentation patterns were characterized by a more prominent mononucleosomal
fragments abundance in 51% of patients (mono-nucleosomal cluster), whereas 33% of
patients had a more prominent shift towards submono-nucleosomal fragment lengths (submono-nucleosomal
cluster) (Figure 1E). Patients belonging to the submono-nucleosomal cluster had different
clinical and biological nuances, including advanced stage, B-symptoms, elevated ESR,
higher TMTV and total lesion glycolysis, higher mutation load, higher circulating
immune suppressive cytokines and chemoattractants for monocytes and Th-cells. Accordingly,
patients with submono-nucleosomal cluster had a lower PFS.
Image:
Summary/Conclusion: ctDNA is a validated prognostic biomarker of cHL, can improve
the accuracy of interim response assessment and provide the bases for a molecular
classification of cHL.
S226: LOSS OF NR4A1 IS ASSOCIATED WITH HIGHER EXPRESSION OF IMMUNE CHECKPOINT COMPONENTS
AND REDUCED T CELL-MEDIATED LYMPHOMA CELL KILLING IN AGGRESSIVE LYMPHOMA
K. Pansy1,*, K. Fechter1, A. Arra2, M. Szmyra1, S. Haingartner1, A. Ramsay3, H. Greinix1,
C. Beham-Schmid4, P. Neumeister1, A. Deutsch1
1Division of Hematology, Medical University of Graz, Graz, Austria; 2Clinic for Experimental
Pedriatics, Otto-von-Guericke-Universität, Magdeburg, Germany; 3Lymphoma Immunology
research group, Kings ‘s College, London, United Kingdom; 4Institute of Pathology,
Medical University of Graz, Graz, Austria
Background: Aggressive lymphomas represent the most common type of lymphoid malignancies
with a five-year survival rate of 60%. Despite effective initial treatment, one-third
of all patients will experience a relapse, warranting more research to discover novel
therapeutic strategies. We recently detected a significant lower expression of NR4A1
that correlates with a poor lymphoma-specific survival. Furthermore, ectopic expression
of NR4A1 induces apoptosis in vitro and suppresses lymphoma growth in xenografts indicating
its tumor suppressive properties.
Aims: The aim of this study was to comprehensively study the function of Nr4a1 loss
in lymphomagenesis.
Methods: Therefore, we intercrossed the EµMyc lymphoma mouse model with the Nr4a1-/-
mouse and monitored them until the onset of disease. Furthermore, we transplanted
lymphoma cells of EµMyc Nr4a1-/- and EµMyc Nr4a1+/+ mice into immune-competent C57BL/6
mice and immune-deficient Fox Chase SCID beige mice. Furthermore, we determined the
expression levels of immune checkpoint components in biopsies of our human diffuse
large B cell lymphoma (DLBCL) cohort and correlated them to NR4A1 content. Finally,
to investigate the immune-regulatory function of Nr4a1, we performed co-culture cytotoxicity
assays using OVA257-264 peptide-pulsed EµMyc Nr4a1+/+ and EµMyc Nr4a1-/- lymphoma
cells and Ctla4+/+ and Ctla4-/- OVA targeting CD8+ T cells derived from OT-I mice
and measured T cell-mediated lymphoma cell lysis via flow cytometry, respectively.
Results: We observed that the loss of Nr4a1 leads to an accelerated lymphomagenesis
in vivo, concomitant with increased expression of immune checkpoint components of
the Pd1-Pdl1-Pdl2- and Ctla4-Cd80-Cd86-axes. Immuno-competent, but not immune-deficient
mice, transplanted with Nr4a1-deficient lymphoma cells exhibited rapid lymphoma development,
reduced survival, and upregulation of the immune checkpoint components like in the
primary model. Importantly, low NR4A1 expression correlated with high expression of
immune checkpoint components in biopsies of our human DLBCL cohort largely resembling
our mouse data. To unravel the impact of Nr4a1 on T cell-mediated lymphoma cell killing
and on the regulation of the Ctla-4-Cd80-Cd86-axis of aggressive lymphomas, we performed
co-culture cytotoxicity assays using OVA peptide-pulsed EµMyc Nr4a1+/+ and EµMyc Nr4a1-/-
lymphoma cells and Ctla4+/+ and Ctla4-/- OVA targeting CD8+ T cells. In these experiments,
we observed a massively diminished lymphoma cell killing in the EµMyc Nr4a1-/- setting,
when we used Ctla4+/+ OT-1 CD8+ T cells. Interestingly, when we used Ctla4-/- OT-1
CD8+ T cells for the cytotoxicity assays, lymphoma cell killing was enhanced in the
Nr4a1-/- setting compared to that using Ctla4+/+ OT-1 CD8+ T cells.
Image:
Summary/Conclusion: Our data suggest that Nr4a1 plays a critical role in regulating
the licensing of immune evasion in aggressive lymphomas by regulating immune checkpoint
expression. Thus, it might act as a promising target to restore anti-lymphoma immune
responses.
S227: SBNO2 IS A SPECIFIC DEPENDENCY OF STAT3-DRIVEN T-CELL MALIGNANCIES
T. Brandstoetter1,*, B. Maurer1, J. Schmoellerl2, S. Kollmann1, J. Huuhtanen3 4, R.
Grausenburger1, S. Mustjoki3 5, J. Zuber2, V. Sexl1
1Dept. of Biomedical Sciences at the Institute of Pharmacology and Toxicology, University
of Veterinary Medicine, Vienna; 2Research Institute of Molecular Pathology, Vienna,
Austria; 3Hematology Research Unit Helsinki, University of Helsinki and Helsinki University
Hospital Comprehensive Cancer Center, Helsinki; 4Department of Computer Science, Aalto
University, Espoo; 5Translational Immunology Research program, University of Helsinki,
Helsinki, Finland
Background: The transcription factor signal transducer and activator of transcription
3 (STAT3) plays important roles in regulating the survival, growth and differentiation
of hematopoietic cells, particularly within the lymphoid lineage. Recently, gain-of-function
mutations within the SH2 domain of STAT3 have been identified in patients suffering
from T-cell large granular lymphocytic leukemia (T-LGLL). The most frequently found
hyperactivating mutation represents STAT3Y640F.
Aims: Transcription factors are notoriously difficult to target, hence a better understanding
of STAT3-dependent transcriptional co-factors and their molecular targets is critical
to identify novel therapeutic approaches.
Methods: To unravel the molecular mechanisms behind STAT3Y640F-induced malignancies,
we developed a murine in vitro model that recapitulates common transcriptional features
that are found in CD8+ transformed T-LGLL cells from patients harbouring STAT3 mutations.
Gene expression analyses and chromatin occupancy profiling of mutated STAT3Y640F identified
a core set of direct transcriptional targets, which were interrogated for their functional
relevance in genome-wide CRISPR/Cas9-based loss-of-function screens.
Results: Expression of STAT3Y640F blocked differentiation of murine hematopoietic
stem and progenitor cells (mHSPC), enhanced their self-renewal capacity and increased
proliferation in comparison to wild type STAT3 expressing cells. Mechanistically,
the STAT3Y640F mutation led to increased and prolonged activation of STAT3 signalling
and decreased their dependence on cytokine stimulation. In contrast to mHSPC expressing
wild-type STAT3, the Y640F-mutated variant resulted in a strong inflammatory gene
expression signature that was characterized by overactivation of TNF alpha/Interferon
gamma pathways, and is conserved in T-LGLL patients harbouring STAT3 mutations. Through
genome-wide loss-of-function screens using CRISPR/Cas9 we identified that the transcriptional
co-regulator strawberry notch homolog 2 (SBNO2) is an essential direct transcriptional
target of STAT3Y640F in transformed mHSPC. SBNO2 was overexpressed in both, STAT3Y640F-transformed
mHSPC (versus STAT3WT) and T-LGLL patients harbouring STAT3 mutations (versus T-LGLL
patients with wild type STAT3). As seen with loss of STAT3-Y640F expression, loss
of SBNO2 also impaired proliferation of transformed mHSPC. Strikingly, global analyses
of gene dependency datasets revealed that various human T-cell malignancies that are
driven by aberrant STAT3 signalling depend on SBNO2 expression.
Summary/Conclusion: Together, our data show that the STAT3/SBNO2 axis could be a promising
therapeutic intervention site in STAT3 driven T-cell malignancies.
S228: ACTIVATED SUMOYLATION RESTRICTS MHC CLASS I ANTIGEN PRESENTATION TO CONFER IMMUNE
EVASION IN LYMPHOMA
U. M. Demel1,*, M. Böger1, S. Yousefian2, C. Grunert1, P. Hotz3, S. Haas2, S. Müller3,
M. Wirth1, M. Schick1, U. Keller1
1Charité Universitätsmedizin Berlin; 2Max Delbrück Center for Molecular Medicine,
Berlin; 3Institute of Biochemistry II, Goethe University Frankfurt, Frankfurt, Germany
Background: During the last decade, the implementation of immunotherapies has revolutionized
cancer treatment. However, despite the striking success, often only subgroups of patients
respond, and loss of the MHC-I antigen presentation machinery (APM) has been identified
as a frequent cause of primary and acquired resistance to immunotherapies. Thus, strategies
to restore these pathways may enhance the efficacy of immunotherapies. The post-translational
protein modification SUMOylation (SUMO) is a crucial regulatory mechanism in the cellular
stress response. Activated SUMOylation is regarded as a hallmark of aggressive cancers.
Notably, a selective small molecule inhibitor of SUMOylation (SUMOi), subasumstat,
is currently tested in clinical trials.
Aims: We aimed to identify the implications of activated SUMOylation for lymphoma
immune evasion, and to develop a pharmacological intervention strategy to improve
cancer immunotherapies.
Methods: A multi-OMICs approach was used to interrogate SUMO-driven immune evasion
mechanisms in lymphoma. In vitro findings were informed by in vivo studies in lymphoma
xenograft models and syngeneic murine tumor models. Single-cell RNA sequencing (CITE-seq)
was applied to decipher in vivo SUMOi-driven changes in blood and immune cell subsets.
Results: Starting from a targeted screening for SUMO-regulated immune evasion mechanisms,
we identified an evolutionarily conserved pathway of MYC-induced SUMOylation, which
attenuated the immunogenicity of lymphoma cells. While activated SUMOylation repressed
the MHC-I antigen processing and presentation machinery (APM) and enabled lymphoma
cells to evade CD8+ T-cell immunosurveillance, pharmacological inhibition of SUMOylation
restored T-cell mediated tumor killing. Importantly, SUMOi amplified the activation
of the STAT1 pathway in response to IFNg, which is one of the key regulators of cytokine-induced
MHC-I expression. Beyond this, we identified a previously unknown mechanism of SUMO-mediated
basal repression of MHC-I and established SAFB, a repressor of immune regulators,
as a SUMO-regulated mediator of MHC-I suppression that mechanistically links SUMOylation
to MHC-I repression. We thus showed that activated SUMOylation converted lymphoma
cells into a less immunogenic state that was restored by pharmacological SUMO inhibition,
enhancing the susceptibility of lymphoma cells to CD8+ T-cell mediated killing. Moreover,
we revealed that activated SUMOylation was associated with lower abundance and activity
of tumor-infiltrating T-cells in vivo, proposing SUMOi as a novel therapeutic strategy
to enhance efficacy of immunotherapies. Moreover, by means of single cell RNA-seq
analysis we accounted SUMOi with a key function in altering the global immune landscape.
Summary/Conclusion: In summary, we show that activated SUMOylation allows tumor cells
to evade anti-tumor immunosurveillance and establish SUMOi as rational therapeutic
strategy for enhancing the efficacy of immunotherapies in lymphoma and other aggressive
cancers.
S229: CHARACTERISATION OF TUMOR MICROENVIROMENT REMODELLING IN THE PROGRESSION OF
AITL WITH SINGLE-CELL RNA SEQUENCING AND IMAGING MASS CYTOMETRY
H. Jin1,*, X. Lu1, L. Fan1, L. Cao1, W. Zhang1, J. Li1, Z. Wu1
1Department of Hematology, The First Affiliated Hospital of Nanjing Medical University,
Jiangsu Province Hospital, Nanjing, China
Background: Angioimmunoblastic T-cell lymphoma (AITL) is a common subtype of peripheral
T-cell lymphoma (PTCL). AITL is an aggressive malignancy with a poor prognosis, and
its clinical manifestations vary greatly among individuals. The current chemotherapy
regimens based on anthracycline show limited efficacy, and there is no best rescue
treatment for patients with relapsed and refractory (RR) AITL. In addition, the lack
of optimal AITL models in vitro greatly limits the basic research on the mechanism
of disease occurrence and progression, and also hinders the development of new drugs
and preclinical trials.
Aims: Our study aims to deeply analyze the tumor heterogeneity and microenviroment
remodelling in the progression of AITL at single cell resolution, discovering key
molecules of drug resistance and potential theraputic targets.
Methods: We detected fresh lymph node samples from initail diagnosed (ID) and relapsed
and refractory (RR) AITL patients using single-cell RNA sequencing (scRNA-seq), combined
with imaging mass cytometry (IMC) and whole exome sequencing. scRNA-seq was performed
to compare the differential transcriptome expression patterns in ID- and RR-AITL samples.
IMC was performed to analyze the spatial position relationship and protein expression
characteristics of different subgroups in the tumor microenvironment of AITL. Immunofluorescence
staining was conducted to detect the expression of relative markers on tissue microarrays
in patients with AITL. In addition, AITL patient-derived organoid model was established
to study the regulatory role of YY1 and its inhibitors in relapsed and refractory
AITL.
Results: We found that RR-AITL samples exhibited significant differences in the tumor
microenvironment compared with ID patients. ScRNA-seq analysis revealed that transcription
factor YY1 was significantly highly expressed in follicular helper T cells (Tfh) of
RR-AITL patients, which promoted the proliferation and drug resistance of AITL cells
(Fig A-D), consistent with the results of IMC (Fig E,F). The proportion of CD8+ T
cells in the RR-AITL sample was reduced, while the proportion of Treg was increased,
as well as the depletion of T cells (Fig G). Furthermore, the stemness of B cells
in RR-AITL was enhanced and exhibits significant malignant characteristics (Fig H).
We also found decreased interaction in RR-AITL samples. B cells and myeloid subgroups
may play important roles in the progression of AITL (Fig I,J). As shown in Fig K,
EBV+ B cells exhibited a wider and more distribution in RR-AITL (Fig L). Interestingly,
EBV+ endothelial cells were presented in both ID- and RR- samples, while the spatial
analysis showed that the distance of EBV+ endothelial cells to B cells in RR-AITL
was obviously greater than in ID group, and the distance of Tfh to B cells also showed
similar result (Fig M,N). Moreover, for the first time, we established AITL patient-derived
organoid models that can be stablely cultured in vitro. On this basis, we could further
clarify the important roles of transcription factor YY1 in the drug resistance of
AITL, evaluate the cytotoxic effect of YY1 inhibitor NP-001 on AITL tumor cells.
Image:
Summary/Conclusion: In conclusion, our study revealed the differences between initial
diagnosed and relapsed /refractory AITL in terms of tumor microenvironment, single-cell
transcriptomes, the spatial distributions of different clusters and their interactions
features. YY1 may serve as an novel target for drug resistance for RR-AITL patients.
These findings may provide a theoretical foundation for improving the clinical treatment
of AITL.
S230: HUMAN ΒETA-DEFENSIN 2 TREATMENT MODULATES THE INTESTINAL MICROBIOME AND THE
ALLOGENEIC T CELL RESPONSE TO LIMIT ACUTE GRAFT-VERSUS-HOST DISEASE
T. Rückert1,*, G. Andrieux2 3, M. Boerries2 3, N. M. Woessner4 5 6, S. Minguet4 5
7, S. Doetsch4 5, K. Aumann8, M Schiff1, L. M. Braun1, E. Haring1 4, B. A. Siranosian9,
A. S. Bhatt9 10, P. Nordkild11, J. Wehkamp12, B. A. H. Jensen13, J. Duyster1 3, R.
Zeiser1 3 14, N. Köhler1 14
1Department of Medicine I; 2Institute of Medical Bioinformatics and Systems Medicine,
Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg;
3German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner
Site Freiburg; 4Faculty of Biology; 5Signalling Research Centres BIOSS and CIBSS;
6Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg; 7Institute
for Immunodeficiency, Center for Chronic Immunodeficiency (CCI); 8Institute of Surgical
Pathology, Medical Center – University of Freiburg, Faculty of Medicine, University
of Freiburg, Freiburg, Germany; 9Department of Genetics; 10Department of Medicine
(Hematology, Blood and Marrow Transplantation), Stanford University, Stanford, United
States of America; 11Defensin Therapeutics, Copenhagen, Denmark; 12Department of Internal
Medicine I, University Hospital Tübingen, Tübingen, Germany; 13Department of Biomedical
Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen,
Denmark; 14CIBSS – Centre for Integrative Biological Signalling Studies, University
of Freiburg, Freiburg, Germany
Background: Allogeneic hematopoietic cell transplantation (allo-HCT) is a curative
therapy option for patients with hematological malignancies. Acute graft-versus-host
disease (GVHD) is primarily driven by allogeneic donor T cells and remains a serious
cause of morbidity and mortality post allo-HCT. In particular, acute GVHD of the gastrointestinal
tract (GI GVHD) is associated with a high mortality rate and treatment options are
limited. Human β-defensin 2 (hBD-2) is an endogenous epithelial cell-derived host-defense
peptide, which is induced by inflammatory stimuli and possesses both antimicrobial
and immunomodulatory functions.
Aims: In this study, we aimed to investigate the expression of beta defensins in human
and murine GI GVHD and the therapeutic effect of recombinant hBD-2 on acute GVHD development
in mouse models.
Methods: To investigate the functional role of hBD-2 post allo-HCT, we employed established
murine GVHD and graft-versus-leukemia (GVL) models and analyzed the allogeneic T cell
response using transcriptome and kinome profiling. Shotgun metagenomic sequencing
was used to examine the effect of hBD-2 on the intestinal microbiome. Furthermore,
we studied beta-defensin expression in two independent acute GVHD patient cohorts
and in mice with acute GVHD.
Results: We found that expression of murine beta-defensin 4 (mBD-4), the murine orthologue
for hBD-2, was reduced in the colon and ileum of mice developing acute GVHD. Oral
treatment of mice with recombinant hBD-2 post allo-HCT reduced weight loss and acute
GVHD severity and mortality. Furthermore, hBD-2 treatment affected the intestinal
microbial composition, including a shift towards higher abundance of Bacteroides species
in hBD-2 treated compared to vehicle treated mice. The changes in the microbiome resulted
in reduced neutrophil infiltration of the ileum during acute GVHD induction in hBD-2
treated mice. Additionally, hBD-2 dampened pro-inflammatory Th1 cytokine production
(TNF, IFN-gamma) by allogeneic T cells in vivo, while preserving the beneficial GVL
effect in two different leukemia models. Mechanistically, oral hBD-2 treatment decreased
alloreactive T cell infiltration and the expression of genes involved in T cell receptor
(TCR) signaling in the ileum of mice with acute GVHD. Using transcriptome and kinome
profiling, we found that hBD-2 directly dampened primary murine and human allogeneic
T cell proliferation, activation and metabolism by reducing proximal TCR signaling.
In patients with acute GI GVHD, intestinal hBD-2 expression was inadequately induced
in response to inflammation at the mRNA and protein level when compared to healthy
subjects and patients with ulcerative colitis.
Summary/Conclusion: In conclusion, our study demonstrates that hBD-2 reduces acute
GVHD severity, likely through its effects in shaping the intestinal microbiota, reducing
neutrophil infiltration in the ileum and dampening allogeneic T cell responses. Both
human and murine acute GVHD are characterized by a lack of intestinal beta-defensin
induction and recombinant hBD-2 represents a potential novel therapeutic strategy
to counterbalance endogenous hBD-2 deficiency.
S231: GLUCOCORTICOID AND GLYCOLYSIS INHIBITORS COOPERATIVELY ABROGATE ACUTE GRAFT-VERSUS-HOST
DISEASE
Q. wen1, Z. xu1,*, Y. wang1, M. lv1, Y. song1, Z. lv1, T. xing1, L. xu1, X. zhang1,
X. huang1, Y. kong1
1Peking University Institute of hematology, Beijing, China
Background: Although glucocorticoids(GCs) are the standard first-line therapy for
acute graft-versus-host disease(aGvHD), approximately 50% of aGvHD patients have no
response to GCs. The role of T cell metabolism in murine aGvHD has been recently reported.
Pharmacological inhibition of glycolysis by targeting 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase
3(PFKFB3) ameliorates aGvHD. However, the question of whether glycolysis is required
for the function of activated T cells and whether abnormal glycolysis is involved
in the occurrence of human aGvHD need to be elucidated. Moreover, the issue as to
whether GCs and metabolism regulators can cooperatively suppress T cell alloreactivity
and ameliorate aGvHD remains to be elucidated.
Aims: The study aimed to analyse the glucose metabolism profiles of activated T cells
in aGvHD patients, and to evaluate the roles of PFKFB3-stimulated glycolysis in alloantigen-activated
T cells and aGvHD induction. Furthermore, the study was performed to explore the issue
of whether GCs could restore activated T cells by regulating glycolysis and the effect
of GCs combined with glycolysis inhibitors on activated T cells.
Methods: In this prospective nested case–control study, a total of 15 aGvHD patients
at diagnosis and 15 matched non-aGvHD patients. The glucose consumption and lactate
production rates were detected by glucose assay kit and lactate assay kit. Subsequently,
to assess whether functional T cell activation is caused by the regulation of PFKFB3,
we used lentivirus transduction for the genetic regulation of PFKFB3 in vitro and
in a humanized xenogeneic aGvHD model. The effect of GCs and glycolysis inhibitors
on activated T cells was further explored in vitro and in a humanized murine model
of aGvHD and graft versus leukaemia (GVL). In addition, to evaluate the synergistic
effect of GCs, 3PO or their combination, combination index (CI) studies were performed
by using the Chou-Talalay method for drug interactions.
Results: Increased glycolysis, which was characterized by elevated PFKFB3, as well
as higher rates of glucose consumption and lactate production in aGvHD T cells. Importantly,
in vitro treatment with glycolysis inhibitor 3PO improved the activity of T cells
derived from aGvHD patients through down-regulating glycolytic activity of T cells.
Genetic upregulation of PFKFB3 induced T cell proliferation and differentiation into
proinflammatory T cells. In a humanized mouse model, PFKFB3-overexpressing T cells
aggravated aGvHD, as characterized by high aGvHD clinical scores, pathological scores
and rapid lethality. Importantly, our integrated data from patient samples in vitro
showed that GCs combined with 3PO decreased proinflammatory T cells and T cell proliferation,
reduced glucose consumption, lactate production and PFKFB3 expression compared with
the single GCs group. Notably, the average CI values of GCs and 3PO were 0.364 for
the synergistic inhibition of IFN-γ synthesis and 0.475 for the synergistic inhibition
of proliferation, respectively. In a humanized murine model of aGvHD and GVL demonstrated
that GCs and 3PO cooperatively reduced the alloreactivity of T cells and ameliorate
aGvHD without a loss of GVL effects.
Summary/Conclusion: Our data indicated that glycolysis is critical for T cell activation
and the induction of human aGvHD. GCs combined with glycolysis inhibitors demonstrated
synergistic effects on reducing T cell alloreactivity and ameliorating aGvHD by regulating
T cell glycolysis in vitro and in vivo. Thus, GCs combined with glycolysis inhibitors
promise to be a novel therapeutic strategy for aGvHD patients.
S232: EXPANDING THE REPERTOIRE OF HLA CLASS I-RESTRICTED MINOR HISTOCOMPATIBILITY
ANTIGENS FOR IMMUNE MONITORING AND MODULATION AFTER ALLOGENEIC STEM CELL TRANSPLANTATION
K. Fuchs1,*, M. van de Meent1, W. Honders1, C. van Bergen1, J. F. Falkenburg1, M.
Griffioen1
1Hematology, LUMC, Leiden, Netherlands
Background: Allogeneic stem cell transplantation is given as curative treatment for
hematological malignancies, but patients face the risk of relapse of their malignancy
as well as Graft-versus-Host Disease. After transplantation, donor T cells recognize
polymorphic peptides presented by HLA surface molecules on the patient’s cells. These
polymorphic peptides, called minor histocompatibility antigens, are caused by genetic
differences in single nucleotide polymorphisms (SNPs) between patient and donor. Dependent
on whether the antigen is presented on tumor cells or healthy non-hematopoietic tissues
of the patient, donor T cells may induce the favourable Graft-versus-Leukemia effect
or Graft-versus-Host-Disease, respectively.
Aims: The aim is to identify the dominant repertoire of minor histocompatibility antigens
in seven common HLA class I molecules for immune monitoring and modulation after allogeneic
stem cell transplantation.
Methods: Donor T cells isolated from patients after allogeneic stem cell transplantation
are tested for recognition of a new panel of 191 selected B cell lines, which are
sequenced in the 1000 Genomes Project. This panel enables the inclusion of seven common
HLA class I molecules and increases SNP coverage to 11 million (MAF > 0.01). SNPs
that strongly associate with T cell recognition are subsequently validated to encode
minor histocompatibility antigens.
Results: Using an optimized approach for whole genome association scanning, more than
80 new minor histocompatibility antigens have been found that are presented by seven
common HLA class I molecules. Antigens targeted in immune responses after transplantation
were often shared between patients, and about 25-30% of antigens were translated in
other reading frames than human proteins with known function. Furthermore, the extended
repertoire was investigated for T cell frequency and tissue distribution of antigen-encoding
genes, pointing towards several antigens as potential targets for immunotherapy.
Summary/Conclusion: In conclusion, despite many SNP mismatches between patients and
donors, our data demonstrate that the repertoire of minor histocompatibility antigens
is confined. As the antigens were identified by an unbiased forward strategy (T cell-to-antigen),
our collection provides relevant insight into the various sources of antigens in annotated
and unannotated reading frames, which is essential for accurate antigen prediction
by reverse strategies (antigen-to-T cell). Furthermore, we more than doubled the number
of minor histocompatibility antigens which are fundamental to predict, follow or manipulate
immune responses after allogeneic stem cell transplantation to improve clinical outcome
of transplanted patients.
S233: THERAPEUTIC GENE EDITING OF T CELLS CORRECTS CTLA4 INSUFFICIENCY.
T. Fox1,*, B. Houghton1, L. Petersone1, N. Edner1, O. Preham1, E. Waters1, C. Hinze1,
A. McKenna1, C. Williams1, A. Kennedy1, A. Pesenacker1, P. Genovese2, L. Walker1,
S. Burns1, D. Sansom1, C. Booth1, E. Morris1
1University College London, London, United Kingdom; 2Boston Children’s Hospital, Boston,
United States of America
Background: Heterozygous mutations in CTLA4 result in an inborn error of immunity
(IEI) (also known as primary immunodeficiency) with a severe clinical phenotype. Autologous
T cell gene therapy may offer a cure without the immunological complications of allogeneic
stem cell transplantation. The mutational landscape and requirement for tight regulation
of CTLA4 make viral gene addition approaches unappealing. Gene editing strategies
permit alteration of CTLA4 while retaining the endogenous gene control machinery.
Aims: We set out to devise a CRISPR/Cas9/AAV6 gene editing strategy to correct CTLA4
insufficiency in T cells.
Methods: We designed several homology directed repair (HDR) editing strategies that
would correct the genetic defect. We first assessed correction of an individual point
mutation. We then evaluated several universal strategies that enable correction of
most disease-causing mutations with a single edit; the first that inserts the CTLA4
cDNA in exon 1, and a second that inserts the CTLA4 cDNA at the 3’end of the first
intron of CTLA4. All AAV6 HDR donor templates included a GFP reporter gene to enable
easy identification of the edited cells.
Results: Superior editing efficiencies were obtained with the intronic approach compared
to the other editing strategies and this strategy was then further evaluated. CTLA4
function and expression kinetics were assessed following editing using flow cytometry-based
assays. Functional studies using CTLA4 transendocytosis (TE) assays, demonstrated
restoration of CD80 and CD86 internalization in the edited CD4+ T cells. Following
gene editing, transgene expression kinetics were comparable to healthy control CD4+
T cells. Gene editing of T cells isolated from patients with CTLA4 insufficiency restored
CTLA4 expression and rescued transendocytosis of CD80 and CD86 in vitro. Using a similar
approach, gene corrected T cells from CTLA4-/- mice engrafted in immunodeficient mice
at clinically relevant frequencies and tail vein bleeds were performed 1, 3 and 4
weeks post adoptive transfer. In the mice which received the GFP+ edited cells, a
stable population of GFP+ cells was detectable at all timepoints demonstrating in
vivo persistence as well as genetic stability. All mice were sacrificed 4 weeks after
cell transfer. To assess lymphoproliferation, the cellularity of peripheral lymph
nodes and spleen weight were analyzed. Spleen and lymph node size, lymph node cell
counts, and spleen weight were all significantly lower in mice which received the
edited cells (n=5) compared to non-edited controls (n=5) and there was no significant
difference in these parameters between mice who had received edited CTLA4-/- T cells
and those who received WT T cells (n=4). Analysis of lymph node and spleen cells confirmed
persistence of CTLA4 expression in the edited (GFP+) cells and revealed that a higher
proportion of Treg than Tconv had been successfully edited.
Summary/Conclusion: Together these data demonstrated that CTLA4 edited T cells survived
in vivo, expressed CTLA4 and were able to control the clinical phenotype of CTLA4
insufficiency, providing a powerful proof-of-principle of our T cell GT approach.
Our data provide proof-of-concept that gene editing can restore CTLA4 function in
T cells demonstrating the potential of this approach to treat CTLA4 insufficiency.
A similar approach could be used in other IEIs that are caused by multiple heterozygous
mutations.
S234: CLINICAL-GRADE MBIL21/4-1BBL EXPANDED NK CELLS EXHIBIT STRONGER COMPETENCE COMPARED
WITH PRIMARY NK CELLS AGAINST HCMV INFECTION
Q.-N. Shang1 2 3,*, X.-X. Yu1 2 3, Z.-L. Xu1 2, T.-T. Han1 2, J. Xie1 2, Z.-Y. Fan1
2, M. Zhao1 2, X.-H. Cao1 2, X.-F. Liu1 2 3, Y.-H. Chen1 2, M. Lv1 2, Y.-Q. Sun1 2,
Y.-J. Chang1 2, Y. Wang1 2, L.-P. Xu1 2, X.-H. Zhang1 2, K.-Y. Liu1 2, X.-Y. Zhao1
2, X.-J. Huang1 2 3
1Peking University People’s Hospital; 2Peking University Institute of Hematology;
3Peking-Tsinghua Center for Life Sciences, Beijing, China
Background: Cytomegalovirus (CMV) infection remains a common complication and leads
to high mortality in subjects who undergo allogeneic hematopoietic stem cell transplantation
(allo-HSCT). Human NK cells are the first lymphocyte recovering after allo-HSCT. Previous
studies indicated the early NK cell reconstitution may be protective against development
of HCMV infection post HSCT. Our previous data also showed that rapid reconstitution
of IFN-γ secreted NK cells at day 15 post transplantation predicted lower HCMV reactivation.
However, little is currently known about whether and how NK cells are responsible
for preventing HCMV infection post transplantation. NK cell adoptive transfer is a
promising method for cancer immunotherapy. Ex vivo mbIL21/4-1BBL expanded NK cells
exhibited high cytotoxicity against leukemia cells. Nevertheless, whether expanded
NK cells owned higher anti-HCMV function compared with primary NK cells in vitro and
in vivo were still unknown.
Aims: To investigate whether expanded NK cells owned higher anti-HCMV function compared
with primary NK cells in vitro and in vivo and the efficacy of adoptive NK cells infusion
to patients post HSCT to prevent HCMV infection.
Methods: NK cells were firstly ex vivo expanded by K562-mbIL21/4-1BBL feeder cells.
We compared not only the cytotoxicity against HCMV infected fibroblast but also the
competence of inhibiting HCMV propagation and reducing HCMV infection between ex vivo
expanded NK cells and primary NK cells. Then primary NK cells and expanded NK cells
were adoptive infused to HCMV-infected humanized mice to investigate the ability to
eliminate HCMV infection in vivo. At last, 20 patients post HSCT were enrolled in
our clinical trial to explore the safety and efficacy of adoptive infusion of K562-mbIL21/4-1BBL
expanded NK cells to protect from HCMV infection.
Results: 1. Most activating receptors, cell adhesion molecule receptors and chemokine
receptors exhibited enhanced expression on expanded NK cells (figure 1a). Expanded
NK cells showed stronger cytotoxicity against HCMV infected fibroblasts and enhanced
abilities to inhibit HCMV propagation compared with primary NK cells in vitro (figure
1b).
2. Both expanded NK cells and primary NK cells showed the ability to migrate to the
spleen, liver, and lung and persisted in these target organs in HCMV-infected humanized
mice. On day 14 post infusion, expanded NK cells showed higher percentage in tissues
than primary NK cells. Moreover, mice with expanded NK cell infusion exhibited more
effective HCMV elimination compared with primary NK cell infusion.
3. The cumulative incidence of HCMV and refractory HCMV infection for patients in
NK infusion cohort were significantly reduced compared with control cohort (figure
1c). Total HCMV persisting time was shortened (14 (5-24) days vs. 17 (7-56) days,
p=0.013). What is more, the absolute number of NK cells was higher for patients in
NK cell infusion group than that in control group. Higher quantitative reconstitution,
more matured NK cell phenotypes and stronger function were determined in patients
with expanded NK cell infusion than those in control group.
Image:
Summary/Conclusion: Based on in vitro and in vivo studies, our data demonstrated that
adoptive NK cells infusion exhibited stronger activities compared with primary NK
cells against HCMV infection.
S235: A TWO-PART, SINGLE- AND TWO-ARM RANDOMIZED, OPEN-LABEL STUDY TO EVALUATE THE
SAFETY, TOLERABILITY AND PHARMACOKINETICS OF THE S1P RECEPTOR MODULATOR KRP203 IN
SUBJECTS WITH HEMATOLOGICAL MALIGNANCIES
S. dertschnig1,*, J. finke2, D. heim3, U. schanz4, E. holler5, U. holtick6, G. socié7,
T. teshima8, C. bucher9, J. passweg10
1Priothera SAS, Saint Louis, France; 2Heamtology, Oncology and SCT, University Freiburg,
Freiburg, Germany; 3Hematology, University Hosptial Basel, Basel; 4University Hospital
Zürich, Zürich, Switzerland; 5Klinik und Poliklinik Innere Medizin III, Unuversitätsklinikum
Regensburg, Regensburg; 6Klinik I für Innere Medizin, Köln, Germany; 7Saint Louis
Hospital, Paris, France; 8Hokkaido University, Faculty of Medicine, Sapporo, Japan;
9Universtity Hospital Basel; 10University Hosptial Basel, Basel, Switzerland
Background: The success of allogeneic hematopoietic stem cell transplantation (HSCT)
is limited by disease relapse. Alloreactive donor T cells have the potential to prevent
relapse by the graft-versus-leukemia (GVL) response. The GVL response is essential,
however, the same allo-T cells cause acute graft-versus-host disease (aGVHD). T cell
trafficking out of lymphoid organs is a major step in raising peripheral immune responses
and is regulated by sphingosine-1-phosphate (S1P) gradients. Pre-clinical data showed
that pharmacological modulation of S1P receptor (S1PR) signalling by KRP203 sequesters
T cells in lymphoid organs and prevents their egress to aGVHD target sites. In murine
models, KRP203 efficiently reduced aGVHD. Because T cell function is not suppressed
by KRP203, GVL was maintained, resulting in improved survival. Here, we show data
of the first clinical trial investigating the safety and tolerability of KRP203 in
patients with hematological malignancies undergoing HSCT.
Aims: The aim of this study was to evaluate the safety, tolerability and pharmacokinitecs
of a S1P receptor modulator towards a novel treatment approach that aims at reducing
GvHD while maintaining Graft versus leukemia.
Methods: This multicentric, phase Ib, prospective, open label, two-part study evaluated
safety, tolerability and pharmacokinetics of KRP203 in intermediate to high-risk patients
undergoing HSCT for hematological malignancies. Three treatment arms were investigated:
3mg KRP203+CsA/MTX, 1mg KRP203+CsA/MTX and 3mg KRP203+Tac/MTX. KRP203 was administered
from day 1 until day 111 and HSCT was performed on day 11.
Results: KRP203 was safe and well-tolerated in these fragile patients. Upon conditioning
and KRP203 treatment absolute lymphocyte counts (ALC) were reduced to about 0.2x109/L,
engrafted normally and during KRP203 exposure ALC stabilized at <0.7x109/L in all
three treatment arms, well below pre-HSCT levels. The treatment effect on ALC counts
resolved when KRP203 was stopped on day 111, resulting in blood ALC to pre-HSCT levels.
CD4+ and CD8+ T cells, were reduced in peripheral blood in response to KRP203 treatment.
CD4+ T cells remained below pre-HSCT level (<100 cells/µl) during the treatment period.
CD8+ T cell counts recovered more rapidly, also during treatment. Overall survival
probability was highest for subjects in the 3mdg KRP203+CsA arm with the first death
reported on day 532. Overall survival at 1 year was 100% in the 3mg KRP203+CsA, 67%
in the 1mg KRP203+CsA and 36% in the 3mg KRP203+Tac arm. The median time to relapse
was 749 days. 2/6 subjects in 1mg KRP203+CsA, 4/10 subjects in 3mg KRP203+CsA and
4/7 subjects in 3mg KRP203+Tac arm relapsed. The median time to any aGVHD was 55 days.
22% of subjects presented with grade III-IV aGVHD. The median time to any chronic
GVHD was 174 days. 57% of subjects presented with moderate and 9% with severe chronic
GVHD.
Summary/Conclusion: KRP203 is safe and well tolerated and shows promising early clinical
outcomes with a limited number of relapses, acute and chronic GVHD. These data support
the initiation of a phase 2b trial investigating KRP203 as an adjunctive and maintenance
treatment as an adjunctive and maintenance treatment to increase leukemia free survival
in adult AML patients undergoing allo-HSCT.
S236: RANDOMIZED MULTICENTER PHASE III STUDY OF HAPLO VERSUS HLA-MATCHED UNRELATED
DONOR (UD) ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION (ALLO HSCT) FOR PATIENTS
OLDER THAN 55 YEARS
S. Harbi1, J.-M. Boher2, E. Forcade3, P. Chevallier4, R. Peffault De Latour5, F. Malard6,
S. Francois7, A. Charbonnier8, E. Hermet9, C. E. Bulabois10, A. Huynh11, A. Berceanu12,
T. Cluzeau13, M. T. Rubio14, S. Furst1, R. devillier15, M. Mohty6, D. Blaise1,*
1hematology; 2DRCI, Institut Paoli Calmettes, Marseille; 3hematology, CHU, Bordeaux;
4hematology, CHU, Nantes; 5Transplant Unit, Hopital Saint Louis; 6hematology, Hopital
Saint Antoine, Paris; 7hematology, CHU, Angers; 8hematology, CHU, Amiens; 9hematolgy,
CHU, Clermont Ferrand; 10hematolgy, CHU, Grenoble; 11hematology, CHU, Toulouse; 12hematology,
CHU, besancon; 13hematology, CHU, Nice; 14hematology, CHU, Nancy; 15hematolgy, Institut
Paoli Calmettes, Marseille, France
Background: In most situations where allo HSCT is wished a matched sibling donor (MSD)
is lacking. The respective advantages of alternative donor such as UD or HaploD are
yet to be precisely assessed notably in older population including the search and
identification process.
Aims: We performed a prospective, multicenter, open-label, randomized controlled trial
(NCT02623309) comparing Haplo versus UD search strategies for allo HSCT in pts with
hematological malignancies older than 55 years after the absence of MSD was established.
Methods: The primary objective was the comparison of Chronic Graft-versus-Host Disease-free
and Relapse-free survival (cGRFS) from time of randomization. Conditioning was RIC
based on fludarabine (150 mg/m2), IV busulfan (260 mg/m2) for haplo HSCT with the
adjunction of thiotepa (5 mg/kg) for UD-HSCT. Pts receiving Haplo-SCT and UD-HSCT
received GVHD prophylaxis based on HD-PTCy (100 mg/kg) and ATG (5 mg/kg), respectively.
Additionnal prophylaxis consisted in CSA and mycophenolate mofetil for all patients
(pts).
Results: 108 pts were enrolled, and 106 pts were analyzed with a median follow-up
of 27 (14-34) months. Median age was 65 years (55-70). Diseases were myeloid malignancies
in 84 pts (79%). DRI was low, intermediate and high in 5(5%), 59(55%) and 42(40%),respectively.
55 and 51 pts were assigned to Haplo and UD group respectively. In case an alternative
donor was not identified in due time, a search for another donor was allowed.
Of the 106 pts, 77 (73%) pts proceeded to transplant (40 (73%) and 37 (73%) pts in
Haplo and UD group respectively). Fifteen (27%) pts in Haplo group did not because
of progression (n=9), patient contraindication (n=5), absence of donor (n=1). Likewise
14 (27%) pts in UD group were not transplanted because of progression (n=8), patient
contraindication (n=6). Overall median time from randomization to allo-SCT was 76
(21-179) and 95 (37-310) days in haplo and UD group respectively (P=NS). Thity-one
(56%) and 26 (51%) pts actually were transplanted according to randomization while
9 (16%) and 11 (22%) pts were transplanted from a UD or a HaploD in haplo and UD search
group respectively. Median time from randomization to allo-SCT was 81 (21-288) when
the initial desired donor was found and 117 (38-310) days when another donor type
was to be searched (p=0,04). In an intent-to-treat analysis from date of randomization,
2-year GRFS, PFS and OS did not differ between the two groups (Haplo vs UD search
group: 29% vs 37%, p=0.22; 45 vs 49 %, p=0.56; 50 vs 59%, p=0.47).
Overall 42 and 35 pts were actually transplanted from a haploidentical and unrelated
donor, respectively whatever the randomization arm. With a median follow up of 24
months after transplant 2-year GRFS and PFS from transplant did not differ between
Haplo and UD HSCT: 40% vs 34%, p=0.66; 48%vs 45%, p=0.78). No statistical difference
was documented for G2-4 aGVHD, severe cGVHD, NRM and relapse probabilities.
Summary/Conclusion: This trial from alternative donor search initiation time-point
establishes for the first time that the search for a HaploD or UD conduct to the same
outcomes in older pts. However, although 77% of the patients can achieved a transplant
with either strategies, only half of the original pts will receive a transplant from
the prospected donor type. Outcomes being similar, this may invite to initially perform
search for both type of donors to avoid delay with secondary searches.
S237: ORCA-T, AN ENGINEERED ALLOGRAFT, RESULTS IN HIGH GVHD-FREE AND RELAPSE-FREE
SURVIVAL FOLLOWING MYELOABLATIVE CONDITIONING FOR HEMATOLOGICAL MALIGNANCIES
E. Meyer1,*, A. Pavlova1, A. Gandhi2, R. Hoeg3, C. Oliai4, R. Mehta5, S. Srour5, J.
McGuirk6, E. Waller7, N. Fernhoff8, M. S. Killian8, J. Mcclellan8, A. Putnam8, B.
Shaw9, M. Abedi10, R. Negrin11
1BLOOD AND MARROW TRANSPLANTATION AND CELLULAR THERAPY, Stanford Hospital and Clinics,
Stanford, CA; 2Department of Medicine, Division of Hematology/Medical Oncology, Oregon
Health and Science University, Portland, OR; 3Department of Medicine, Division of
Bone Marrow Trasnplant, University of California, Davis, Comprehensive Cancer Center,
Davis, CA; 4Department of Medicine, Blood and Bone Marrow Transplant Program, University
of California, Los Angeles, Los Angeles, CA; 5Department of Stem Cell Transplantation
and Cellular Therapy, Division of Cancer Medicine, MD Anderson Cancer Center, Houston,
TX; 6Department of Hematologic Malignancies and Cellular Therapeutics, University
of Kansas Medical Center, Kansas City, KS; 7Bone Marrow and Stem Cell Transplant Center,
Winship Cancer Institute of Emory University, Atlanta, CA; 8Orca Bio, Menlo Park,
CA; 9Department of Medicine, Division of Hematology and Oncology, BMT Program, Medical
College of Wisconsin, Wilwaukee, WI; 10Department of Medicine, Division of Bone Marrow
Transplant, University of California, Davis Medical Center, Davis, CA; 11Department
of Blood and Marrow Transplantation and Cellular Therapy, Stanford Hospital and Clinics,
Stanford, CA, United States of America
Background: Rates of graft versus host disease (GVHD) and non-relapse mortality (NRM)
following myeloablative allogeneic hematopoietic stem cell transplant (MA-alloHSCT)
remain unacceptably high. Strategies to reduce GVHD and NRM have been compromised
by limited efficacy or increased risk of infection and relapse, emphasizing the need
for new approaches that holistically improve outcomes.
Orca-T is a high-precision, allogeneic investigational cell therapy product comprised
of stem and immune cells that leverages highly purified, polyclonal donor regulatory
T cells to control alloreactive immune responses, reducing the need for pharmacologic
GVHD prophylaxis. Orca-T is produced in a central GMP facility and has been successfully
scaled to clinical centers throughout the U.S.
Aims: The aim of these studies was to evaluate the safety and efficacy of Orca-T in
patients with hematologic malignancies.
Methods: As of 28 February 2022, 138 patients with high-risk hematologic malignancies
have received Orca-T in a single-center Phase 1-2 study (NCT01660607, n=41) and a
multicenter Phase 1b study (NCT04013685, n=97) and have ≥ 100 days of follow-up. Informed
consent was obtained from all transplant recipients and donors, and the studies received
IRB approval from participating institutions. Orca-T was produced from G-CSF-mobilized
peripheral blood (PB) from matched related donors (n=72), matched unrelated donors
(n=62), or mismatched unrelated donors (MMUD, n=4). Median follow-up for recipients
was 300 days (range: 27-1941). Median age was 49 years, and diagnoses included AML
(43%), ALL (27%), MDS (10%), myelofibrosis (7%), and CML (6%). Patients received myeloablative
conditioning (busulfan-based, n=109; TBI-based, n=27; BCNU, n=2) followed by GVHD
prophylaxis with either single-agent tacrolimus (tac, n=127), sirolimus (n=7), or
tac plus mycophenolate (n=4, MMUD). A contemporaneous CIBMTR-based control arm was
obtained that consisted of patients with similar diagnoses who received myeloablative
alloHSCT from a PB source followed by tac/methotrexate PPX.
Results: Orca-T was successfully manufactured, distributed, and infused for all patients
enrolled. Overall time from donor centers to recipient centers was under 60 hours
in all cases. Median time to neutrophil engraftment was 13 days. The rates of grade
≥ 3 acute GVHD in the first 180 days and moderate to severe chronic GVHD through 1
year were low with Orca-T at 4% and 5%, respectively. NRM was infrequent at 4% through
1 year. Orca-T exhibited GRFS of 71% & OS of 90% at 1 year. No formal comparison to
the CIBMTR cohort was performed, and a Phase 3 study has been initiated to confirm
these findings. Longitudinal immune reconstitution data was collected and will be
presented. Clinical data is summarized in Table 1.
*MAGIC Criteria **NIH Consensus Grading
Image:
Summary/Conclusion: Results from patients treated with Orca-T, a high-precision Treg-engineered
donor product, suggest a reduction in cGVHD, improved GRFS, and low toxicity relative
to historic data. Orca-T manufacturing was accomplished with consistent and reliable
cell manufacturing and distribution across a wide geographic area. A multicenter randomized-control
trial phase 3 trial comparing Orca-T to SOC has been initiated.
S238: MATCHED RELATED VERSUS UNRELATED VERSUS HAPLOIDENTICAL DONORS FOR ALLOGENEIC
TRANSPLANTATION IN AML PATIENTS ACHIEVING FIRST COMPLETE REMISSION AFTER TWO INDUCTION
COURSES: A STUDY FROM THE ALWP/EBMT
A. Nagler1,*, M. Labopin2, S. Mielke3, J. Passweg4, D. Blaise5, T. Gedde-Dahl6, J.
J. Cornelissen7, U. Salmenniemi8, I. Yakoub-Agha9, P. Reményi10, G. Socié11, G. Van
Gorkom12, H. Labussière-Wallet13, X.-J. Huang14, M. Thérèse Rubio15, J. L Byrne16,
C. Craddock17, L. Griskevicius18, F. Ciceri19, M. Mohty20
1Hematology Division, Sheba Medical Center, Ramat-Gan, Israel; 2EBMT ALWP office,
Hôpital Saint-Antoine, Paris, France; 3Hematology, Karolinska University Hospital;
4Department of Cell Therapy and Allogenic Stem Cell Transplantation (CAST), Department
of Laboratory Medicine (LabMED), Karolinska University Hospital and Institutet, Karolinska
Comprehensive Cancer Center, Stockholm, Sweden; 5Programme de Transplantation & Therapie
Cellulaire, Centre de Recherche en Cancérologie de Marseille, Paoli Calmettes, Marseille,
France; 6Hematology Dept., Section for Stem Cell Transplantation, Oslo University
Hospital, Rikshospitalet, Clinic for Cancer Medicine, Oslo, Norway; 7Department of
Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam,
Netherlands; 8Stem Cell Transplantation Unit, HUCH Comprehensive Cancer Center, Helsinki,
Finland; 9CHU de Lille LIRIC, INSERM U995, Université de Lille, Lille, France; 10Haematology
and Stem Cell Transplant, Dél-pesti Centrumkórház – Országos Hematológiai és Infektológiai
Intézet, Budapest, Hungary; 11Hematology – BMT, Hopital St. Louis, Paris, France;
12Department of Internal Medicine, Division of Hematology, GROW School for Oncology
and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands;
13Service Hematologie, Centre Hospitalier Lyon Sud, Lyon, France; 14Institute of Haematology,
Xicheng District, Peking University People´s Hospital, Beijing, China; 15CHRU BRABOIS,
Vandoeuvre les Nancy, Nancy, France; 16Nottingham University, Hucknall Road, Nottingham;
17Haematology, University Hospital Birmingham NHSTrust, Queen Elizabeth Medical Centre,
Edgbaston, Birmingham, United Kingdom; 18Haematology, Oncology & Transfusion Center,
Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania; 19Hematology and
BMT, San Raffaele Scientific, Milan, Italy; 20EBMT ALWP office Paris, Saint Antoine
Hospital, Paris, France
Background: Transplantation (HSCT) outcome is significantly inferior in AML patients
(pts) who required 2 rather than 1 induction course to achieve CR1, with a higher
relapse (RI) and lower leukemia-free (LFS) and overall survival (OS) and lower graft-versus-host
disease (GVHD)-free, relapse-free survival (GRFS).
Aims: To allocate the best donor for HSCT in AML pts achieving CR1 after 2 inductions
comparing matched related sibling donors (MSD) with 9-10/10 HLA compatible unrelated
donors (UD) and non-T cell depleted haploidentical (Haplo) donors, respectively.
Methods: This was a retrospective analysis including adult pts aged ≥18 years with
AML undergoing HSCT while in CR1 achieved after 2 inductions during the period 2010-2020.
Allografts were from MSD, UD or Haplo donors. Multivariate analysis (MVA) adjusting
for potential confounding factors was performed using a Cox’s proportional-hazards
regression model for main outcomes.
Results: 1295 pts were included: MSD (n=428), UD 10/10 (n=554), UD 9/10 (n=135), Haplo
(n=178). Median follow up was 48.4, 46.0,40.5 and 35.2 months, respectively. Median
age was 52.1, 53.8, 52.2 and 49.5 years, respectively. The 4 groups did not differ
with respect to pts’ gender, type of AML, cytogenetics, and performance status. Haplo
transplants were performed more recently, with more BM grafts and lower frequency
of pt CMV seropositivity. Post-transplant cyclophosphamide (PTCy) was the main anti
GVHD prophylaxis in HSCT from Haplo donors, while in vivo T-cell depletion was more
frequently used in transplants from UD. Myeloablative conditioning was less frequently
used in transplants from UD and time from diagnosis to HSCT was shorter in transplants
from MSD. Cumulative incidence of ANC >0.5x109/L at day 30 was 98.1%, 98.5%, 95.5%
and 92.1%, respectively (global P value=0.03). Univariate analysis of 2- and 5-year
outcomes after transplantation are given in Table 1. On MVA, acute (a) GVHD II-IV
but not III-IV was higher in all groups compared to MSD: UD 9/10, HR=2.53; 95% CI
1.68-3.82, P<0.0001; UD 10/10, HR=1.96; 95% CI 1.42-2.69, P<0.0001 and Haplo HR=2.15;
95% CI 1.41-3.3, P=0.0004. Incidence of extensive chronic (c) GVHD was significantly
higher in UD 9/10 (HR=2.52; 95% CI 1.55-4.11, P=0.0002) and UD 10/10 (HR=1.48; 95%
CI 1.03-2.13, P=0.036) and cGVHD all grades was higher in UD 9/10 vs MSD (HR=1.77;
95% CI 1.26-2.49, P=0.0009), while it did not differ in Haplo transplants. Non-relapse
mortality (NRM) was higher in HSCT from UD 10/10, 9/10 and Haplo donors compared with
those from MSD (HR= 1.75; 95% CI 1.16-2.63, P= 0.007, HR =2.22; 95% CI 1.29-3.83,
P= 0.004 and HR=2.53; 95% CI 1.47-4.34, P=0.0008), respectively. RI, LFS and OS did
not differ significantly between donor types: RI HR=0.84, P=0.18; HR=0.9, P=0.59 and
HR=0.76, P=0.17 in UD 10/10, UD 9/10 and Haplo vs MSD, respectively. LFS HR=1.02,
P=0.87; HR=1.15, P=0.35 and HR=1.11, P=0.49 and OS HR=1.12, P=0.3, HR=1.27, P=0.14
and HR=1.33, P=0.081, respectively. Finally, GRFS was lower in HSCT from UD 9/10 (HR=1.56,
95% CI 1.20-2.03, P<0.001) but not in those from UD 10/10 (HR=1.13, P=0.22) and Haplo
donors (HR=1.12, P=0.43) compared to MSD, respectively. Main cause of death in all
groups was original disease, followed by infections, and GVHD.
Image:
Summary/Conclusion: In AML patients undergoing allogeneic transplantation in CR1 achieved
after two induction courses which has been shown to be a poor prognostic factor, and
those who do not have an MSD, HLA matched UD and Haplo donors are comparable alternatives.
40-50% of these pts achieved long term LFS.
S239: COMPARABLE LONG-TERM OUTCOMES BETWEEN UPFRONT HAPLOIDENTICAL AND IDENTICAL SIBLING
DONOR HSCT IN APLASTIC ANEMIA: A NATIONAL REGISTRY-BASED STUDY
Z. xu1,*, L. xu1, D. wu2, S. wang3, X. zhang4, R. xi5, S. gao6, L. xia7, J. yang8,
M. jiang9, X. wang10, Q. liu11, J. chen2, M. zhou3, X. huang1
1Peking university institute of hematology, Beijing; 2The First affiliated Hospital
of Soochow University, Soochow; 3Guangzhou First People’s Hospital, Guangzhou; 4Xinqiao
Hospital affiliated to Third Military Medical University, Chongqing; 5General Hospital
of Lanzhou Military Region of PLA, Lanzhou; 6The First Hospital of Jilin University,
Changchun; 7Xiehe Hospital; 16affiliated to Huazhong University of Science and Technology,
Wuhan; 8Changhai Hospital affiliated to Second Military Medical University, Shanghai;
9The First affiliated Hospital of Xinjiang Medical University, Urumchi; 10Shandong
Provincial Hospital, Jinan; 11Nanfang Hospital affiliated to Southern Medical University,
Guangzhou, China
Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains
a curative option for severe aplastic anemia (SAA), and transplantation from identical
sibling donors (ISDs) has been recommended as a first-line treatment.
Aims: Haploidentical donor (HID) transplantation for SAA has made great advances;
thus, an increased role of HID-SCT in SAA should be considered if an ISD is unavailable.
Methods: We performed a national registry-based analysis comparing long-term outcomes
in the upfront HID or upfront ISD SCT setting.
Results:
A total of 342 SAA patients were enrolled, with 183 patients receiving HID SCT and
159 receiving ISD SCT.
The estimated 9-year overall survival (OS) and failure-free survival (FFS) were 87.1±2.5%
and 89.3±3.7% (P = 0.173) and 86.5±2.6% vs. 88.1±3.8% (P = 0.257) for patients in
the HID and ISD SCT groups, respectively. Transplantation from HID or ISD SCT has
greatly improved quality of life (QoL) levels post-HSCT compared to pre-HSCT. The
occurrence of chronic graft versus host disease was the only identified adverse factor
affecting each subscale of QoL. Physical and mental component summaries in adults
as well as physical, mental, social, and role well-being in children were all similar
between HID and ISD SCT at 5-year points. At the last follow-up, the proportion of
returning to society was comparable between the HID and ISD groups, showing 78.0%
versus 84.6% among children and 74.6% versus 81.2% among adults.
A total of 342 SAA patients were enrolled, with 183 patients receiving HID SCT and
159 receiving ISD SCT. The estimated 9-year overall survival (OS) and failure-free
survival (FFS) were 87.1±2.5% and 89.3±3.7% (P = 0.173) and 86.5±2.6% vs. 88.1±3.8%
(P = 0.257) for patients in the HID and ISD SCT groups, respectively. Transplantation
from HID or ISD SCT has greatly improved quality of life (QoL) levels post-HSCT compared
to pre-HSCT. The occurrence of chronic graft versus host disease was the only identified
adverse factor affecting each subscale of QoL. Physical and mental component summaries
in adults as well as physical, mental, social, and role well-being in children were
all similar between HID and ISD SCT at 5-year points. At the last follow-up, the proportion
of returning to society was comparable between the HID and ISD groups, showing 78.0%
versus 84.6% among children and 74.6% versus 81.2% among adults.
Image:
Summary/Conclusion: These data suggest the recommendation for upfront transplantation
with a haploidentical donor for SAA patients in the absence of an ISD, especially
in experienced transplant centers.
S240: CD7 CHIMERIC ANTIGEN RECEPTOR T CELLS BRIDGING TO ALLOGENEIC HEMATOPOIETIC STEM
CELL TRANSPLANTATION IMPROVED DISEASE-FREE SURVIVAL IN REFRACTORY/RELAPSED T-CELL
ACUTE LYMPHOBLASTIC LEUKEMIA
Z. Li1,*, X. Wang1, X. Wen1, T. Xu1, Y. Song1, T. Wu1
1BEIJING GOBROAD BOREN HOSPITAL, Beijing, China
Background: Our previous clinical study has shown that the patients with refractory/relapsed
T-cell acute lymphoblastic leukemia (r/r T-ALL) have achieved 90% complete remission
(CR) with donor-derived CD7 chimeric antigen receptor T cells (CART), but some cases
have developed donor-recipient mixed chimerism and remarkable pancytopenia (Pan J.
et al. JCO 2021). It is crucial to quickly bridge to allogeneic hematopoietic stem
cell transplantation (allo-HSCT) for hematopoietic reconstitution.
Aims: In current study, the safety and efficacy of allo-HSCT in r/r T-ALL after CD7-CART
are investigated.
Methods: From February 2018 to December 2021, total 57 patients with r/r T-ALL who
underwent allo-HSCT in our hospital were included. The median age was 13(4-69) years
old. Extramedullary disease (EMD) was found in 32 patients (56.1%). Somatic gene mutations
(NOTCH 18, IL7R 3, JAK 2, FBXW7 2, WT1 2, NRAS 2) were detected in 17 patients. Eighteen
patients who were resistant to chemotherapy received CD7-CART (CART group) before
transplant, and all patients except one achieved CR. Thirty-nine r/r T-ALL patients
were managed with chemotherapy before transplant (non-CART group), and 25 patients
achieved CR (CR arm), 14 patients were in non-remission (NR) (NR arm) before transplantation.
Conditioning regimens in CART group were either busulfan/fludarabine (n=12) or TBI/fludarabine
(n=6). Conditioning regimens in non-CART group were either busulfan/fludarabine (n=11)
or TBI/ fludarabine (n=28). ATG was used for haploidentical or unrelated transplants.
Cyclosporine, mycophenolate mofetil and short-term methotrexate were employed for
GVHD prophylaxis.
Results: In CART group, the median time of neutrophils and platelets engraftment were
15 (10-22) and 18 (8-50) days. With the median follow-up 283 (14-496) days, 13 patients
survived and 5 patients died (infection 4, cerebral hemorrhage 1). Three of 18 (16.7%)
patients relapsed (CD7- 1, CD7 + 2) after transplantation and have been receiving
treatment so far. One-year overall survival (OS) and disease-free survival (DFS) were
77.8% and 61.2%. Transplantation-related mortality (TRM) was 16.6%. In non-CART group,
the median time of neutrophils and platelets engraftment were 15 (10-22) and 15 (8-28)
days. Total 25 patients survived and 14 cases died (relapse 3, infection 4, GVHD 1,
multiple organ failure 6). Fifteen of 39 (38.5%) patients relapsed. In CR arm, with
the median follow-up 637(9-1357) days, one-year OS and DFS were 71.0% and 59.2%. In
NR arm, with the median follow-up 517 (7-513) days, one-year OS and DFS were 59.2%
and 40%. TRM was 28.2%.
Summary/Conclusion: Donor-derived CD7-CART followed by allo-HSCT has achieved similar
survival (OS, DFS) and lower relapse rate and TRM compared with chemotherapy followed
by allo-HSCT in r/r T-ALL. CD7-CART may result in better disease control which will
translate into less disease recurrence after transplant.
S241: NON-RESTRICTIVE DIET DOES NOT INCREASE GASTROINTESTINAL INFECTIONS AND FEBRILE
NEUTROPENIA IN PATIENTS WITH NEUTROPENIA AFTER STEM CELL TRANSPLANTATION: DATA FROM
A MULTICENTRE, RANDOMIZED TRIAL
F. Stella1,*, V. Marasco2, G. Levati2, A. Guidetti1, A. Chiappella2, G. Perrone2,
M. Pennisi2, C. Tecchio3, N. Mordini4, G. Ferrara2, G. Gobbi2, L. Saracino2, C. Carniti2,
P. Corradini1 2
1Dept. of Oncology and Hematology, Università degli studi di Milano; 2Division of
Hematology and Bone Marrow Transplant, Fondazione IRCCS Istituto Nazionale dei Tumori,
Milan; 3Department of Medicine, University of Verona, Verona; 4Division of hematology,
Azienda Ospedaliera S. Croce e Carle, Cuneo, Italy
Background: Infections represent a major cause of morbidity and mortality during neutropenia,
especially after hematopoietic stem cell transplantation (HSCT). Multiple preventive
measures, including low microbial diet, have been adopted to reduce these complications.
However, the effect of a protective diet (PD) in this setting has never been explored
prospectively and evidence-based results lack. Conversely, there is evidence that
PD could negatively affect the quality of life and that a prolonged fasting duration
can increase the incidence of acute gastrointestinal graft-versus-host disease (aGVHD)
in allo-HSCT recipients.
Aims: To prospectively compare infection and aGVHD rates in patients receiving a non-restrictive
diet (NRD) compared to PD after HSCT.
Methods: Since July 2016 we conducted a multicentre randomized interventional study
comparing the use of a PD (Arm A) vs NRD (Arm B) in hematological patients aged≥18
years hospitalized to receive autologous or allogenic HSCT. Stratification for allo-HSCT
patients was planned. Patients received the assigned diet during the entire period
of neutropenia. For PD, foods cooked >80°C and/or thick peel fruit were considered
diet-specific. For the NRD, raw fruit and vegetables (adherent to hospital hygiene
standards) were considered diet-specific.
The primary objective was to demonstrate the lack of significant differences in incidence
of infections grade >2 (according to CTCAE 4.0) and deaths during neutropenia between
the two arms.
Secondary endpoints included assessment of gastrointestinal infections and fever of
undetermined origin (FUO), change in body weight, length of hospitalization, overall
survival estimated at 30 days and cumulative incidence of aGVHD within 100 days after
allo-HSCT.
Results: Overall, 162 patients were analyzed at interim analysis, 80 patients in PD
group and 82 in NRD group. The two arms were well balanced in terms of: sex, age,
disease type, number of previous therapeutic lines, disease status at enrollment,
antimicrobial prophylaxis, and reason for hospital admission. Moreover, 32 patients
received an allo-HSCT, 17 in the PD and 15 in the NRD group respectively. Detailed
patients’ characteristics are summarized in Table 1.
We did not observe a significant difference in terms of infections in the two randomized
arms; infections grade >2 or death were reported in 35 (43.7%) patients in arm A and
in 34 (41.5%) patients in arm B [relative risk RR=1.05, confidence interval (CI)95%=0.76-1.4].
The number of patients developing gastrointestinal infections and FUO during hospitalization
was 8 (10%) vs 8 (9.8%) [RR=1.01, CI95%=0.57-1.53] and 32 (40%) vs 28 (34.1%) [RR=1.13,
CI95%=0.82-1.54] in arm A and arm B, respectively. No differences in weight variations
from admission to discharge were observed comparing arm A and arm B (mean 4.15kg vs
3.66kg, p=0.3). Average hospitalization length in the two arms was respectively 20.6
vs 21.5 days (p=0.4). No deaths were reported at day+30. For 32 allo-HSCT recipients,
aGVHD grade ≥2 incidence at day +100 was 5% vs 1.2% in arm A and arm B, respectively
(RR 1.65, CI95%=0.77-2.19).
Image:
Summary/Conclusion: Results of this multicentre prospective trial show similar rate
of infections and deaths between patients receiving a PD versus a NRD during neutropenia
after HSCT. These data suggest that the use of NRD could be considered for transplanted
patients without risks of more infective events.
S242: EARLY INFUSION OF MSCS IN THE PROPHYLAXIS OF GVHD AFTER HAPLO-HEMATOPOIETIC
STEM-CELL TRANSPLANTATION
R. Huang1,*, T. Chen1, S. Wang2, J. Wang3, Y. Su4, J. Liu5, Q. Wen1, P. Kong1, C.
Zhang1, L. Gao1, L. Gao1, X. Zhang1
1Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing,
2920th Hospital of Joint Logistics Support Force of People’s Liberation Army of China,
Sichuan; 3The Affiliated Hospital of Guizhou Medical University, Guiyang; 4The General
Hospital of Western Theater Command PLA, Sichuan; 5The Third Xiangya Hospital of Central
South University, Hubei, China
Background: After the efficiency of repeated infusion umbilical cord-derived mesenchymal
stromal cells (UC-MSCs) in preventing chronic graft versus host disease (GVHD) from
100 days after haploidentical hematopoietic stem-cell transplantation (haplo-HSCT)
was proved in the last trial, we design a sequel clinical trial to further explore
the early repeated infusion of UC-MSCs in GVHD prophylaxis.
Aims: To evaluate the efficacy of repeated infsion of MSCs in GVHD prophylaxis from
D45 after haplo-HSCT.
Methods: 128 qualified subjects aged between 18 and 60 with haplo-HSCT in five transplant
centers in Mid-West of China were enrolled and randomly assigned equally into the
MSCs group and the control group. Patients in the MSCs group received 4 rounds infusion
of UC-MSCs (1×106 cells/kg per two weeks from 45 days after transplantation). We investigated
the incidence of GVHD and relapse free survival (GRFS), the incidence and severity
of aGVHD and cGVHD and the accumulative relapse rate.
Results: The one-year GRFS in the MSCs group is 65.6% in the MSCs group, which is
significantly higher than the control group (43.7%, P= 0.0129). The accumulative incidence
of III-IV aGVHD and severe cGVHD of patients in the MSCs group significantly decreased,
which is 3.1% and 6.3%, while the control group is 25.6% and 18.8% (P=0.0089 and P=0.0260).
The MSCs infusion did not influence the accumulative relapse rate, which is 10.9%
in both groups.
Image:
Summary/Conclusion: In this trial, we proved that early repeated infusion of UC-MSCs
could effectively improve the GRFS rate for patients after haplo-HSCT by decreasing
the incidence and severity of GVHD without increasing the incidences of relapse for
these patients.
S243: ANTIBODY AND T-CELL RESPONSE SIX MONTHS AFTER INITIATION OF BNT162B2 VACCINATION
IN PATIENTS WITH MULTIPLE MYELOMA OR CHRONIC LYMPHATIC B-CELL LEUKEMIA COMPARED TO
HEALTHY CONTROLS
L. D. Heftdal1 2 3,*, L. Pérez-Alós4, S. R. Ham2, J. R. Madsen4, K. Fogh5, C. C. Kronborg2,
A. P. Vallentin1 3, R. B. Hasselbalch5, S. R. Ostrowski6 7, R. Frikke-Schmidt7 8,
E. Sørensen6, L. Hilsted8, H. Bundgaard7 9, K. Iversen5 7 10, P. Garred4 7, S. D.
Nielsen2 7 11, K. Grønbæk1 3 7
1Department of Hematology; 2Department of Infectious Diseases, Rigshospitalet, Copenhagen
University Hospital; 3Biotech Research and Innovation Centre, University of Copenhagen;
4Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631,
Rigshospitalet, Copenhagen University Hospital, Copenhagen; 5Department of Cardiology,
Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev; 6Department
of Clinical Immunology, Section 2034, Rigshospitalet, Copenhagen University Hospital;
7Department of Clinical Medicine, University of Copenhagen; 8Department of Clinical
Biochemistry; 9Department of Cardiology, Rigshospitalet, Copenhagen University Hospital,
Copenhagen; 10Department of Emergency Medicine, Herlev and Gentofte Hospital, Copenhagen
University Hospital, Herlev; 11Department of Surgical Gastroenterology and Transplantation,
Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
Background: The ongoing COVID-19 pandemic has resulted in more than 419 million cases
and more than 5.9 million deaths. Previous studies have indicated inferior responses
to SARS-CoV-2 vaccination across different hematological diseases. Through this prospective
cohort study, we examined the development and durability of anti-receptor binding
domain (RBD) IgG after two doses of BNT162b2 in 179 patients with either multiple
myeloma (MM) or Chronic Lymphatic B-cell Leukemia (B-CLL) six months after vaccination
and compared to immunocompetent controls.
Aims: We aimed to investigate the durability of immune responses to COVID-19 vaccination
in patients with MM or B-CLL compared to healthy controls, and to identify risk factors
for humoral non-response, including type of diagnosis.
Methods: We measured anti-receptor binding domain (RBD) IgG after two doses of BNT162b2
in 179 patients (MM: n=78, B-CLL: n=101) and 179 age and sex matched healthy controls
up to six months after first vaccination. Anti-RBD IgG levels and neutralizing capacity
of antibodies were measured at first and second dose of BNT162b2 and two and six months
after first dose. Humoral response was defined as anti-RBD IgG > 225 AU/mL with a
neutralizing index ≥ 25%. Humoral non-response was defined as the absence of a humoral
response. T-cell responses were assessed six months after the first dose using an
ELISA-based interferon-gamma release assay. A positive T-cell response was defined
as IFN-ɣ release > 200 mIU/mL. Data on diagnoses were obtained through medical records,
and data on vaccination status were obtained from the Danish Vaccination Register.
Results: In patients with MM or B-CLL, the geometric mean concentration (GMC) of anti-RBD
IgG increased from baseline 1.49 AU/mL (95% CI: 1.21-1.84) to three weeks after the
first vaccine dose 15.10 AU/mL (95% CI: 9.39-24.29) and after receiving the second
dose 1179.60 AU/mL (95% CI: 727.78-1919.85). From two to six months after first vaccine
there was a significant decline in the GMC of anti-RBD IgG to 252.75 AU/mL (95% CI:
159.17-403.43). The mean neutralizing capacity in patients with MM or B-CLL was lower
than in controls at all time points after the first vaccine dose. Six months after
first vaccine dose, 79 of 179 (44.1%) patients with MM or B-CLL had a positive humoral
response, while this was the case for 170 of 179 controls (95.0%), p<0.001.
Having MM or B-CLL was significantly associated with risk of humoral non-response.
This was most pronounced in B-CLL patients who had an age and sex adjusted risk ratio
(RR) of 12.25 (95% CI: 6.42-23.38, p< 0.001) of humoral non-response compared to healthy
controls. For MM patients the RR was 4.65 (95% CI: 2.21-9.80, p< 0.001).
T-cell response was assessed in a subset of 48 patients with MM (n=28) or B-CLL (n=20)
and 26 controls, six months after first vaccine dose. A total of 21 (43.8%) patients
with MM (12/28) or B-CLL (9/20) and 14 (53.8%) controls had a positive T-cell response
(p =0.56). Seven of 20 (35.0%) patients with MM or B-CLL who did not develop a humoral
response, developed a T-cell response (MM: 3/8, B-CLL: 4/12), while 14 of 28 (50.0%)
patients with MM or B-CLL who developed a humoral response developed a T-cell response
(p =0.46, MM: 9/11, B-CLL: 5/8). In healthy controls 14 of 25 (56.0%) people who developed
a humoral response also developed a T-cell response.
Summary/Conclusion: Humoral response to BNT162b2 was impaired in patients with MM
or B-CLL compared to healthy controls. Both patients with MM and B-CLL were at higher
risk of humoral non-response compared to healthy controls.
S244: EFFECT OF ANTI-SPIKE NEUTRALIZING MONOCLONAL ANTIBODIES ON COVID-19 PROGRESSION
AND TIME TO VIRAL CLEARANCE IN PATIENTS WITH HEMATOLOGICAL MALIGNANCIES AND SARS-COV-2
INFECTION: THE GIMEMA EXPERIENCE
V. Marasco1,*, A. Guidetti1 2, A. Piciocchi3, A. Candoni4, M. Bocchia5, R. Bruna6,
P. Musto7, A. Visentin8, M. Turrini9, A. Tucci10, C. Selleri11, E. Crea3, P. Fazi3,
F. Passamonti12, P. Corradini1 2
1Hematology and Bone Marrow Transplantation, Istituto Nazionale Dei Tumori di Milano;
2School of Medicine, Università degli Studi di Milano, Milan; 3GIMEMA Foundation,
Rome; 4Dipartimento di Medicina Specialistica, University of Udine, Udine; 5Hematology
Unit, University of Siena, Azienda Ospedaliero Universitaria Senese, Siena; 6Division
of Hematology, Department of Translational Medicine, University of Eastern Piedmont
and Ospedale Maggiore della Carità, Novara; 7Department of Emergency and Organ Transplantation,
“Aldo Moro” University School of Medicine and Unit of Hematology and Stem Cell Transplantation,
AOU Consorziale Policlinico, Bari; 8Hematology and Clinical Immunology unit, Department
of Medicine, University of Padova, Padova; 9Hematology, Ospedale Valduce, Napoli;
10Department of Hematology, ASST Spedali Civili di Brescia, Brescia; 11Hematology,
Ospedale San Giovanni di Dio e Ruggi D’Aragona, Salerno; 12Department of Medicine
and Surgery, niversity of Insubria and ASST Sette Laghi, Ospedale di Circolo of Varese,
Varese, Italy
Background: Patients with hematological malignancies (HM) infected with SARS-CoV-2
have a higher risk of developing severe coronavirus disease (COVID-19) with consequent
death, due to immune system impairment. Anti-spike Neutralizing Monoclonal Antibodies
(nMoAbs) are indicated for the treatment of paucisymptomatic COVID-19 patients, but
evidence of safety and efficacy among HM subjects is still lacking.
Aims: To assess the efficacy of different nMoAbs approved by Agenzia Italiana del
Farmaco (AIFA) on HM patients affected by paucisymptomatic SARS-COV-2.
Methods: Multicenter retrospective observational study at ten sites in Italy, which
enrolled consecutive patients with SARS-CoV-2 infection and treated with nMoAbs from
February 2020 to December 2021. Only HM subjects on treatment or in disease remission
within 6 months from treatment discontinuation with paucisymptomatic SARS-COV-2 infection
were included. nMoAbs approved by AIFA include Bamlanivimab, Bamlanivimab/Etesevimab,
Casirivimab/Imdevimab, Sotrovimab, and Regdanvimab. The primary endpoint was to assess
the time to SARS-CoV-2 molecular swab negativization. A comparison to an historical
control not receiving nMoAbs was assessed. Secondary endpoints consisted in evaluation
of hospitalization rate due to COVID-19, including intensive care unit (ICU) admission
rate due to respiratory failure, and safety assessment.
Results: Overall 51 HM patients (median age 62 years; 35% women) were evaluated. Seventeen
of them had non-Hodgkin lymphomas, 9 multiple myeloma, 6 chronic lymphocytic leukemia,
6 acute myeloid leukemia, 3 Hodgkin lymphoma, 2 acute lymphoblastic leukemia, 2 myeloproliferative
neoplasm, 1 Waldenstrom macroglobulinemia and 5 had other HM diagnosis. Thirty-six
patients were on active treatment, whereas 11 had completed their therapies within
6 months from nMoAbs administration, for 4 patients data were missing. In 7 subjects
the last treatment was chemotherapy, in 19 immunotherapy with or without chemotherapy,
in 9 target therapy, in 4 autologous stem cell transplantation, in 2 allogeneic stem
cell transplantation, for 4 patients data were missing. Detailed description of patients’
characteristics is reported in table 1. Twenty-six patients were treated with Bamlanivimab/Etesevimab,
17 with Casirivimab/Imdevimab, 3 with Bamlanivimab, and 2 with Sotrovimab, for 3 patients
data were missing. Median time to SARS-CoV-2 molecular swab negativization was evaluable
in 41 subjects and was 17 days (min 5, IQR 12-26, max 174). This result compared well
with the previous finding of 28 days reported in an historical group of HM patients
not treated with nMoAbs. We did not find any subpopulation, according to age, diagnosis,
period of infection or type of nMoAbs who achieved a major benefit from nMoAbs treatment.
The rate of hospitalization due to COVID-19 progression was 19% (10/51), with an extremely
low percentage of patients requiring ICU admission due to sever COVID-19 (2%,1/51).
Most frequent side effects included chills (8%), diarrhea (6%), headache (2%), nausea
(2%) and vomiting (2%).
Image:
Summary/Conclusion: Among paucisymptomatic SARS-CoV-2 positive HM patients on active
treatment or in disease remission within 6 months from treatment discontinuation,
the administration of nMoAbs substantially reduced the time to swab negativization
compared to an historical control of HM subjects. This treatment was also able to
reduce the rate of hospitalization and death due to COVID-19 progression in this high
risk group.
S245: ALTERATION OF BONE MARROW NICHE BY ALLOGENEIC IMMUNE REACTION AFTER HSCT
M Kimura1,*, N. Asada2, M. Matsuda1, M. Abe1, W. Kitamura1, K.-I. Matsuoka1, H. Fujiwara2,
M. Ono3, W. Ziyi3, T. Mukai4, Y. Morita5, Y. Maeda1
1Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School
of Medicine, Dentistry and Pharmaceutical Science; 2Hematology and Oncology, Okayama
University Hospital; 3Molecular Biology and Biochemistry, Okayama University Graduate
School of Medicine, Dentistry and Pharmaceutical Science, Okayama; 4Immunology and
Molecular Genetics; 5Rheumatology, Kawasaki Medical School, Kurashiki, Japan
Background: Allogeneic stem cell transplantation (allo-SCT) has been considered as
a curable therapy for hematologic diseases. Post-transplant myelosuppression is one
of the life-threatening complications. Hematopoietic stem cell (HSC) functions are
tightly regulated by a specialized microenvironment called “niche” in the bone marrow
(BM) and we have identified differential contributions of perivascular stromal cells
as HSC niche (Asada et al. Nat Cell Biol 2017). Recent studies indicate that BM niche
can be targeted by allo-immune reaction but it remains unclear how the perivascular
niche cells are involved in the mechanism of HSC dysfunction after allo-SCT.
Aims: In this study, we aim to investigate the mechanisms of the impairment of the
niche cells by allo-immune reactions, and to explore a targetable pathway for myelosuppression
after allo-SCT.
Methods: To evaluate the alterations in niche cells after allo-SCT, we utilized allo-SCT
mice models (C3H/HeJ into C57BL/6, C57BL/6 into B6D2F1). Hematopoietic cells, including
HSCs and mature cells, and perivascular stromal cells in BM were analyzed by flow
cytometry (FACS) after transplantation. In imaging studies, we visualized perivascular
stromal cells using Nestin-GFP (Nes-GFP), Myh11-CreER/tdTomato, LeptinR-Cre/tdTomato
transgenic mice and observed them with confocal laser scanning microscopy. The gene
expression analysis of niche factors was performed by real-time PCR. To assess the
landscape of transcriptional remodeling in BM after allo-SCT, we performed single-cell
RNA sequencing (scRNAseq) of BM cells.
Results: FACS analyses showed that Nes-GFP+ stromal cells were dramatically reduced,
and HSC recovery was severely impaired in allo-SCT mice compared to control mice at
day 21 after SCT. Imaging studies also revealed that perivascular stromal cells are
morphologically damaged in allo-SCT mice. Nes-GFP+ stromal cells in allo-SCT mice
presented a decreased expression of niche factors, including Cxcl12 and Scf, which
are essential for HSC maintenance, indicating the dysfunction of niche cells. We confirmed
the impaired niche function in allo-SCT mice by tandem transplantation analysis in
vivo, in which the recovery of syngeneic HSCs was also impaired in damaged microenvironment
in allo-SCT mice. Prevention of allo-reactive T cell expansion by post-transplant
cyclophosphamide ameliorated the niche cell reduction along with improved recovery
of HSCs. These results indicate that niche impairment caused by allo-immune reaction
leads to HSC dysfunction. The scRNAseq of BM highlighted the elevated expression levels
of inflammation response genes, including Cxcl9 and Cxcl10, and MHC classⅡ, together
with the deceased expression of niche factors in niche cells in allo-SCT mice. These
alterations seemed to be induced by proinflammatory cytokines such as interferon-gamma
(INF-γ) derived from allo-T cells, as the increased number of T cells and high levels
of INF-γ in BM were observed in allo-SCT mice. Additionally, evaluation of T cell
localization by using imaging techniques of BM revealed that allo-T cells reside significant
closer to LepR+ niche cells in allo-SCT mice. Since activation of CXCR3, which is
a receptor for chemokines Cxcl9 and Cxcl10, has been shown to induce chemotaxis of
activated T cells, we treated allo-SCT mice with AMG487, an antagonist of CXCR3, resulted
in an improved recovery of both perivascular niche cells and hematopoiesis in allo-SCT
mice.
Summary/Conclusion: Collectively, allo-immune reaction severely damages perivascular
HSC niche cells, leading to impaired recovery of hematopoiesis after allo-SCT.
S246: A HUMAN BONE MARROW ORGANOID FOR DISEASE MODELLING AND DRUG SCREENING IN BLOOD
CANCERS
A. Khan1 2,*, M. Colombo2, J. Reyat1, G. Wang2 3, A. Rodriguez-Romera2, W. X. Wen2
3, L. Murphy2, B. Grygielska1, C. Mahoney4, A. Stone5, A. Croft4, D. Bassett6, G.
Poologasundarampillai7, A. Roy8, S. Gooding8, J. Rayes1, K. Machlus5, B. Psaila2
1Institute of Cardiovascular Sciences, University of Birmingham, Birmingham; 2MRC
Weatherall Institute of Molecular Medicine; 3Centre for Computational Biology, MRC
Weatherall Institute of Molecular Medicine, University of Oxford, Oxford; 4Rheumatology
Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham,
United Kingdom; 5Vascular Biology Program, Boston Children’s Hospital, Department
of Surgery, Harvard Medical School, Boston, United States of America; 6Healthcare
Technologies Institute, School of Chemical Engineering; 7School of Dentistry, Institute
of Clinical Sciences, University of Birmingham, Birmingham; 8MRC Weatherall Institute
of Molecular Medicine, Department of Paediatrics and National Institute of Health
Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United
Kingdom
Background: Blood cancers are the most common cancer affecting children, and among
the top 10 most common cancers in adults, in whom they remain largely incurable. In
the era of advanced “omics”, a major bottleneck to the development of novel therapies
is target prioritization and validation. There is a pressing need for experimental
platforms that model the cellular and molecular complexity of the bone marrow niche,
to enable mechanistic studies into the role of the bone marrow microenvironment in
the initiation and propagation of blood cancers.
Aims: Organoid technologies have been transformative in other settings, but an organoid
with adequate homology to native haematopoietic tissues has not yet been reported.
We therefore sought to develop organoids that emulate the cellular, molecular and
spatial architecture of human bone marrow.
Methods: A directed-differentiation protocol was devised in which human iPSCs formed
mesodermal aggregates before hydrogel embedding and commitment to vascular and hematopoietic
lineages. Multi-modal imaging and single cell RNA-sequencing were used to confirm
cellular, molecular and architectural homology to native human hematopoietic tissues.
Results: We optimized hydrogel compositions and cytokine cocktails to support the
differentiation of a 3D bone marrow perivascular niche. The resulting organoids contained
distinct haematopoietic stem/progenitor cells (HSPCs), stromal and myeloid cellular
subtypes including pro-platelet forming megakaryocytes and lumen-forming vasculature.
3D whole-organoid imaging revealed a vascular network with sinusoid-like vessels invested
with MSCs/fibroblasts (Fig1A), and the extravasation of haematopoietic cells into
vessel lumens. Megakaryocytes closely associated with vessels, and extended pro-platelet
protrusions into the vasculature with remarkable similarity to native bone marrow.
scRNAseq analysis confirmed transcriptional homology to human bone marrow cellular
subtypes, and highlighted strong cell-cell communication networks and haematopoietic
support from organoid stroma. We first explored the utility of the organoids to model
myelofibrosis, an exemplar malignancy that involves cancer-induced bone marrow remodelling
and where there is a need for improved models for target prioritization and novel
therapies. Treatment of organoids with TGFβ resulted in a dose-dependent increase
in hallmarks of fibrosis (Fig 1B), which was effectively reversed using pharmacological
agents, enabling drug screening. Given their homology to native bone marrow, we hypothesized
that the organoids may support engraftment of primary human cells. Indeed, cells from
healthy donors and patients with blood cancers efficiently engrafted throughout the
organoids. After 14 days, organoids engrafted with myelofibrosis but not healthy cells
showed dramatic fibrotic remodelling (Fig 1C). The organoids also supported primary
cells from other myeloid and lymphoid blood cancers, including those notoriously difficult
to maintain in vitro – e.g. myeloma cells engrafted into organoids were >90% viable
at 10 days following seeding, while rapidly dying within 48 hrs without organoid support.
The ability to study primary cells ex vivo overcomes a huge hurdle to translational
research in the myeloma field.
Image:
Summary/Conclusion: This platform is an enabling technology for the interrogation
of disease mechanisms in haematological disorders. Target identification and screening
using a species-specific, 3D model that can incorporate primary patient cells will
reduce dependence on animal models and may accelerate translational research.
S247: EMERGENCY GRANULOPOIESIS INDUCES A LYMPHOID TO MYELOID BIAS SWITCH IN A SUBSET
OF HEMATOPOIETIC STEM CELLS
K. Vanickova1 2,*, P. Danek1, M. Milosevic3, M. K. Adamcova1 4, N. Rai5, F. Matteini6
7, M. C. Florian6 7 8, J. D’Armiento9, J. Rohlena3, K. Rohlenova3, M. Alberich-Jorda1
4
1Department of Hemato-oncology, Institute of Molecular Genetics of the CAS, Prague;
4; 2Faculty of Science, Charles University; 3Institute of Biotechnology of the Czech
Academy of Sciences; 4Childhood Leukaemia Investigation Prague, Department of Pediatric
Haematology and Oncology; 2nd Faculty of Medicine, Charles University in Prague, University
Hospital Motol, Prague, Czechia; 5Department of Pediatrics, Columbia University, New
York, United States of America; 6Stem Cell Aging Group, Regenerative Medicine Program,
The Bellvitge Institute for Biomedical Research (IDIBELL); 7Program for Advancing
the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], Barcelona;
8Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine
(CIBER-BBN), Madrid, Spain; 9Department of Anesthesiology, Columbia University, New
York, United States of America
Background: Granulocytes represent the first line of defense against bacteria and
fungi, thus their production needs to be adapted to specific demands. While daily
production of granulocytes is sustained by steady-state granulopoiesis, upon infection
this program switches to emergency granulopoiesis (EG), to ensure enhanced and accelerated
granulocytic production. Recently, it was shown that unperturbed hematopoiesis is
sustained by multipotent progenitors, however, during stress conditions, hematopoietic
stem cells (HSCs) become active and responsible for coping with stress situations.
Yet, whether and how HSCs play a role during EG is unknown.
Aims: The aim is to understand the role of HSCs at early stages of acute infection,
and to determine the mechanisms that drive emergency granulopoiesis at the stem cell
level.
Methods: The methods used in the project include single cell RNA sequencing, qRT-PCR,
ELISA, whole-mount bone marrow microscopy, flow cytometry sorting and analysis, stem
cell in vitro cultures, Wnt10b knockout mouse model and murine LPS administration.
Results: To understand the contribution of HSCs to the EG response, we performed single
cell RNA sequencing analysis of sorted murine HSCs (Lin-c-Kit+Sca-1+CD48-CD150+) 4
hours after lipopolysaccharide (LPS) stimulation, which mimics a bacterial infection
and activates EG in vivo. Strikingly, we observed radical changes in the HSC cluster
composition between PBS control and LPS treated mice. Interestingly, these changing
population dynamics were marked by alterations in HSC lineage bias, demonstrating
that under EG there is an expansion of the myeloid-bias HSCs at expenses of the lymphoid-bias
HSCs. Remarkably, we identified Procr (CD201) as a specific cell surface marker for
this lymphoid to myeloid transition, allowing us to distinguish, sort and track the
switch from CD201+ lymphoid-bias HSCs (present in PBS control) towards CD201- myeloid-bias
HSCs (present upon LPS stimulation). Further, the abundant CD201- myeloid-bias HSC
clusters were characterized by an inflammatory gene signature. Particularly interesting,
was the elevated expression of Wnt10b. We verified that Wnt10b is upregulated in murine
primary bone marrow cells upon LPS stimulation in vitro using qRT-PCR. Further, we
demonstrated that Wnt10b is released in the bone marrow following LPS treatment in
vivo using ELISA and whole-mount microscopy. We showed that Wnt10b activates the canonical
Wnt/β-catenin signaling pathway and promotes myeloid differentiation of Lin-c-Kit+Sca-1+
cells in vitro. In addition, our preliminary data suggests that the EG response is
impaired in Wnt10b KO mice following LPS stimulation in vivo.
Summary/Conclusion: In conclusion, we observed that HSCs actively take part in early
stages of EG by activating an inflammatory signature associated with the switch from
lymphoid to myeloid bias, therefore ensuring sufficient production of myeloid cells
to fight the infection.
Acknowledgements: This work was partially supported by a GACR grant 22-18300S and
institutional funding from the IMG CAS (RVO 68378050).
S248: THE ENDOSOMAL ADAPTOR PROTEIN MYCT1 CONTROLS ENVIRONMENTAL SENSING IN HUMAN
HSCS
J. Aguade-Gorgorio1,*, Y. Jami-alahmadi1, M. Kardouh1, I. Fares1, V. Calvanese2, H.
Johnson1, M. Magnusson3, J. Shin4, H. Goodridge4, J. Wohlschlegel1, H. Mikkola1
1UCLA, Los Angeles, United States of America; 2University College London, London,
United Kingdom; 3Lund University, Lund, Sweden; 4Cedars Sinai, Los Angeles, United
States of America
Background: Hematopoietic stem cells (HSC) integrate diverse environmental cues to
maintain the balance between quiescence, self-renewal and differentiation throughout
life. However, the molecular programs governing HSC stemness become dysregulated during
culture, compromising HSC self-renewal and engraftment ability. Despite recent advances
in optimizing culture conditions for human HSC, our ability to expand or modify functional
human HSCs in culture for therapeutic use is still limited. Our data uncovered MYCT1
(Myc target 1) as a critical human HSC regulator that is selectively expressed in
undifferentiated HSPCs and endothelial cells (EC) but becomes drastically downregulated
during culture, concomitantly with loss of engraftment potential.
Aims: We aimed to understand the molecular programs controlled by MYCT1 that are essential
for maintaining human HSC function.
Methods: We analyzed multiple RNAseq gene expression data sets of cultured and uncultured
human HSPCs from developmental and postnatal hematopoietic tissues to identify genes
associated with self-renewing HSCs. To assess the importance of MYCT1 expression in
human HSC function we transduced fetal liver (FL) and cord blood (CB) HSPCs (CD34+CD38-CD90+
and GPI80+ in FL) with lentiviral shRNA vectors. We quantified the expansion of control
or knockdown (KD) HSPCs at distinct time points, and determined engraftment ability
at 6, 12, and 24 weeks after transplanting equal numbers of sorted HSPCs into conditioned
immunodeficient mice.
To define MYCT1 localization and interactome we performed subcellular fractionation,
immunofluorescence, and immunoprecipitation coupled with high-sensitivity mass spectrometry
in endothelial cells and the HSC-like KG1 cell line. We assessed endocytosis by quantifying
the internalization of fluorescently labeled dextran and transferrin by FACS in human
HSPCs and ECs after MYCT1 KD or overexpression (OE). To determine the signaling and
cellular consequences of MYCT1 loss we performed phospho-proteomics, western blot,
and single cell RNAseq in ECs and/or human HSPCs. HSPC proliferation was quantified
after MYCT1 KD or OE by CFSE dilution assay and monitoring of single cell divisions
for 5 days.
Results: MYCT1 KD experiments revealed that MYCT1 is critical for human HSPC ex vivo
expansion and engraftment after transplantation. We determined that MYCT1 is a membrane-associated
protein localized in endosomes, where it interacts with vesicle trafficking components
and signaling receptors with critical functions in HSC biology. MYCT1 KD in human
ECs and HSPCs led to hyperactivation of endocytosis, a crucial regulatory step determining
the responsiveness to extracellular cues and governing cell fate decisions. MYCT1
KD then led to widespread dysregulation of signaling pathways, including a hyperactivation
Akt, and cellular functions, including defective proliferation, whereas MYCT1 overexpression
had the opposite effect
Summary/Conclusion: Our data suggest that MYCT1 governs human HSC stemness and fate
decisions by controlling the sensing of extracellular signals and their downstream
signaling through endocytosis. As MYCT1 expression is downregulated in cultured HSPC,
this work suggests that the inability to properly sense microenvironmental signals
in cultured HSCs is a key mechanism contributing to culture-associated HSC dysfunction
that will need to be overcome to improve transplantability of ex vivo expanded human
HSCs.
S249: EX VIVO HEMATOPOIETIC STEM CELL (HSC) EXPANSION USING BONE-LINING REINVIGORATING
MESENCHYMAL STROMAL CELLS (RMSCS)
S. Sood1 2,*, L Klein1, P. Albert1, F. Pilz1, N. Schmitt3, D. Nowak3, M. Essers1 2
1Inflammatory Stress in Stem Cells; 2HI-STEM, DKFZ, Heidelberg; 3Department of Hematology
and Oncology, Heidelberg University, Mannheim, Germany
Background: Bone marrow transplants (BMTs) have highlighted the Hematopoietic Stem
Cell (HSC) potential to restore a new functional hematopoietic system in diseased
recipients. However, a major roadblock for the clinical application and translational
research on HSCs is our limited potential for ex vivo HSC expansion.
Aims: To develop an approach for improved ex vivo HSC expansion using novel bone-lining
reinvigorating Mesenchymal Stromal Cells (rMSC)-based culture system.
Methods: Using a functional approach, we developed a robust pipeline for the fluorescence-activated
cell sorting (FACS)-based isolation and ex vivo expansion of rMSCs from both murine
and patient-individualized human samples. We tested the long-term HSC expansion potential
in the rMSC-based co-culture system, phenotypically using FACS based proliferation
analysis and functionally using limiting dilution and colony forming assays. Further,
we performed transplantation experiments to study the hematopoietic reconstitution
ability of the ex vivo expanded HSCs. Moreover, we expanded a single HSC over multiple
cell divisions using our culture system to demonstrate bona fide HSC self-renewal
potential of the rMSC-based system.
Results: We propose a potent ex vivo HSC expansion system based on novel bone lining-derived
reinvigorating Mesenchymal Stromal Cells (rMSCs). Both bulk- and single-HSCs expanded
long-term using the rMSC co-culture system maintained phenotypic stemness over multiple
cell differentiation cycles and possessed functional bone marrow reconstitution capabilities
upon transplant. Notably, our rMSC co-culture system outperformed existing alternatives
for HSC expansion including systems using stromal cells, non-cellular coating factors,
or different medium compositions. Further, our results highlight the reliable isolation
and robust culture of human rMSCs using our experimental strategy, and the potential
to also utilize the rMSC co-culture system for the ex vivo expansion of human HSCs.
Summary/Conclusion: We could demonstrate that our rMSC-based system for HSC expansion
can play a pivotal role in research to reduce the number of mice used for ex vivo
experiments. Moreover, our data also shows that the rMSC-based co-culture can be used
for human HSC expansion, opening possibilities of applying this potent system for
research on numerous diseases including immunodeficiencies and leukaemia.
S250: CHOLINERGIC SIGNALS PROMOTE THE QUIESCENCE OF NORMAL OR LEUKAEMIC STEM CELLS
THROUGH THE ACTIVATION OF ALPHA 7-NICOTINIC RECEPTOR IN BONE MARROW MESENCHYMAL STROMAL
CELLS
C. Fielding1 2 3,*, A. García-García1 2 3, C. Korn1 2 3, S. Gadomski1 2 3 4 5, Z.
Fang1 2 3, C. Kapeni1 2 3, J. L. Reguera6, J. A. Pérez-Simón6 7, B. Göttgens1 3, S.
Méndez-Ferrer1 2 3 8 9
1Department of Haematology, University of Cambridge; 2National Health Service Blood
and Transplant, Cambridge Biomedical Campus; 3Wellcome-MRC Cambridge Stem Cell Institiute,
University of Cambridge, Cambridge, United Kingdom; 4Skeletal Biology Section, National
Institute of Dental and Craniofacial Research, Bethesda; 5NIH-OxfordCambridge Scholars
Program in partnership with Medical University of South Carolina, Charleston, United
States of America; 6Department of Haematology, University Hospital Virgen del Rocio,
41013 Sevilla, Spain; 7National Health Service Blood and Transplant, NIH-OxfordCambridge
Scholars Program in partnership with Medical University of South Carolina, Charleston,
United States of America; 8Instituto de Biomedicina de Sevilla (IBiS/CSIC); 9Departamento
de Fisiología Médica y Biofísica, Universidad de Sevilla, 41013 Sevilla, Spain
Background: Our recent research has suggested that the sympathetic nervous system,
a master regulator or homeostasis and stress responses, regulates the activity of
hematopoietic stem cells (HSCs) in different bone marrow niches. We recently showed
that chemotherapy or irradiation, which are routinely used for cancer treatment or
as conditioning regimens for HSC transplantation, increase the release of cholinergic
signals by sympathetic neurons and osteolineage cells. As a result, not all HSCs become
activated (which could cause their exhaustion), but some remain quiescent, thereby
retaining their full potential. Cholinergic signals activate α7 nicotinic receptor
(α7nAChR) in bone marrow mesenchymal stromal cells (BMSCs), which in turn preserve
stem cell quiescence under proliferative stress (Nat Commun 13:543). In addition,
α7nAChR signalling exhibits anti-inflammatory properties (PMID:32595942). However,
whether this signalling pathway influences acute myeloid leukaemia (AML) development
and response to treatment is unclear.
Aims: Investigate the influence of cholinergic signals on leukaemogenesis and therapy
response in AML.
Methods: Analysis of bone marrow (BM) from mouse models lacking sympathetic cholinergic
innervation or α7nAChR in BMSCs, transplanted with BM cells from an inducible MLL-AF9+
AML model (Cancer Cell 15:791) and treated with cytarabine. Flow cytometry, histology,
FACS, qRT-PCR.
Results: We confirmed reduced density of sympathetic noradrenergic innervation in
the BM of AML mice; however, interestingly, the cholinergic innervation, which also
has sympathetic origin (Cell Stem Cell, in press) was preserved. To investigate the
role of these fibres in AML development, we utilised a mouse model lacking sympathetic
cholinergic innervation due to loss of the neurotrophic receptor GFRα2. AML developed
faster in cholinergic-neural-deficient mice, compared with WT mice, transplanted with
iMLL-AF9+ BM cells. This was associated with reduced quiescence of leukaemia-initiating
cells (LIC) upon acute cytarabine treatment of mice lacking cholinergic skeletal innervation.
This effect was more pronounced after repeated cytarabine treatment, leading to increased
proliferation of LICs and augmented leukaemia burden at relapse.
Given that α7nAChR transduces the cholinergic regulation of normal HSCs by BMSCs,
we utilised LepR-Cre;Chrna7f/f mice to conditionally delete this signalling pathway
in leukaemic mice. Reproducing the observations in mice lacking cholinergic innervation,
α7nAChR deletion in BMSCs accelerated AML development, and repeated treatment with
cytarabine further increased LIC proliferation, spleen infiltration and leukaemia
burden after relapse.
Summary/Conclusion: These results suggest that cholinergic signals increase the quiescence
of normal or leukaemic stem cells through the activation of a7-nicotinic receptor
in BMSCs and might influence AML response to chemotherapy.
S251: DISRUPTION OF SUCCINATE RECEPTOR SIGNALING PROMOTES MYELOPROLIFERATION AND ACUTE
MYELOID LEUKAEMIA
V. Cuminetti1,*, J. Konieczny1, A. Bernal1, A. Villatoro1, H. Taman2, M. Ristic1,
N. Villaplana-Lopera3, R. H. Paulssen2, G. Giovinazzo4, A. Vik5, P. Garcia3, L. Arranz1
1IMB, Stem Cells Ageing and Cancer; 2Genomics Support Center Tromsø, UiT The Arctic
University of Norway, Tromsø, Norway; 3Institute of Cancer and Genomic Sciences, University
of Birmingham, Birmingham, United Kingdom; 4Pluripotent Cell Technology Unit, Centro
Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; 5Department
of Hematology, University Hospital of North Norway, Tromsø, Norway
Background: Unlike healthy haematopoietic stem cells (HSC), leukaemia stem cells (LSC)
have high oxidative phosphorylation and hence targeting the tricarboxylic acid (TCA)
cycle seems a promising therapeutic strategy in acute myeloid leukemia (AML) treatment.
Accumulation of the TCA cycle oncometabolite succinate is associated with tumourigenesis,
but its potential involvement in HSC function and leukaemia is unkown. Succinate stabilizes
the oxygen-sensing transcription factor hypoxia-inducible factor (HIF)-1-α and activates
interleukin (IL)-1β production in macrophages. Succinate may also be exported and
has cell extrinsic effects through its receptor, succinate receptor-1 (SUCNR1), which
may be pro- or anti-inflammatory.
Aims: To investigate the in vivo role and molecular mechanisms of succinate and its
receptor in normal and malignant haematopoiesis.
Methods: To study the effect of succinate receptor on haematopoiesis, we characterized
the immunophenotype of Sucnr1-KO mice. To dissect the role of Sucnr1 on healthy haematopoiesis
via cell-autonomous effects, we transplanted fl/Sucnr1/fl control or Mx1-Cre fl/Sucnr1/fl
bone marrow (BM) cells into wild-type (WT) C57BL/6J mice, which were later injected
with pIpC. To study the role of Sucnr1 through non-cell autonomous effects, we transplanted
WT BM into Nes-Cre
ERT2
fl/Sucnr1/fl recipients or fl/Sucnr1/fl controls, which were induced with tamoxifen.
We characterized the effects of Sucnr1 deletion at the molecular level by bulk and
single-cell RNA-sequencing (scRNA-seq), and aimed to uncover the molecular mechanisms
mediating the effects of Sucnr1 ablation using in vivo tageting with drugs. To study
the effect of succinate on leukaemic progression, we used two different mouse models,
i.e. Mx1-Cre Nras
G12D
and MLL-AF9 knock-in (KI). In human AML, we quantified SUCNR1 expression in peripheral
blood mononuclear cells (PBMC) from AML patients versus healthy volunteers, and treated
xenograft recipients of CD34+ AML cells with succinate or vehicle.
Results:
In vivo deletion of Sucnr1 induced myeloid bias and expansion of the haematopoietic
stem and progenitor cell (HSPC) compartment. In vivo deletion of Sucnr1 from Nestin+
stromal cells promoted expansion of long-term HSC (LT-HSC) versus intact recipients.
Conversely, in vivo deletion of Sucnr1 from the haematopoietic system induced expansion
of multipotent progenitors (MPP) versus intact haematopoietic cells. RNA-seq in HSPC
from Sucnr1-KO versus WT mice showed changes in genes associated with myeloid output,
and pinpointed related pathways potentially involved. scRNA-seq confirmed expansion
of LT-HSC in Sucnr1-KO versus WT mice. In vivo treatments targeted signalling pathways
at least partially responsible for the haematopoietic abnormalities in Sucnr1-KO.
We found higher content of succinate in the BM extracellular fluid of leukaemic Mx1-Cre
Nras
G12D
. Succinate injection to xenograft recipients of CD34+ AML cells increased leukaemic
output versus vehicle, promoted myeloid bias in Mx1-Cre Nras
G12D
mice and reduced survival in mice transplanted with BM of MLL-AF9 leukaemic donors.
PBMC from AML patients expressed lower level of SUCNR1 versus healthy donors and BM
nucleated cells from Mx1-Cre Nras
G12D
also expressed less Sucnr1 than their healthy counterparts.
Summary/Conclusion: We show a novel role for succinate and its receptor in the haematopoietic
system and pinpoint this pathway as a novel means of communication within the BM HSC
microenvironment, whose disruption may be involved in myeloid malignancy. These findings
may help design new therapeutic strategies for AML patients.
S252: M2-POLARIZED MACROPHAGES CONTROL LEUKEMIC STEM CELL FATE BY PROMOTING METABOLIC
REPROGRAMMING
I. Weinhäuser1 2 3,*, D. A. Pereira-Martins1 2 3, J. R. Hilberink1, L. Y. Almeida2
3, D. R. A. Silveira4, L. Quek4, S. M Hogeling1, J. M. Mota5, E. Ammatuna1, A. R.
Lucena-Araujo6, G. A. Huls1, E. M. Rego7, J. J. Schuringa1
1Experimental Hematology, University medical centre groningen, Groningen, Netherlands;
2Center for Cell Based Therapy, University of Sao Paulo; 3Internal Medicine, Medical
School of Ribeirao Preto, Ribeirao Preto, Brazil; 4Myeloid Leukaemia Genomics and
Biology Group, Kings College London, London, United Kingdom; 5Medical Oncology Service,
Sao Paulo State Cancer Institute, Sao Paulo; 6Department of Genetics, Federal University
of Pernambuco, Recife; 7Center for Cell Based Therapy, University of Sao Paulo, Sao
Paulo, Brazil
Background: Being a crucial part of the tumor microenvironment (TME) in solid tumors,
tumor-associated macrophages are often associated with poor prognosis (Bruni et al.
Nat Rev Cancer 2020). Yet, in acute myeloid leukemia (AML) the role of macrophages
remains unclear.
Aims: Here, we evaluated the impact of M2 macrophages in AML using a patient derived
xenograft (PDX) model.
Methods: To do so, we first injected 1x105 peripheral blood (PB) derived M2 macrophages
into NSGS mice and next transplanted notoriously difficult to engraft primary Acute
Promyelocytic Leukemia (APL) cells (1 x 106 cells - n=7 different patient samples)
via the retro-orbital vein.
Results: As a result, mice with co-injected human M2-macrophages developed full-blown
leukemia with increased spleen weight in comparison with the control. Perhaps even
more strikingly, ex vivo culture of APL and other favorable AML subtypes such as inv
(16) or NPM1 mutant leukemic cells on M2-macrophages for 48h was sufficient to “train”
these cells to engraft and induce fatal leukemia. Maintenance of self-renewal was
shown in a secondary transplant assay and an enhanced frequency of leukemic stem cells
was assessed by in vivo LTC-IC assays (LSC frequency: Control: 1/6x106 cells vs M2
pre-culture: 1/7.2x104 cells).
To better understand the biological changes induced on leukemic blast when exposed
to M2 macrophages, we performed an RNA sequencing analysis comparing AML/APL samples
at diagnosis to cells that were “trained” (48h) on M2-macrophages or on MS5 mesenchymal
bone marrow stromal cells, as a control. Gene ontology and gene set enrichment analysis
on the genes up regulated upon M2 co-culture were significantly enriched for oxidative
phosphorylation (OxPhos) signatures. Evaluation of functional respiration using seahorse
measurements, confirmed the increase of oxygen consumption rate (OCR, basal and maximum)
in primary AML/APL cells (n=7) after exposure to M2 macrophages compared to MS5. The
increase of basal and maximum OCR suggested enhanced mitochondrial metabolism, which
prompted us to determine whether macrophages, similar to MSC cells, can transfer mitochondria
to primary AML cells (van der Vlist et al. Neuron 2022). Flow cytometry analysis revealed
an efficient mitochondrial transfer from M2 macrophages to leukemic blasts, which
was more efficient compared to mitochondrial exchange from MS5 cells. Treatment with
the CPT1A inhibitor Etomoxir (50 µM), prevented the gain in functional respiration
and enhanced proliferation of AML cells when those were co-cultured on M2-macrophages,
while no changes were observed for MS5 co-cultures. These results suggest increased
fatty acid oxidation to drive the OXPHO-like state in AML.
Summary/Conclusion: Overall, we revealed that M2 macrophages can support leukemic
growth of favorable AML subtypes that are commonly difficult to engraft in PDX models.
Even an in vitro exposure to M2 macrophages suffices to alter the biology of AML cells
transforming a non-engraftable cell into a cell with high leukemic potential. In vitro
we show that AML blast cells cultured on M2 macrophage adapt a more OxPhos-like state
linked to intrinsic changes associated with increased uptake of mitochondria. In summary,
our study uncovers how the TME can contribute to leukemic transformation which provides
alternative avenues for therapeutic interventions.
S253: THE SHEDDASE DOMAIN OF ADAM10 AUGMENTS THE INTERACTION OF LEUKEMIA CELLS WITH
THE BONE MARROW NICHE IN VIVO AS SHOWN BY RECONSTITUTING PDX LEUKEMIA CELLS WITH CRISPR-CAS9-INDUCED
KNOCKOUT
J. P. Schmid1 2,*, E. Bahrami1, M. Becker1, A. K. Jayavelu3, A.-K. Wirth1, V. Jurinovic1
4 5, R. Öllinger6 7 8, R Rad2 6 7 8, B. Vick1 2, M. Mann3, T. Herold1 5, I. Jeremias1
2 4
1Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German
Research Center for Environmental Health (HMGU); 2German Cancer Consortium (DKTK),
partner site Munich, Munich; 3Department of Proteomics and Signal Transduction, Max
Planck Institute of Biochemistry, Martinsried; 4Department of Pediatrics, University
Hospital, Ludwig Maximilian University (LMU); 5Laboratory for Leukemia Diagnostics,
Department of Medicine III, University Hospital, LMU Munich; 6Center for Translational
Cancer Research (TranslaTUM), TUM School of Medicine, Technische Universität München;
7Department of Medicine II, Klinikum rechts der Isar, Technische Universität München;
8Institute of Molecular Oncology and Functional Genomics, Technische Universität München,
Munich, Germany
Background: Tumor-microenvironment interactions are critically important determinants
contributing to leukemia formation and maintenance. Interrupting the leukemia-bone
marrow interaction represents an attractive therapeutic approach in acute leukemia
(AL). Functional genomics significantly increases our understanding of the vulnerabilities
and gene dependencies of individual tumors.
Aims: Here, we developed a CRISPR-Cas9 screening approach for functional analysis
of surface molecules in patient-derived xenograft (PDX) AL models in vivo.
Methods: Size of CRISPR library was determined by genetic barcoding. Stable expression
of fluorescently labelled Cas9 and sgRNA constructs in two PDX samples. Enrichment
of double positive cells by MACS and injection into NSG mice. Gene depletion analysis
using MAGeCK algorithm to screen and functional competitive in vivo assays to validate
the candidates. Characterization of the ADAM10 KO or inhibitor (GI254023X) treated
cells for engraftment capacity by homing assay, frequency of leukemic stem cells by
competitive limiting dilution transplantation assay (LDTA), sensitivity towards routine
chemotherapy by in vivo competitive chemotherapy trials in both lineages. Rescue assay
by reconstitution of ADAM10 variants in functional competitive in vivo assays.
Results: When running a customized CRISPR-Cas9 screen targeting about 100 cell surface
candidates in two AL PDX samples, several sample-specific, but also commonly depleted
candidates were identified. CRISPR screen findings were confirmed on the level of
single molecules, using a competitive molecular in vivo approach and testing the PDX
cells with and without knockout in the same mouse. These experiments validated an
essential function for the two well-known depleted candidates CXCR4 and ITGB1 in both
PDX models in vivo. Of note, various members of the Solute Carrier Family (SLC) were
among the list of drop-out candidates. ADAM10 was identified as a commonly depleted
candidate in both PDX models. In vivo competitive experiments confirmed the essential
role of ADAM10 in PDX models from 6 additional patients with either acute lymphoblastic
leukemia (ALL) or acute myeloblastic leukemia (AML), indicating a broad essential
role of ADAM10 in both, ALL and AML, independent from their oncogenic-driver mutations
and chromosomal abnormalities. Moreover, treating PDX cells with an ADAM10 chemical
inhibitor resulted in significantly reduced engraftment capacity into the bone marrow
(BM), indicating a role for ADAM10 in the early engraftment and homing process in
the BM microenvironment. Knockout of ADAM10 reduced the frequency of leukemia stem
cells, indicating that a relevant fraction of stem cells depends on ADAM10. ADAM10
KO ALL and AML PDX samples showed increased sensitivity towards routine chemotherapy
treatments, indicating that inhibition of ADAM10 sensitizes AL towards conventional
chemotherapy. When ADAM10 knockout cells were reconstituted with different recombinant
ADAM10 variants, PDX in vivo experiments revealed that wildtype ADAM10 rescued the
phenotype, while an ADAM10 variant lacking the enzymatic domain did not, highlighting
the importance of the sheddase activity for ADAM10 function in leukemia maintenance.
Summary/Conclusion: In summary, we established CRISPR-Cas9 drop-out screens in PDX
models in vivo as technology to explore patient-specific tumor dependencies. Our data
revealed a yet unknown function of ADAM10 to maintain patient leukemic cells in the
bone marrow microenvironment niche. ADAM10 thus represents an attractive future therapeutic
target for the treatment of acute leukemia.
S254: CD4+ T CELL-DERIVED IL21 REGULATES STEM CELL FATE IN ACUTE MYELOID LEUKEMIA
V. Rubino1 2 3,*
1Department for BioMedical Research, University of Bern; 2Department of Medical Oncology,
Inselspital, Bern University Hospital; 3Graduate School of Cellular and Biomedical
Sciences, University of Bern, Bern, Switzerland
Background: Acute myeloid leukemia (AML) is characterized by poor prognosis for patients,
due to the very high rate of relapse, which occurs even after chemotherapy-induced
complete remission. Therapy-insensitive AML cells, so-called leukemia stem cells (LSCs),
are thought to be the main driver of relapse. In fact, high LSCs number and active
LSC self-renewal programs are related to poor therapy response and adverse outcome
in AML. Therefore, eliminating LSCs is an urgent, yet unmet, medical need. LSCs properties
are crucially regulated and maintained also through interactions with the bone marrow
microenvironment, which includes immune cells. Despite evidences of mutual interactions
between AML cells and immune cells, their interplay is still poorly understood. A
better understanding of these interactions might provide novel methods to effectively
eradicate LSCs and achieve durable AML cure.
Aims: The aim of this study was to investigate the role of IL21/IL21R signaling in
the so far unexplored context of AML. Our aims involved identification of IL21 source
in murine and human AML and elucidation of the mechanisms how IL21/IL21R signaling
regulates LSCs properties.
Methods: We used a combination of flow cytometry, ELISA and qRT-PCR to determine protein
and mRNA expression of both IL21 and IL21R in primary AML samples and analyzed the
correlation between IL21 serum levels with patients’ survival and complete remission
rate. Colony-forming assays and short-term cultures were used to determine the in
vitro effect of IL21 on primary normal and leukemic stem cells. In addition, patient-derived
xenografts were employed to study IL21 effect on stem cell frequency and AML engraftment
capacity.
We used various syngeneic murine AML models (e.g. MLL/AF9 and MLL/ENL) to study the
functional role of IL21/IL21R signaling in vivo. We performed RNA-Seq, together with
analysis of symmetric cell division, expression of differentiation markers, NF-kB
signaling, stem cell frequencies by limiting-dilution secondary transplantation experiments
to investigate the mechanisms how IL21 regulates LSCs.
Results: In human AML, serum IL21 was identified as an independent positive prognostic
biomarker for overall survival (OS). Furthermore, high dose chemotherapy was more
effective in patients with high levels of IL21 at diagnosis, as illustrated by a significantly
higher rate of complete remission (CR rate: 81 vs 59 %) and prolonged OS (median survival:
915 vs. 364 days, HR: 1.8). Functionally, IL21 inhibited cell growth and clonogenic
potential of primary AML stem and progenitor cells but not normal hematopietic stem
cells (HSCs). In murine AML models, IL21/IL21R signaling in LSCs reduced stem cell
frequency, prolonged survival of the mice and resulted in the downregulation of stemness-related
and the up-regulation of differentiation-promoting pathways. In both human and murine
AML, CD4+ T cells were identified as the main source of IL21.
Image:
Summary/Conclusion: In this work, we have identified IL21/IL21R signaling pathway
as an important regulator of cell fate in human and murine LSCs, but not HSCs. We
found that IL21 is a positive prognostic marker for OS and that higher serum IL21
levels correlate with better survival and higher rate of complete remission in patients
that undergo chemotherapy. Our findings therefore suggest that promoting IL21/IL21R
signaling on LSCs may be a novel approach to decrease stemness and increase differentiation
in AML.
S255: EFFICACY AND SAFETY OF TISAGENLECLEUCEL IN PEDIATRIC AND YOUNG ADULT PATIENTS
(PTS) WITH RELAPSED OR REFRACTORY (R/R) MATURE B-CELL NON-HODGKIN LYMPHOMA (NHL):
THE PHASE II BIANCA STUDY
V. Minard-Colin1,*, J. Buechner2, F. Locatelli3, B. Gonzalez Martinez4, B. J. Vormoor5,
S. Cooper6, J. Krueger7, S. Napolitano8, A. Attarbaschi9, A. Baruchel10, S. Ghorashian11,
M. L. Hermiston12, H. Hiramatsu13, M. Ifversen14, S. John15, S. L. Khaw16, T. A. O’Brien17,
C. L. Phillips18, C. Diaz de Heredia19, D. Tomizawa20, K. Vettenranta21, A. S. Wayne22,
S. Newsome23, R. Awasthi24, S. Redondo25, A. Masood24, S. L. Maude26, B. Burkhardt27
1Department of Pediatric and Adolescent Oncology, Gustave Roussy, Université Paris-Saclay,
Villejuif, France; 2Department of Pediatric Hematology and Oncology, Oslo University
Hospital, Oslo, Norway; 3Department of Pediatric Hematology and Oncology, IRCCS Ospedale
Bambino Gesù Children’s Hospital, Sapienza, University of Rome, Rome, Italy; 4Hospital
Universitario La Paz, Madrid, Spain; 5Prinses Maxima Centrum, Utrecht, Netherlands;
6Johns Hopkins Comprehensive Cancer Center, Baltimore, MD, United States of America;
7Division of Haematology/Oncology/Bone Marrow Transplantation, Department of Paediatrics,
The Hospital for Sick Children, Toronto, ON, Canada; 8Hematology-Oncology and Bone
Marrow Transplantation Unit, Pediatric Department, and Monza and Brianza Foundation
for Children and their Mums, San Gerardo Hospital, Lombardia, Monza, Italy; 9St. Anna
Kinderspital and Children’s Cancer Research Institute, Vienna, Austria; 10University
Hospital Robert Debré (APHP) and Université de Paris, Paris, France; 11Great Ormond
Street Hospital for Children NHS Trust Haematology Department, London, United Kingdom;
12University of California San Francisco Benioff Children’s Hospital, San Francisco,
CA, United States of America; 13Department of Pediatrics, Graduate School of Medicine,
Kyoto University, Kyoto, Japan; 14Department of Pediatrics and Adolescent Medicine,
Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; 15University of
Texas Southwestern Medical Center, Dallas, TX, United States of America; 16Children’s
Cancer Centre, Royal Children’s Hospital and Murdoch Children’s Research Institute,
Parkville, VIC; 17Kids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW, Australia;
18Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of
America; 19Hospital Universitari Vall d’Hebron, Vall d’Hebron Institut de Recerca,
Barcelona, Spain; 20Children’s Cancer Center, National Center for Child Health and
Development, Tokyo, Japan; 21University of Helsinki, Helsinki, Finland; 22Children’s
Hospital Los Angeles, USC Norris Comprehensive Cancer Center, Keck School of Medicine,
University of Southern California, Los Angeles, CA, United States of America; 23Novartis
Pharma AG, Basel, Switzerland; 24Novartis Pharmaceutical Corporation, East Hanover,
NJ, United States of America; 25Novartis Farmaceutica S.A., Madrid, Spain; 26Division
of Oncology, Center for Childhood Cancer Research and Cancer Immunotherapy Program,
Children’s Hospital of Philadelphia and Department of Pediatrics, Abramson Cancer
Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,
United States of America; 27Pediatric Haematology and Oncology, University Hospital
Muenster, Muenster, Germany
Background: Chimeric antigen receptor (CAR)-T cell therapy (tx) targeting CD19 is
approved for treatment of adult r/r large B-cell lymphoma (LBCL) and pediatric r/r
B-cell acute lymphoblastic leukemia (ALL). Pediatric and young adult pts with r/r
mature B-NHL have dismal prognoses, especially those with Burkitt lymphoma (BL), and
limited clinical benefit from available tx.
Aims: Herein we report primary efficacy and safety outcomes from the global, multicenter,
open-label, single-arm Phase II BIANCA trial (NCT03610724).
Methods: Pts ≤25 y and ≥6 kg at screening with histologically confirmed CD19+ r/r
mature B-NHL after ≥1 prior lines of tx were eligible following successful leukapheresis.
Pts received optional bridging chemotherapy and fludarabine/cyclophosphamide or cytarabine/etoposide
for lymphodepletion prior to a single intravenous injection of tisagenlecleucel (target
dose range, 0.2-5x106/kg bodyweight [pts ≤50 kg] or 0.1-2.5x108 [pts >50 kg] CAR+
viable T cells). The primary endpoint was overall response rate (ORR=complete response
[CR]+partial response [PR]) by local investigator assessment in the efficacy analysis
set (EAS), which excludes pts with pre-infusion CR. Secondary endpoints included progression-free
survival (PFS), overall survival (OS), safety, and CAR-T cell kinetics.
Results: As of October 10, 2021, 33/34 pts enrolled received tisagenlecleucel (EAS,
N=28). Median age was 13 y (range, 3-22 y), 70% were male, 55% had BL, and 45% had
LBCL. Pts received a median of 2 prior tx (18% received prior stem cell transplant
[SCT]), 85% had stage III/IV disease at initial diagnosis, 15% were primary refractory,
30% were refractory, and 55% had relapsed/progressed at study entry. Median time from
enrollment to infusion was 35 d (range, 7-62 d). Median time from tisagenlecleucel
infusion to data cutoff was 16 mo (range, 6-30 mo). Bridging chemotherapy was given
to 94%. ORR was 32% (95% CI: 15.9-52.4); 7% had a CR. Subgroup analysis suggested
that pts with BL had a lower ORR than pts with LBCL (20% vs 46%). Median PFS was 2.5
mo (95% CI: 1.1-2.9); median OS was 11.4 mo (95% CI: 3.4-not estimable); 12-mo estimated
PFS and OS were 23% (95% CI: 8.9-40.3) and 47% (95% CI: 26.6-65.0), respectively.
Of the 18 BL pts, 7 (39%) are alive post infusion (1 received allogeneic SCT, 1 surgery,
3 other tx, and 2 no other tx). There were no tx-related deaths; most tx-related grade
(gr) ≥3 (94%) and serious (52%) adverse events occurred ≤8 w post infusion. A neurologic
event (NE) occurred in 27%; 15% had a gr ≥3 NE, and none had gr 5 NE. Cytokine release
syndrome (CRS; per Lee 2014 criteria) occurred in 70%, 9% had gr 3 CRS, and there
was no gr ≥4 CRS. Median time to CRS onset and duration were 6 d (range, 1-27 d) and
5 d (range, 1-13 d), respectively. Only 1 pt died ≤30 d post infusion, due to disease
progression. Geometric mean maximal expansion (Cmax) across all pts was 5730 copies/μg,
and median time to Cmax was 12.8 d (range, 2.5-21.9 d) by qPCR. Median CAR-T cell
persistence in pts with CR or PR was 182 d (range, 20.9-562 d).
Image:
Summary/Conclusion: Tisagenlecleucel demonstrated efficacy in pediatric and young
adult pts with r/r mature B-NHL and comparable safety to that recorded in adults with
DLBCL. The OS is encouraging; however, optimal positioning of tisagenlecleucel in
the treatment of pediatric r/r B-NHL requires further exploration, especially for
BL, which is highly aggressive and challenging to treat in the relapse setting.
S256: TABELECLEUCEL FOR EBV-DRIVEN POST-TRANSPLANT LYMPHOPROLIFERATIVE DISEASE FOLLOWING
ALLOGENEIC HEMATOPOIETIC CELL OR SOLID ORGAN TRANSPLANT AFTER FAILURE OF RITUXIMAB
± CHEMOTHERAPY (ALLELE)
S. Prockop, MD1,*, A. Beitinjaneh, MD2, S. Choquet, MD3, S. Dahiya, MD4, R. Dinavahi,
MD5, R. Farah, MD6, L. Gamelin, MD, PhD5, A. Ghobadi, MD7, A. Mehta5, P. Nayak, MD8,
R. Reshef, MD9, G. Satyanarayana, MD10, P. Stiff, MD11, W. Ye, PhD8, K. M. Mahadeo,
MD, MPH12
1Boston Children’s Hospital/Dana Farber Cancer Institute, Boston; 2University of Miami/Jackson
Memorial Hospital, Miami, United States of America; 3Sorbonne Université, Hôpital
de la Pitié-Salpêtrière, Paris, France; 4University of Maryland School of Medicine,
Baltimore; 5Atara Biotherapeutics, Thousand Oaks; 6UPMC Hillman Cancer Center, Pittsburgh;
7Washington University, Division of Oncology, St. Louis; 8Atara Biotherapeutics, South
San Francisco; 9Columbia University Medical Center, New York; 10Ingram Cancer Center,
Vanderbilt Health, Nashville; 11Loyola University Medical Center, Chicago; 12MD Anderson
Cancer Center, Houston, United States of America
Background: Tabelecleucel is an investigational, off-the-shelf, allogeneic Epstein–Barr
Virus (EBV)-specific T-cell immunotherapy being explored in patients (pts) with EBV-driven
post-transplant lymphoproliferative disease (EBV+ PTLD). Median overall survival (OS)
after rituximab (R) ± chemotherapy (C) failure is 0.7 months in EBV+ PTLD post allogeneic
hematopoietic cell transplant (HCT) (Sanz ASH 2021) and 4.1 months post solid organ
transplant (SOT) after R+C failure (Dharnidharka ASH 2021), demonstrating an urgent
need for therapies in this ultra-rare disease. Tabelecleucel has shown promising outcomes
(Prockop JCI 2020). Additionally, an investigator-assessed objective response rate
(ORR) of >60% with >80% 2-year OS (Prockop EBMT 2021, Prockop ATC 2021, Prockop ASH
2021) was seen in pts with relapsed/refractory (R/R) EBV+ PTLD, with either complete
or partial response to tabelecleucel. Safety data in >180 pts with tabelecleucel treated
EBV+ PTLD demonstrate tumor flare reaction (TFR) as the only identified risk (Atara
Biotherapeutics, Data on file).
Aims: Report data from the ongoing Phase 3 ALLELE study (NCT03394365).
Methods: ALLELE is a multicenter, open-label study investigating tabelecleucel in
pts with EBV+ PTLD post HCT after R failure (n=33) and post SOT after R±C failure
(n=33). Pts receive tabelecleucel at 2 x 106 cells/kg on Days 1, 8 and 15 in 35-day
cycles. Response is evaluated by investigator and by independent oncologic response
adjudication (IORA; primary assessment) using Lugano Classification with LYRIC modification.
Efficacy endpoints include ORR, duration of response (DOR), time to response (TTR),
and OS. Pts are assessed post treatment for up to 5 years for survival.
Results: As of May 2021, 38 pts (14 HCT, 24 SOT) were evaluable by IORA and had the
opportunity for 6 months follow-up. Median age was 52.9 (range, 3.2–81.5) years, 44.4%/47.2%
of pts were high/intermediate risk per PTLD-adapted prognostic index, and median number
of lines of prior systemic treatment was 1 (range, 1–5). R/R HCT and SOT pts received
a median (range) of 3 (1–5) and 2 (1–6) cycles of tabelecleucel, respectively. ORR
was 50.0% (19/38, 95% CI: 33.4, 66.6) overall, 50.0% (7/14, 95% CI: 23.0, 77.0) in
HCT, and 50.0% (12/24, 95% CI: 29.1, 70.9) in SOT (Table). Overall, median TTR was
1.1 (0.7–4.7) months, 11 of 19 responders had DOR > 6 months, and median DOR was not
reached (Table).
Median OS was 18.4 (95% CI: 6.9, NR) months overall, not yet reached for HCT, and
16.4 (95% CI: 3.5, NR) months for SOT. 1-year survival rates were 61.1% (95% CI: 42.9,
75.0) overall, 66.8% (95% CI: 32.4, 86.6) for HCT, and 57.4% (95% CI: 35.2, 74.5)
for SOT. Median OS was NR (95% CI: 16.4, NR) for responders and 5.7 (95% CI: 1.8,
12.1) months for non-responders. Responders had a higher 1-year survival rate vs non-responders:
89.2% (95% CI: 63.1, 97.2) vs 32.4% (95% CI: 12.1, 54.9) (Table).
Serious treatment emergent AEs (TEAEs) and fatal TEAEs were reported in 57.1% and
7.1% of HCT and 62.5% and 16.7% of SOT pts respectively. No fatal TEAE was treatment
related. There were no reports of TFR, infusion reactions, cytokine release syndrome,
marrow rejection, or transmission of infectious diseases, and no events of graft vs
host disease or organ rejection reported as related to tabelecleucel.
Image:
Summary/Conclusion: Tabelecleucel Phase 3 data show clinically meaningful outcomes
and promising ORR and OS in a pt population with poor survival and no approved therapies.
Tabelecleucel was well tolerated without evidence of safety concerns typically observed
with other adoptive T-cell therapies.
S257: A PHASE 1, FIRST-IN-HUMAN, DOSE-ESCALATION CLINICAL TRIAL OF MEMORY-ENRICHED
CD30-CAR T-CELL THERAPY FOR THE TREATMENT OF RELAPSED OR REFRACTORY HODGKIN LYMPHOMA
AND CD30+ T-CELL LYMPHOMA
A. C. Caballero1,*, L. Escribà-Garcia1, R. Montserrat-Torres1, E. Escudero-López1,
P. Pujol-Fernández1, C. Ujaldón-Miró1, I. Garcia-Cadenas1, A. Esquirol1, R. Martino1,
J. Sierra1, C. Alvarez-Fernández1, J. Briones1
1Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
Background: Up to 30% of Hodgkin lymphoma (HL) patients are refractory or relapse
(R/R) after first treatment and their prognosis is poor. We have developed a refined
CD30-CAR-T (HSP-CAR30) targeting a proximal epitope within the CD30 molecule to overcome
soluble CD30 and generated products enriched in memory T-cells to ensure efficient
engraftment, persistence and enhancement of antitumor efficacy (Alvarez-Fernandez
et al, 2021). Here, we report the results of our Phase 1 study evaluating HSP-CAR30
for the treatment of R/R HL and CD30+ T-cell non-Hodgkin lymphoma (T-NHL) (NCT04653649).
Aims: Primary endpoints were to assess safety of HSP-CAR30 and to establish maximum
tolerated dose (MTD) recommended for the following Phase 2. Secondary objectives include
best response rates after infusion.
Methods: We conducted a phase 1 dose-escalation study in 11 patients with R/R HL or
CD30+ T-NHL. HL patients were R/R to treatments including chemotherapy, brentuximab
and anti-PD-1 antibodies, while T-NHL patients were R/R to at least 2 chemotherapy
treatments. T-cells were transduced with a lentivirus encoding a second-generation
4-1BB costimulated CAR, containing a scFv directed against an epitope from the proximal
non-cleavable part of CD30 protein. Three cell-dose levels were evaluated: DL1 (3x106/kg),
DL2 (5x106/kg) and DL3 (10x106/kg) CAR30+ T-cells.
Results: From February 2021 to December 2021, 11 patients (9 HL and 2 CD30+ T-NHL)
were enrolled and underwent leukapheresis. Of these, 10 patients received HSP-CAR30:
3 patients at DL1, 3 at DL2 and 4 at DL3. Demographic characteristics and baseline
disease features are summarized in the Table. Median age was 49.9 years (range 21–65).
Median number of prior lines of treatment was 4.6 (range 3–7). One patient did not
received treatment due to lack of T-cell expansion. All patients received LD before
infusion, fludarabine/bendamustine in HL patients (n=8) and fludarabine/cyclophosphamide
in T-NHL (n=2). Mean HSP-CAR30 expression was 94.79±3,38% (±SD). Memory T-cell subset
comprised 93.07±4,8% (±SD) in CD4+ and 91.64±4,9% (±SD) in CD8+. Mean time to HSP-CAR30
cell peak level across all doses was 29 days (range 6–98). CAR+ T-cells were detectable
by flow cytometry up to 11 months after infusion. HSP-CAR30 infusion was well tolerated;
there were no dose limiting toxicities (DLTs). Relevant adverse events are shown in
the Table. Grade 1 cytokine release syndrome (CRS) was observed in 6 (60%) patients.
No patient developed neurotoxicity. Self-limited skin rash was seen in 4 (40%) patients.
One patient with history of cytomegalovirus (CMV) infections had CMV pneumonia. Another
patient developed pulmonary tuberculosis. At data cutoff (February 21st, 2021), the
median follow-up was 204 days (60–351). Best objective response was 100%, including
5 (50%) patients with complete response (CR), all with HL (DL1=1; DL2=3; DL3= 1).
Three patients have died of progressive disease (2 T-NHL and 1 HL). There were no
non-relapse mortality events. Median PFS and median overall survival (OS) was not
reached. Six-month PFS for HL patients was 75%.
Image:
Summary/Conclusion: This is the first European academic CART clinical trial evaluating
a T-cell memory-enriched CART 30. Our Phase 1 study provides evidence for feasibility
and safety of HSP-CAR30. Additionally, HSP-CAR30 has shown promising efficacy in heavily
treated HL patients that is being explored in a phase 2 trial already started.
S258: LISOCABTAGENE MARALEUCEL (LISO-CEL) AS SECOND-LINE THERAPY FOR R/R LARGE B-CELL
LYMPHOMA (LBCL) IN PATIENTS NOT INTENDED FOR HSCT: PRIMARY ANALYSIS FROM THE PHASE
2 PILOT STUDY
A. Sehgal1,*, D. Hoda2, P. A. Riedell3, N. Ghosh4, M. Hamadani5, G. C. Hildebrandt6,
J. E. Godwin7, P. Reagan8, N. Wagner-Johnston9, J. Essell10, R. Nath11, S. R. Solomon12,
R. Champion13, E. Licitra14, S. Fanning15, N. Gupta16, R. Dubowy17, A. D’Andrea18,
L. Wang17, L. I. Gordon19
1University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh; 2Intermountain
Healthcare, Loveland Clinic for Blood Cancer Therapy, Salt Lake City; 3University
of Chicago Comprehensive Cancer Center, Chicago; 4Levine Cancer Institute, Atrium
Health, Charlotte; 5BMT & Cellular Therapy Program, Medical College of Wisconsin,
Milwaukee; 6Markey Cancer Center, University of Kentucky, Lexington; 7Providence Cancer
Center, Earle A. Chiles Research Institute, Portland; 8University of Rochester Medical
Center, Rochester; 9Johns Hopkins Hospital, Baltimore; 10Oncology Hematology Care,
Cincinnati; 11Banner MD Anderson Cancer Center, Gilbert; 12Northside Hospital Cancer
Institute, Atlanta; 13Norton Cancer Institute, Louisville; 14Astera Cancer Care, East
Brunswick; 15Prisma Health, Greenville; 16Stanford Cancer Genetics Clinic, Palo Alto;
17Bristol Myers Squibb, Seattle, United States of America; 18Celgene, a Bristol-Myers
Squibb Company, Boudry, Switzerland; 19Northwestern University, Feinberg School of
Medicine, Robert H. Lurie Comprehensive Cancer Center, Chicago, United States of America
Background: Patients with relapsed or refractory (R/R) LBCL after first-line treatment
who are unable to undergo high-dose chemotherapy (HDCT) and hematopoietic stem cell
transplantation (HSCT) have poor outcomes and limited treatment options.
Aims: PILOT (NCT03483103) evaluated liso-cel, an autologous, CD19-directed chimeric
antigen receptor (CAR) T cell product, as second-line treatment in patients with R/R
LBCL not intended for HSCT.
Methods: Eligible patients were adults with R/R LBCL after first-line treatment who
were not deemed candidates for HDCT and HSCT by their physician and met ≥ 1 frailty
criteria as follows: age ≥ 70 years, Eastern Cooperative Oncology Group performance
status (ECOG PS) of 2, diffusing capacity for carbon monoxide ≤ 60%, left ventricular
ejection fraction < 50%, creatinine clearance < 60 mL/min, or alanine aminotransferase/aspartate
aminotransferase > 2 × the upper limit of normal. Bridging therapy was allowed. Patients
received lymphodepletion with cyclophosphamide and fludarabine, followed 2–7 days
later by liso-cel infusion at a target dose of 100 × 106 CAR+ T cells. Cytokine release
syndrome (CRS) was graded per Lee 2014 criteria. Neurological events (NE) were defined
as investigator-identified neurological adverse events related to liso-cel and graded
using the National Cancer Institute Common Terminology Criteria for Adverse Events,
version 4.03. The primary endpoint was objective response rate (ORR) per independent
review committee; all patients had ≥ 6 months of follow-up from first response.
Results: Of 74 patients who underwent leukapheresis, 61 received liso-cel and 1 received
nonconforming product (ie, product wherein one of the CD8 or CD4 cell components did
not meet one of the requirements to be considered liso-cel). Common reasons for preinfusion
dropout included death and loss of eligibility (5 each). For liso-cel–treated patients,
median age was 74 years (range, 53–84; 79% ≥ 70 years) and 69%, 26%, and 5% met 1,
2, and 3 frailty criteria, respectively; 26% had ECOG PS of 2 and 44% had Hematopoietic
Cell Transplantation-specific Comorbidity Index score ≥ 3. After first-line treatment,
54% were chemotherapy refractory, 21% relapsed within 12 months, and 25% relapsed
after 12 months; 51% of patients received bridging chemotherapy. Median (range) on-study
follow-up was 12.3 months (1.2–26.5). ORR and complete response rate were 80% and
54%, respectively (Table). Median duration of response and progression-free survival
were 12.1 months and 9.0 months, respectively. Median overall survival has not been
reached. The most frequent treatment-emergent adverse events (TEAE) were neutropenia
(51%), fatigue (39%), and CRS (38%), with grade 3 CRS in 1 patient (2%) and no grade
4/5 CRS events. Any-grade NEs were seen in 31% (n = 19) of patients; grade 3 NEs occurred
in 5% (n = 3) of patients and no grade 4/5 NEs were reported. Seven percent (n = 4)
received tocilizumab only, 3% (n = 2) received corticosteroids only, and 20% (n =
12) received both tocilizumab and corticosteroids for treatment of CRS and/or NEs.
Overall, grade ≥ 3 TEAEs occurred in 79% (n = 48) of patients, with grade 5 TEAEs
in 2 patients (both due to COVID-19). Two patients (3%) had grade 3/4 infections and
15 (25%) had grade ≥3 neutropenia at Day 29.
Image:
Summary/Conclusion: In the PILOT study, liso-cel as second-line treatment in patients
with LBCL who met ≥ 1 frailty criteria and for whom HSCT was not intended demonstrated
substantial and durable overall and complete responses, with no new safety concerns.
S259: DUAL ANTIGEN TARGETING WITH CO-TRANSDUCED CD19/22 CAR T CELLS FOR RELAPSED/REFRACTORY
ALL
S. Ghorashian1,*, G. Lucchini2, R. Richardson3, K. Nguyen3, C. Terris3, J. Yeung3,
J. Chu2, L. Williams2, K. Ko2, C. Walding4, K. Watts5, S Inglott1, S. Adams1, E. Gravett1,
K. Gilmour6, A. Lal7, S. Kunaseelan7, B. Popova7, A. Lopes7, Y. Ngai7, E. Kokalaki8,
K. Rao2, R. Chiesa2, J. Silva2, K Mullanfiroze2, A. Lazareva2, D. Bonney5, R Wynn5,
M. Pule8, R. Hough4, P. Amrolia2
1Haematology; 2Bone Marrow Transplant, Great Ormond St Children’s Hospital; 3Molecular
and Cellular Immunology, UCL Great Ormond St Institute of Child Health; 4Haematology,
University College London Hospital NHS Trust, London; 5Blood and Marrow Transplant,
Royal Manchester Children’s Hospital, Manchester; 6Cell Therapy and Immunology, Great
Ormond St Children’s Hospital; 7Cancer Research UK & UCL Cancer Trials Centre; 8Autolus
Ltd, London, United Kingdom
Background: CD19 negative escape is a major cause of relapse after CD19 CAR T cell
therapy for relapsed/refractory (r/r) paediatric ALL and dual targeting of CD19/CD22
may overcome this. We have previously shown that AUTO1, a fast off rate autologous
CD19 CAR T cell therapy was highly active in ALL with a favorable safety profile and
excellent persistence (Ghorashian et al Nat.Med. 2019). Building on these properties,
we developed AUTO1/22 in which autologous T cells are co-transduced with 2 different
lentiviral vectors encoding our existing CD19 CAR and a novel CD22CAR designed to
recognise targets with low antigen density. AUTO1/22 was evaluated in a Phase I study
in children/young adults with r/rALL (NCT02443831).
Aims: To determine the safety/biological efficacy of AUTO1/22
Methods: Patients with r/r B-ALL age < 25 ywho were ineligible for/relapsed after
Tisagenlecleucel were recruited. Following fludarabine/cyclophosphamide lymphodepletion,
patients received 1x106 /kg CAR+ T cells. The presence of CAR T cells in the blood/bone
marrow (BM) was assessed by flow cytometry + qPCR and BM MRD was assessed by IgH qPCR
+ flow cytometry. Primary end-points were incidence of grade 3-5 toxicity and the
proportion of patients achieving MRD negative remission.
Results: Ten patients have been treated and 8 are evaluable with >1 month follow-up.
The median age was 12 years and patients had a median of 3.5 prior lines of therapy
(range 2-6). Five of 8 patients had relapsed post allogeneic SCT, 4 had received prior
Blinatumomab/Inotuzumab and 3 had relapsed after prior Tisagenlecleucel. Prior to
lymphodepletion, 2 patients had >5% BM disease, 5 had MRD between 10-2 and 10-5 and
1 was BM MRD negative. CAR T cell products had a central memory phenotype with predominance
of CD19/22 double positive cells (median 59.3%) and balanced populations of CD19 and
CD22 single positive cells (16% and 10.9% respectively).Cytokine release syndrome
(CRS) occurred in 7/8 patients (grade 1 n=2, grade 2 n=5) requiring Tocilizumab in
3 cases, but severe (≥ grade 3) CRS was not seen and no patients required ICU admission
for CRS. Grade 1-2 ICANS was observed in 3 patients. One patient had delayed grade
4 leucoencephalopathy (MRI/brain biopsy were more indicative of fludarabine toxicity
than CAR T related) and has ongoing neurological recovery. 7 patients had grade 3-4
cytopenia persisting beyond/recurring after day 28, requiring a CD34+ stem cell top
up in 1 case. 5/8 patients had CD19CAR T cells and 3/8 patients had CD22CAR T cells
detectable at last follow-up. 7 of 8 evaluable patients (88%) achieved MRD negative
CR/CRi at 1 month post-infusion. One patient did not respond with CD19+ CNS relapse
+ MRD level BM disease at day 28. Of the 7 responding patients, 1 had frank CD19+CD22+
BM and extramedullary relapse at 3 months and 1 had emergence of MRD level disease
at 10.5 months post infusion, in both cases associated with loss of CAR T cells. One
other patient had early loss of CAR T cells with B cell recovery but ongoing MRD negative
CR at 3 months post-infusion and remains in MRD negative CR on maintenance chemotherapy.
Overall, at a median follow-up of 4.8 months, 5/8 patients remain in MRD negative
CR at last follow-up.
Summary/Conclusion: We demonstrate that dual CD19/22 targeting CAR T cells generated
by co-transduction show an acceptable safety profile, with robust expansion/persistence
and early efficacy in a heavily pre-treated cohort. To date with limited follow-up
we have not observed antigen negative relapse but longer follow up is needed.
S260: A MATCHED COMPARISON OF TISAGENLECLEUCEL AND AXICABTAGENE CILOLEUCEL CAR T CELLS
IN RELAPSED OR REFRACTORY DIFFUSE LARGE B-CELL LYMPHOMA: A REAL-LIFE LYSA STUDY FROM
THE FRENCH DESCAR-T REGISTRY
E. Bachy1,*, S. Le Gouill2, P. Sesques3, R. Di Blasi4, M. Guillaume5, G. Cartron6,
D. Beauvais7, L. Roulin8, F. X. Gros9, M. T. Rubio10, P. Bories11, J. O. Bay12, C.
Castilla Llorente13, S. Choquet14, R.-O. Casasnovas15, M. Mothy16, S. Guidez17, M.
Joris18, M. Loschi19, S. Carras20, J. Abraham21, A. Chauchet22, L. Drieu La Rochelle23,
J. Zerbit24, O. Hermine25, T. Gastinne26, J. J. Tudesq6, E. Gat27, F. Broussais27,
C. Thieblemont28, R. Houot5, F. Morschhauser7
1Hematology, Hospices Civils de Lyon, Pierre Bénite; 2Institut Curie, Paris; 3Hospices
Civils de Lyon, Pierre Bénite; 4AP-HP, Paris; 5CHU Rennes, Rennes; 6CHU Montpellier,
Montpellier; 7CHU Lille, Lille; 8CHU Mondor, Paris; 9CHU Bordeaux, Bordeaux; 10CHU
Nancy, Nancy; 11CHU Toulouse, Toulouse; 12CHU Clermont Ferrand, Clermont Ferrand;
13GUSTAVE ROUSSY CANCER CAMPUS GRAND PARIS, Villejuif; 14CHU Pitié Salprétrière, Paris;
15CHU Dijon, Dijon; 16CHU Saint Antoine, Paris; 17CHU Poitiers, Poitiers; 18CHU Amiens,
Amiens; 19CHU Nice, Nice; 20CHU Grenoble, Grenoble; 21CHU Limoges, Limoges; 22CHU
Besancon, Besancon; 23CHU Tours, Tours; 24APHP Cochin; 25CHU Necker, Paris; 26CHU
Nantes, Nantes; 27LYSARC, Lyon; 28CHU Saint Louis, Paris, France
Background: Axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tisa-cel) have
both demonstrated impressive clinical activity in relapsed/refractory (R/R) diffuse
large B-cell lymphoma (DLBCL). In a previous propensity score matching (PSM) analysis
(Bachy et al., ASH 2021) allowing for balanced comparison between axi-cel and tisa-cel
outcomes, we reported on a prolonged progression-free survival (PFS) but a higher
toxicity associated with axi-cel compared with tisa-cel. No OS difference was observed
but the follow-up was short (6 months).
Aims: We aim at reporting on PSM analysis with longer follow-up and with additionnal
patients treated with axi-cel or tisa-cel.
Methods: All patients treated in France with axi-cel or tisa-cel from the 1st July
2018 to the 1st October 2021 and included in the DESCAR-T registry were considered.
Propensity score matching (PSM) was used to create a balanced covariate distribution
between a cohort of patients treated with tisa-cel and a cohort of patients treated
with axi-cel. An exhaustive list of covariates was used for PSM: age, sex, LDH level,
C reactive protein (CRP), time between last treatment and infusion, Eastern Cooperative
Oncology Group (ECOG) performance status (PS), Ann Arbor stage, number of prior lines
of treatment before CAR-T, bridging and response to bridging, prior stem cell transplant
(SCT) either autologous or allogeneic, bulk assessed at lymphodepletion, centre, histological
diagnosis. PSM was performed considering a 1:1 matching without replacement and with
optimal matching applying a calliper width of the propensity score set at 0.1. Inverse
probability of treatment weighting (IPTW) was used as another approach to further
validate PSM analysis.
Results: 809 patients from 25 French centres with R/R DLBCL after at least 2 lines
of previous therapy had a commercial CAR-T order with axi-cel or tisa-cel and were
registered in DESCAR-T. Out of 809 patients with a CAR-T order, 60 were not infused
due to progression or death between leukapheresis and lymphodepletion and 20 did not
proceed to lymphodepletion for other reasons. Finally, 729 proceeded to lymphodepletion
and CAR-T infusion. In the 1:1 matched population (N=418, 209 patients treated with
tisa-cel and 209 patients treated with axi-cel), the best ORR/CRR was 66/42% versus
80/60% for patients treated with tisa-cel compared with axi-cel, respectively (P<0.001
for both ORR and CRR comparisons). After a median FU of 11.7 months (95% CI, 10.5-12.0
months) the 1-yr PFS was 33% for tisa-cel and 47% for axi-cel (HR=1.65, 95% CI 1.26-2.18,
P=0.0003). OS was also significantly longer following axi-cel infusion than following
tisa-cel infusion (1-yr OS 63% versus 49%; HR=1.58, 95% CI, 1.13-2.21; P=0.0072).
Similar findings were found using IPTW statistical approach. Grade 1-2 CRS were significantly
more frequent with axi-cel than tisa-cel (P=0.004) but no significant difference was
observed for grade 3 or more CRS (9% versus 5% for tisa-cel and axi-cel respectively,
P=0.130). Regarding ICANS, both all grades and severe (i.e. grade ≥3) ICANS were significantly
more frequent with axi-cel than tisa-cel. 48% of patients experienced ICANS after
axi-cel infusion compared to 22% after tisa-cel infusion. 29 patients (14%) presented
a grade ≥3 ICANS with axi-cel compared with 6 (3%) only with tisa-cel.
Image:
Summary/Conclusion: In conclusion, our matched-comparison study supports a higher
efficacy but also a higher toxicity of axi-cel compared with tisa-cel in third or
more treatment line for R/R DLBCL.
S261: SAFETY AND PRELIMINARY EFFICACY FINDINGS OF AUTO4, A TRBC1-TARGETTING CAR, IN
RELAPSED/REFRACTORY TRBC1 POSITIVE SELECTED T CELL NON-HODGKIN LYMPHOMA
K. Cwynarski1,*, E. Tholouli2, G. Iacoboni3, T. Menne4, D. Irvine5, L. Wood6, N. Balasubramaniam7,
J. Shang8, M Zhang8, K. Duffy9, B. Huber10, M. Vinson11, W. Brugger9, M. Pule12 13
1Haematology, University College London, London; 2Manchester Royal Infirmary, Manchester,
United Kingdom; 3VHIO Vall d’Hebron Hospital, Barcelona, Spain; 4Freeman Hospital
Newcastle, Newcastle; 5University of Glasgow, Glasgow; 6Cancer Clinical Trials Unit,
University College London Hospitals; 7Cancer Clinial Trials Unit, University College
London, London, United Kingdom; 8Autolus Therapeutics, Rockville, United States of
America; 9Clinical Development, Autolus Therapeutics, London, United Kingdom; 10Autolus
Therapeutics, Munich, Germany; 11Autolus Therapeutics, London, United Kingdom; 12Research
& Development, Autolus Therapeutics; 13University College London, London, United Kingdom
Background: Peripheral T cell lymphomas (PTCL) are typically aggressive, treatment
resistant and associated with poor prognosis. Clinical application of immunotherapy
is limited by a lack of target antigens that discriminate malignant from normal T
cells. Unlike B cell depletion, pan–T cell aplasia is prohibitively toxic. We recently
described a targeting strategy based on the mutually exclusive expression of T cell
receptor beta-chain constant domains 1 and 2 (TRBC1 and TRBC2) (Maciocia, PM. et al,
Nat Med 2017) which can spare a proportion of the normal T cell compartment.
Aims: Here we describe early clinical findings of AUTO4, a TRBC1 directed autologous
CAR T cell therapy, tested against relapsed/refractory (r/r) TRBC1+ PTCL.
Methods: NCT03590574 is multi-centre, single-arm study of AUTO4 with a phase I dose
escalation component and a phase II expansion cohort. Here we report the initial findings
of the phase I component. Biopsies from patients >18 years of age were screened for
TRBC1-positive PTCL using next-generation sequencing. Four flat dose levels were explored:
25 x 106, 75 x 106, 225 x 106, and 450 x 106 CAR T cells administered as a single
dose. CAR T-cell products are generated using a semi-automated closed process. Patients
received lymphodepletion with fludarabine (30mg/m2 x4, day-6 to day-3) and cyclophosphamide
(500mg/m2 x2 on day-6 and day-5) (Flu/Cy) prior to AUTO4 infusion on Day 0. Primary
endpoints were incidence of Grade 3 to 5 toxicity occurring within 60 days of AUTO4
infusion and the frequency of dose limiting toxicities within 28 days of AUTO4 infusion
(DLT period). Overall response (CR+PR) rate post AUTO4 infusion by PET-CT (Lugano
2014 criteria) was a secondary endpoint.
Results: As of 09-FEB-2022, n=64 patients consented for screening of TRBC1-positive
PTCL. N=24 samples were TRBC1-positive; 7 patients were screen failures including
1 patient who died during screening. 11 products were manufactured; one patient relapsed
prior to AUTO4 infusion, and one patient screen failed after product manufacture.
To date 9 patients have been treated with AUTO4. The median age in these 9 patients
was 57 years (range 34 to 63 years). The T-cell lymphoma subtypes treated were PCTL-NOS
(n=4), ALCL (n=1), and AITL (n=4). Two patients had prior stem cell transplantation.
The median number of prior treatment lines was 3 (range 1-5). After lymphodepletion
with Flu/Cy, 3 patients received 25 × 106 CAR T cells, 2 patients received 75 × 106
CAR T cells, 1 patient received 225 × 106 CAR T cells and 3 patients received 450
× 106 CAR T cells. No patient experienced any dose limiting toxicities. The median
(range) number of CD3+ T-cells/µl in blood prior to lymphodepletion and at the end
of the DLT period (Day 28) was 204 (94-698) and 123 (19-458), respectively. 3 patients
(33%) experienced CRS (1 patient with Grade 1, 1 patient with Grade 2, and 1 patient
with Grade 3). None of the patients experienced neurotoxicity/ICANS. The most common
treatment-emergent adverse events were cytopenias (anemia and neutropenia). Of the
9 patients treated, 5 patients had achieved complete metabolic responses (CMR) by
PET-CT at Month 1, one patient remains with a PR 6 months post AUTO4 infusion, and
3 patients did not respond. All 3 patients at the 450x106 cell dose achieved a CMR
at Month 1.
Summary/Conclusion: AUTO4 has a tolerable safety profile in patients with r/r TRBC1+
peripheral T-cell lymphoma. Early data shows encouraging response rates. Updated data
and longer follow up will be presented.
S262: THE COBALT-LYM STUDY OF CTX130: A PHASE 1 DOSE ESCALATION STUDY OF CD70-TARGETED
ALLOGENEIC CRISPR-CAS9–ENGINEERED CAR T CELLS IN PATIENTS WITH RELAPSED/REFRACTORY
(R/R) T-CELL MALIGNANCIES
S. P. Iyer1,*, R. A. Sica2, P. J. Ho3, B. Hu4, J. Zain5, A. Prica6, W.-K. Weng7, Y.
H. Kim8, M. S. Khodadoust9, M. L Palomba10, F. M. Foss11, K. Tipton12, E. L. Cullingford12,
Q. He12, A. Sharma12, S. M. Horwitz10
1Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center,
Houston; 2Department of Oncology, Montefiore Medical Center, Albert Einstein Cancer
Center, Bronx, United States of America; 3Institute of Haematology, Royal Prince Alfred
Hospital, Camperdown, Australia; 4Division of Hematology and Hematologic Malignancies,
Huntsman Cancer Institute, Salt Lake City; 5Department of Hematology and Hematopoietic
Cell Transplantation, City of Hope, Duarte, United States of America; 6Princess Margaret
Cancer Centre, Toronto, Canada; 7Division of Blood and Marrow Transplantation and
Cellular Therapy; 8Department of Dermatology; 9Division of Oncology, Department of
Medicine, Stanford University School of Medicine, Stanford; 10Memorial Sloan Kettering
Cancer Center, New York; 11Department of Dermatology, Yale School of Medicine, New
Haven; 12CRISPR Therapeutics, Cambridge, United States of America
Background: Overall survival (OS) in a subset of patients (pts) with T-cell lymphoma
(TCL) has improved with front-line combination chemotherapy; however, R/R TCL pts
continue to have very limited treatment options. For pts with R/R peripheral (PTCL)
and transformed cutaneous TCL (CTCL), median OS is 1-2.5 and <5 yrs, respectively.
Adapting autologous chimeric antigen receptor (CAR) T cell therapy for TCL continues
to be challenging due to poor function of donor T cells, fratricide effect, and risk
of infusing transduced malignant CAR T cells into pts. CTX130TM is a first-in-class,
CD70-targeting allogeneic (allo) CAR T therapy that may allow for CAR T therapy in
pts whose own T cells are not ideal to manufacture auto CAR T cells. CD70 is a co-stimulatory
protein with temporally limited expression on activated lymphocytes and is highly
expressed in many TCLs. CTX130 is modified with CRISPR/Cas9-editing to eliminate expression
of: 1) T-cell receptor (TCR) by TCR alpha constant disruption, 2) major histocompatibility
complex class I expression by β2-microglobulin disruption, and 3) CD70 to mitigate
fratricide and enhance performance.
Aims: Investigate safety and efficacy of CTX130 in pts with R/R TCL.
Methods: COBALTTM-LYM (NCT04502446) is an open-label, multicenter, global study evaluating
the safety and efficacy of CTX130 in pts ≥18 y with CD70+ (≥10% by immunohistochemistry)
R/R TCL (PTCL or CTCL). Pts with PTCL and CTCL must have received ≥1 or ≥2 prior lines
of systemic therapy, respectively. Pts received lymphodepleting chemotherapy (LDC)
with fludarabine 30mg/m2 and cyclophosphamide 500mg/m2 for 3 days, followed by CTX130.
Pts were treated with CTX130 at doses from 3x107 (dose level [DL]1) to 9x108 (DL4)
CAR+ T cells. Pts could receive a second course of CTX130 if response was not achieved
but had experienced clinical benefit or disease progression. The primary endpoint
is safety (incidence of dose limiting toxicities [DLTs]). Key secondary endpoints
include overall response rate (ORR, by Lugano and ISCL criteria for PTCL and CTCL,
respectively), disease control rate (DCR; ≥stable disease [SD]), duration of response
and OS.
Results: As of 6 Dec, 2021, 17 pts with TCL were enrolled; 15 received CTX130, were
evaluable for a Day 28 assessment and are included in the analysis. Among the pts
who received CTX130, median age was 67 y, 7 pts had PTCL, and 8 pts had CTCL. Median
CD70 expression was 90% (range, 20-100%). Median follow up was 3.1 months. At DL ≥3,
ORR was 71%, CR rate was 29% and DCR was 100% (Table). Responses were observed in
PTCL (75% ORR at DL≥3) and CTCL (67% ORR at DL≥3) and across disease compartments.
CTX130 had an acceptable safety profile across all DLs. There were no instances of
graft versus host disease, tumor lysis syndrome, hemophagocytic lymphohistiocytosis,
or infusion reactions with LDC or CTX130. There were no DLTs, no Grade (Gr) ≥3 cytokine
release syndrome (CRS) or immune effector cell-associated neurotoxicity syndrome (ICANS);
Gr 1-2 CRS and ICANS were 47% and 20%. There was no increase in frequency or severity
of CRS in pts who received second infusions of CTX130. 1 pt (7%) experienced a Gr
≥3 infection. There was a sudden death in 1 pt with William’s syndrome in the context
of a lung infection.
Image:
Summary/Conclusion: We have observed clinically meaningful responses, including CRs
with CTX130, the first CAR T directed against the novel target, CD70. CTX130 has an
acceptable safety profile in pts with heavily pretreated R/R TCL and will be investigated
further in an expansion phase of the study.
S263: PHASE I OPEN-LABEL SINGLE ARM STUDY OF GPRC5D CAR-T CELLS (ORICAR-017) IN PATIENTS
WITH RELAPTSED/REFRACTORY MULTIPLE MYELOMA (POLARIS)
M. Zhang1, Y. Hu1, X. Ding2, Y. Tang2, Y. Yang2, S. Chen2, H. Huang1,*
1Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou; 2Oricell Therapeutics Co., Ltd, Shanghai, China
Background: G-protein–coupled receptor class 5-member D (GPRC5D), a type-C 7-pass
transmembrane receptor protein, is predominantly expressed on malignant plasma cell
phenotype, including most malignant plasma cells from pts with multiple myeloma (MM).
The autologous GPRC5D-directed CAR-T cell (OriCAR-017), owning to an additional proprietary
Ori element, is the standard second generation CAR-T cell with improvement in expansion
and durability of CAR-T cells post-transfusion.
Here we report initial results from the phase I open-label single arm study of GPRC5D
CAR-T Cells (OriCAR-017) in pts with Relapsed/Refractory (R/R) MM (NCT05016778).
Aims: The primary objective in the dose escalation phase was to evaluate safety and
tolerability of OriCAR-017. Secondary endpoints included efficacy and pharmacokinetics
(expansion and persistence of OriCAR-017.
Methods: Key enrolment criteria included adults with measurable MM, R/R or intolerant
to established MM therapies, prior BCMA-targeted therapy allowed. Patient received
lymphodepleting chemotherapy with Fludarabine 30mg/m2 daily and cyclophosphamide 300mg/m2
daily for 3 days followed by a single infusion of OriCAR-017. The trial followed a
standard 3 + 3 design with the following dose cohorts: 1×106/kg, 3×106/kg and 6×106/kg
CAR+ T cells.
Results: Eleven pts with R/R MM were enrolled and underwent apheresis during June
9, 2021 and February 25, 2022. Nine of them have completed OriCAR-017 infusion to
date. 2 pts were suspended infusion due to rapid disease progression.
Of the 9 pts infused, median age of 65 years (range: 41-71) and a median of 6 (range
3-17) prior lines of therapy; 4 (44.4%) prior BCMA CART therapy. 3 (33.3%) pts with
extramedullary plasmacytoma ≥ 1 and 7 (77.8%) pts with bone marrow plasma cells ≥60%
at baseline. Five out of 6 pts had high-risk cytogenetic profiles, including 2 pts
with del(17p).
No dose-limiting toxicities occurred. The most common treatment-related AEs were hematological
toxicities. All pts experienced Cytokine Release Syndrome (CRS, with 8(88.9%) pts
in G1, 1 (11.1%) patient in G2), no G3/4 CRS was observed. All CRS cases were rapidly
relieved after conventional CRS intervention, including tocilizumab and steroids.
No neurologic toxicities were reported to date.
All 9pts median follow up time was 118 days (range33—220). A 100.0% ORR were observed
with 3 (33.3%) pts achieved CR/sCR, 3 pts (33.3%) were VGPR, 3 pts were PR (33.3%).
4 pts relapsed from BCMA CAR-T therapy had responses with 1 sCR, 2 VGPR, 1 PR. All
9 pts were Minimal Residual Disease (MRD) negative in the bone marrow by flow cytometry
(sensitivity: 10-5) at day 28 after infusion, and 6pts continued MRD negative at month
3 and 2 pts at month 6 after infusion.
Robust OriCAR-017 expansion in peripheral blood by using qPCR in 3 dose cohorts, median
peak was 349866.1copies/ml (range 60837.5 – 2240358.5), median time to peak expansion
was 10 days (range 7-14). 8 out of 9 patients detected CAR + cell in bone marrow at
D28 after infusion with median 498048.3copies/ml (range 495.4- 2358538.6).
Image:
Summary/Conclusion: In this phase I study, OriCAR-017 was safe and showed impressive
efficacy among previously heavily treated R/RMM pts. Majority of AEs were transient,
manageable, and reversible. 100% ORR and 100% MRD negative rate, along with favorable
safety evidenced support OriCAR-017 could be a competitive therapy for pts with R/RMM.
Furthermore, pts who had relapsed from BCMA CAR-T therapy may still benefit from OriCAR-017
treatment.
S264: PRELIMINARY ANALYSES OF A NON-GENE-EDITING ALLOGENTIC CAR-T IN CD19+ RELAPSED
OR REFRACTORY NON-HODGIN’S LYMPHOMA
X. Wang1, L. Xue1,*, S. Li1, Q. Fan1, K. Liu2, R. Jin2, X. Yang1, T. Wang2, L. He2,
J. Li2
1Department of Hematology, The First Affiliated Hospital of USTC, Hefei; 2Fundamenta
Therapeutics Inc., Suzhou, China
Background: A novel non-gene-editing allogeneic CAR-T platform was developed on the
base of intracellular retention of membrane proteins, and named ThisCART (This = TCR
and/or HLA-I intracellular sequestered). CD19-directed ThisCART (ThisCART19A) cells
are readily produced with a single lentiviral vector, encoding both a CD19-targeting
CAR and an anti-CD3 single chain antibody with the KDEL peptide fused to its C-termini
(Figure 1). ThisCART19A cells, deficient of surface TCR/CD3 complexes in extended
culture, induced no xegoneic GvHD reaction in murine model. Similar to conventional
CD19-directed CAR-T, they exhibited potent CD19-specific cytotoxicities both in vitro
and in vivo.
Aims: To evaluate the safety, pharmacokinetics and clinical activity of ThisCART19A
in patients with refractory or relapsed CD19 positive B cell malignancies.
Methods: A single-center, open-label, single-arm study (NCT04384393) is ongoing to
evaluate the safety, pharmacokinetics and clinical activity of ThisCART19A in patients
with refractory or relapsed CD19 positive B cell malignancies, including acute or
chronic lymphoblastic leukemia, non-Hodgkin’s lymphoma. Standard or enhanced lymphodepletion
with fludarabine and cyclophosphamide was followed by infusion of ThisCART19A at a
dose range of 0.2-60x10
6
cells/kg. The pharmacokinetics of ThisCART19A cell was assessed by flow cytometry
or quantitative PCR.
Results: As of January 30th, 2022, eight evaluble subjects with aggressive or advanced
non-Hodgkin’s lymphoma received ThisCART19A infusion (Table 1). Subject #3 had received
two subsequent autologous CAR-T therapies and relapsed after each treatment. Adverse
event profile is acceptable: no ≥3 grade of CRS or ICANS and no evidence of GvHD reaction.
Two subjects experienced reversible Hemophagocytic lymphohistiocytosis with decreased
lymphocytes and neutrophils. Dose-dependent CAR-T expansion was observed in six subjects
pretreated with enhanced lymphodepletion. Lack of CAR-T expansion in the other two
is likely due to the standard lymphodepletion or quick disease progress. PET/CT (≥28
days) scan demonstrated one partial response and five complete responses. Among these
five, three remain in remission with one in continued follow-up over 8 months.
Table 1
Patient baseline characteristics.
Patients No.
Pt1
Pt2
Pt3
Pt4
Pt5
Pt6
Pt7
Pt8
Age/Sex
41/F
54/M
42/M
58/F
44/M
40/M
67/F
51/F
Pathological type
DLBCL
MCL
DLBCL
DLBCL
tFL-DLBCL
FL
DLBCL
MZL
Ann Arbor/ IPI/FLIPI
IV B/4
IV A/3
IV B/3
III B/4
IV B/3
IV A/4
IV B/3
IV B/1
Since Initial Diagnosis
3 yrs
1 yr
4 yrs
1 yr
2 yrs
1 yr
1 yr
3yrs
Extranodal disease
Yes
No
Yes
Yes
No
Yes
Yes
Yes
Lines of prior therapies
4
4
4
5
7
5
2
3
Prior auto CAR T
No
No
Auto CART CD19
Auto CART CD20
No
No
No
No
No
R/R
Refractory
Refractory
Relapsed
Refractory
Relapsed
Refractory
Relapsed
Relapsed
Best Response
PR
CR
CR
CR
CR
CR
PD
PD
Image:
Summary/Conclusion: ThisCART19A cells demonstrated potent CD19-dependent cytotoxicity
and no xenogentic GvHD in murine models. In this preliminary analysis on evaluable
lymphoma subjects, ThisCART19A exhibited an acceptable safety and an encouraging clinical
response profiles, both comparable to those for marketed autologous products.
S265: RADIOMICS AND ARTIFICIAL INTELLIGENCE FOR IDENTIFICATION AND MONITORING OF SILENT
CEREBRAL INFARCTS IN SICKLE CELL DISEASE: FIRST ANALYSIS FROM THE GENOMED4ALL EUROPEAN
PROJECT
M. P. Boaro1, R. Biondi2, N. Biondini2, A. Collado Gimbert3, E. F. JM4, V. Pinto5,
N. Romano6, V. Voi7, G. B. Ferrero8, M. Casale9, M. Cirillo10, G. Palazzi11, F. Cavalleri12,
G. L. Forni5, G. Reggiani1, S. Perrotta13, M. Manu Pereira14, S. Zazo15, K. Marias16,
M. De Montalembert17, P. Bartolucci18, E. van Beers19, F. Alvarez20, F. Cremonesi21,
T. Sanavia22, P. Fariselli22, G. Castellani23, R. Manara24, R. Colombatti1,*
1Woman’s and Child’s Health, University of Padova, Padova; 2Of Experimental, Diagnostic
and Specialty Medicine, University of Bologna, Bologna, Italy; 3Servei d’Oncologia
i Hematologia Pediàtriques, Hospital Universitari Vall d’Hebron; 4Radiology, Hospital
Universitari Vall d’Hebron, Barcelona, Spain; 5Centro della Microcitemia, Anemie Congenite
e Dismetabolismo del Ferro; 6Department of Diagnostic and Interventional Neuroradiology,
Ospedali Galliera, Genova; 7Dipartimento di Scienze Cliniche e Biologiche, Università
di Torino, Ospedale San Luigi Gonzaga, Orbassano; 8Dipartimento di Scienze Cliniche
e Biologiche, University of Torino, Torino; 9Donna, del Bambino e di Chirurgia Generale
e Specialistica, Università̀ degli Studi della Campania “Luigi Vanvitelli”; 10Department
of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli,
Napoli; 11Dipartimento Integrato Materno Infantile, Azienda Ospedale-Università di
Modena e Reggio Emilia; 12Neurological Department, University of Modena, Modena; 13Dipartimento
della Donna, del Bambino e di Chirurgia Generale e Specialistica, Università̀ degli
Studi della Campania “Luigi Vanvitelli”, Napoli, Italy; 14Vall d’Hebron Research Institute,
Barcelona; 15Information Processing and Telecommunications Center, Universidad Politécnica
de Madrid, Madrin, Spain; 16Computational Medicine Laboratory, Institute of Computer
Science, FORTH, Heraklion, Greece; 17Department of Pediatrics, Hopital Necker, Paris;
18Red Cell Genetic Disease Unit, Université Paris Est, Institut Mondor de Recherche
Biomédicale (IMRB), Creteil, France; 19Universitair Medisch Centrum Utrecht, Utrecht,
Netherlands; 20Universidad Politecnica Madrid, Madrid, Spain; 21Datawizard s.r.l,
Roma; 22Medical Sciences, Division of Gastroenterology and Hepatology, Città della
Salute e della Scienza di Torino, University of Turin, Torino; 23Department of Physics,
University of Bologna, Bologna; 24Neuroscience, University of Padova, Padova, Italy
Background: The use of Artificial Intelligence (AI) for personalized medicine has
recently guided improvements in the diagnostic pathway of many diseases. The EU Project
GENOMED4ALL: “Genomics and Personalized Medicine for All through Artificial Intelligence
in Haematological Diseases” aims at using European data of patients affected by Sickle
Cell Disease (SCD) to find correlation between -omics data – and phenotype, seizing
the opportunity to improve diagnostics through AI.
Silent Cerebral Infarcts (SCIs) are a significant cause of morbidity in SCD: they
affect 25% of children by the age of 6 and 40% by the age of 18 with consequences
on cognition, schooling, working capacity and quality of life. Hence, one of the aims
of the SCD clinical case in GENOMED4ALL is the use of radiomics – quantitative method
for the evaluation and interpretation of medical images- and AI firstly to develop
an automatic and uniform identification and characterization of SCI on MRIs, secondly,
to correlate imaging data with other types of omics data in order to predict risk
of recurrence
Aims: The first phase of the GENOMED4ALL project aimed at collecting anonymized MRI
data available in Centers of the EuroBloodNet Network to develop an algorithm that
could identify SCI on MRI from different sources, distinguishing them from other lesions
Methods: MRI protocol included 3D-T1, FLAIR and DWI sequences. Neuroradiological reports
were checked for consistency. A stepwise segmentation (identification of lesion volume),
pre-processing and extraction of MRIs was performed utilizing different open-source
software: Lesion Segmentation Tools and UNet for segmentation, Freesurfer for brain
extraction. To optimize SCI identification, the best software for segmentation was
selected
Results: Six SCD expert centers participated in the first phase with 501 MRIs: 225
were classified as abnormal by the local neuroradiologists due to presence of SCI;
70% were pediatric MRIs. Different instruments were used in the centers: Philips 1.5
T (n.2), Siemens 1.5 T and 3 T (n.2), GE 3T (n.2).
A stepwise procedure allowed optimization of SCI identification, with distinction
between SCI and non-clinically significant background (periventricular areas) or other
white matter hyperintensities (transient glial maturation).
As shown in Figure 1, to segment SCI in FLAIR MRI, we used a pre-trained UNet [Li
H, 2018] winner of the MICCAI challenge. Firstly, we extracted the brain registering
the MNI152 on the T1 image. Then we applied the brain mask and a threshold taking
only the largest component. The UNet performed the segmentation of hyperintensities.
We removed the regions surrounded by less than 90% of White Matter or near the brain
ventricles to remove non-SCI region (refinement). We then enlarged the remaining ones.
To date, segmentation on 303 exams from different centers showed very few false positive
and some false negatives highlighting the need to take into account different technical
characteristics (various equipment in different centers)
Image:
Summary/Conclusion: SCD is a rare systemic disorder with extreme phenotypic variability.
Radiomics and AI offer the opportunity to seize the potential of big dataset analysis
to understand natural history and optimize diagnostics. SCI can be detected automatically
from different datasets. Four more European Centers will add their MRI in the second
phase, in order to increase variability and allow correlation of detailed Radiomics
features with clinical-hematological variables and other omics data
S266: OXYGEN GRADIENT EKTACYTOMETRY-DERIVED BIOMARKERS ARE ASSOCIATED WITH THE OCCURRENCE
OF ACUTE COMPLICATIONS IN SICKLE CELL DISEASE
M. Rab1,*, C. Kanne2, C. Boisson3, J. Bos1, B. van Oirschot1, M. Houwing4, C. Renoux5,
R. Schutgens6, M. Bartels6, A. Rijneveld7, E Nur8, M. Cnossen4, P. Joly5, R. Fort9,
P. Connes3, R. van Wijk1, V. Sheehan2, E. van Beers6
1Central Diagnostic Laboratory-Research, University Medical Center Utrecht, Utrecht,
Netherlands; 2Department of Pediatrics, Emory University School of Medicine, Atlanta,
United States of America; 3Laboratory LIBM EA7424, University of Lyon; 1, Lyon, France;
4Department of Pediatric Hematology, Erasmus University Medical Center, Rotterdam,
Netherlands; 5Laboratory of Biochemistry and Molecular Biology, Hospices Civils de
Lyon, Lyon, France; 6Van Creveldkliniek, University Medical Center Utrecht, Utrecht;
7Department of Hematology, Erasmus University Medical Center, Rotterdam; 8Department
of Hematology, Amsterdam University Medical center, Amsterdam, Netherlands; 9Department
of Internal Medicine, Hospices Civils de Lyon, Lyon, France
Background: Sickle cell disease (SCD) is a monogenetic disorder with a highly complex
pathophysiology. There is an unmet need for robust reproducible biomarkers that can
assess red blood cell (RBC) function and predict disease severity and complications.
Aims: To explore the association between oxygen gradient ektacytometry-derived biomarkers,
and blood viscosity with incidence of major (acute) SCD-related complications.
Methods: Oxygen gradient ektacytometry measures RBC deformability continuously while
the sample is gradually deoxygenated and subsequently reoxygenated, and identifies
the oxygen tension at which sickling occurs. We examined associations between the
occurrence of acute chest syndrome, cerebral infarction and vaso-occlusive crisis
(VOC) and known biomarkers such as fetal hemoglobin, blood viscosity as well as exploratory
oxygen gradient ektacytometry-derived biomarkers in an adult cohort of 50 individuals
with SCD (HbSS or HbS/βo-thalassemia) and a pediatric cohort consisting of 177 children
with SCD (HbSS or HbS/βo-thalassemia). A substantial number of subjects were on hydroxyurea
therapy (64% of adults and 89% of children). Subjects that received a blood transfusion
less than three months prior to measurements were excluded from the study. A logistic
regression analysis was performed; odds ratios were adjusted for age and hydroxyurea
therapy.
Results: In the adult cohort, for every 10 mmHg increase in Point of Sickling (PoS,
the pO2 tension where RBCs start to sickle, reflecting sickling tendency) the likelihood
of >1 acute complication increased; the adjusted odds ratio (aOR) was 3.00 (p=0.015).
For every 0.1 increase in EImax (reflecting RBC deformability at normoxia), the aOR
was 0.33 (p=0.035, Table 1). In the pediatric cohort, for every 10 mmHg increase in
PoS, the likelihood of >1 acute complication increased; the aOR was 1.65 (p=0.006).
For every 0.1 increase in EImin (reflecting RBC deformability at hypoxia), the aOR
was 0.50 (p=0.007). Fetal hemoglobin and blood viscosity levels were not associated
with likelihood of multiple acute complications. However, fetal hemoglobin was associated
with reduced likelihood of VOC in adults (aOR of 0.32 for every 10% increment, p=0.010)
but not in children (aOR of 0.68, p=0.231, data not shown). In the adult cohort higher
EImax was associated with reduced likelihood of VOC (aOR 0.31, p=0.029). There was
a trend found for an association between higher PoS and greater likelihood of VOC
(aOR 2.22, p=0.050), and no association for EImin. In the pediatric cohort only EImin
was associated with VOC (aOR 0.68, p=0.036).
Image:
Summary/Conclusion: These findings indicate that oxygen gradient ektacytometry generates
novel clinically relevant biomarkers and provide a rationale for further development
of these biomarkers in the evaluation of novel therapies, as part of clinical care,
or clinical trial endpoints. In particular, in assessment of treatment strategies
that do not target HbF induction, such as pyruvate kinase activators, voxelotor and
l-glutamin, oxygen gradient ektacytometry can generate relevant biomarkers.
S267: PROPERDIN-BLOCKING ANTIBODIES ATTENUATE COMPLEMENT ALTERNATIVE PATHWAY ACTIVATION
TRIGGERED BY CELL-FREE HEME IN SICKLE CELL DISEASE MODELS
T. Trovati Maciel1, R. Cofiell2, C. Carvalho1, S. Allali1, R. Rignault1, A. Bennet3,
Y. Dai3, P. Tamburini4, C. Gasteyger5, O. Hermine1, S. K. Kim2,*
1Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutical
Implications, Imagine Institute, Inserm, Paris, France; 2Translational Research, Alexion
AstraZeneca Rare Disease, New Haven, CT; 3Clinical Development and Translational Sciences,
Alexion AstraZeneca Rare Disease, Boston, MA; 4Research, Alexion AstraZeneca Rare
Disease, New Haven, CT, United States of America; 5Clinical Development and Translational
Sciences, Alexion AstraZeneca Rare Disease, Zurich, Switzerland
Background: Complement activation may play an important role in sickle cell disease
(SCD) pathophysiology. Heme has been identified as a trigger of complement activation
in SCD patients and is present in excess during severe vaso-occlusive crises. Heme
is also a DAMP (damage associated molecular pattern) and induces thrombo-inflammation
through tissue factor expression.
ALXN1820 is a bi-specific VHH nanobody that simultaneously binds human albumin and
properdin, thereby effectively and selectively inhibiting alternative pathway (AP)
activation and formation of C3 and C5 convertases. The safety of ALXN1820 has been
assessed in non-human primates and is currently being tested in a phase 1 study in
healthy participants.
Aims: 1) To assess if complement activation and heme are functionally linked; 2) To
investigate the therapeutic potential of ALXN1820 in in vitro SCD models and in in
vivo SCD models using a mouse properdin-blocking monoclonal antibody (mAb), 14E1.
Methods:
In vitro, complement inactivated C3b (iC3b) and C5b-9 deposition on red blood cells
(RBCs) from patients with SCD and complement iC3b and C5b-9 deposition on the endothelial
cell line HMEC-1 were assessed by flow cytometry after exposure to heme +/- ALXN1820.
In vivo, in Townes SCD mice, vaso-occlusion and hyper-hemolysis were induced by intravenous
heme injection (50 µmol/kg) or hypoxia-reoxygenation (8% O2 for 3h then 21% O2 for
1h) in the presence and absence of 14E1 (40 mg/kg), a mouse-specific properdin inhibitor.
Vaso-occlusion was measured by staining RBCs with immunofluorescence-labeled Ter-119
antibodies on lung and liver sections. C3b and C5b-9 deposition on SCD RBCs was assessed
by flow cytometry. Markers of hemolysis (bilirubin, lactate dehydrogenase, free hemoglobin,
free heme) were quantified in plasma using commercial assays.
Results: In RBCs from patients with SCD, cell-free heme triggered marked iC3b and
C5b-9 deposition (Table). Deposition was blocked in the presence of ALXN1820 by >95%
for iC3b and by >85% for C5b-9. HMEC-1 cells exposed to heme showed marked deposition
of iC3b and C5b-9. In the presence of ALXN1820, deposition was blocked by >70% for
iC3b and >85% for C5b-9. Pretreatment of mice with 14E1 markedly ameliorated vaso-occlusion
and reduced both C3b and C5b-9 deposition on RBCs, and hemolysis biomarkers.
Image:
Summary/Conclusion: These data strengthen the hypothesis that in SCD, cell-free heme
is a potent trigger of complement AP activation, which can be blocked by targeting
properdin. Investigating the efficacy and safety of anti-properdin ALXN1820 in patients
with SCD is warranted as a novel approach to treatment of acute and chronic SCD complications.
S268: SAFETY, TOLERABILITY, AND PHARMACOKINETIC/PHARMACODYNAMIC RESULTS FROM PHASE
1 STUDIES OF GBT021601, A NEXT-GENERATION HBS POLYMERIZATION INHIBITOR FOR TREATMENT
OF SICKLE CELL DISEASE
C. Brown1,*, C. Key2, I. Agodoa3, J. Olbertz3, K. Duchin3, A. Barth3, E. Lisbon3
1Aflac Cancer and Blood Disorder Center of Children’s Healthcare of Atlanta and Department
of Pediatrics, Emory School of Medicine, Atlanta; 2ICON plc, San Antonio; 3Global
Blood Therapeutics, South San Francisco, United States of America
Background: Sickle cell disease (SCD) is caused by polymerization of sickle hemoglobin
(HbS). Voxelotor is a first-in-class HbS polymerization inhibitor approved by the
United States Food and Drug Administration for the treatment of SCD in adult and pediatric
patients aged ≥4 years, and by the European Medicines Agency for the treatment of
hemolytic anemia due to SCD in adult and pediatric patients aged ≥12 years as monotherapy
or in combination with hydroxycarbamide. GBT021601 is a next-generation HbS polymerization
inhibitor with improved pharmacokinetic (PK) properties that stabilizes Hb in the
oxygenated state, thus inhibiting polymerization. GBT021601 has the potential to achieve
higher Hb occupancies at lower doses than voxelotor, potentially reducing treatment
burden and improving clinical outcomes for patients with SCD.
Aims: To explore the safety, tolerability, PK, and pharmacodynamics of GBT021601 in
healthy volunteers and patients with SCD.
Methods: Two studies are in progress: (1) GBT021601-011: a phase 1, randomized, double-blind,
placebo-controlled, parallel-group study of single ascending doses (SADs) and multiple
ascending doses (MADs) of GBT021601 in healthy volunteers aged 18 to 55 years; and
(2) GBT021601-012: a phase 1, single-arm, intrapatient, single-dose and MAD study
in patients with SCD (homozygous HbS) aged 18 to 60 years. In the GBT021601-011 SAD
phase, participants (n=8) were randomized 6:2 to receive a single oral dose of GBT021601
(50 to 2200 mg) or placebo. In the GBT021601-011 MAD phase, participants (n=10) were
randomized 7:3 to 14 days of GBT021601 (3 days of loading doses followed by 11 days
of once-daily maintenance doses) or placebo. Data from the 50 mg MAD cohort are reported
in this analysis. In the GBT021601-012 study, patients with SCD (N=6) received a single
100 mg dose of GBT021601, followed by an 8-week washout, then 2 continuous MADs: once-daily
maintenance doses of 50 mg (5 weeks) then 100 mg (3 weeks). Each MAD phase participant
received 2 days of loading doses.
Results: In the GBT021601-011 50 mg MAD cohort, blinded safety data demonstrated that
GBT021601 was well tolerated. Adverse events (AEs) were predominantly grade 1 and
not related to treatment. Drowsiness, headache, and constipation were the most frequent
AEs. Mean (SD) Hb occupancy of GBT021601, determined via PK analysis, was 25.8% (3.9%).
In GBT021601-012, patients with SCD completed treatment with no AEs leading to study
discontinuation. Three vaso-occlusive crises (grades 2 and 3) occurred in 2 patients
and were deemed unrelated to GBT021601. After dosing for 8 weeks, the mean (SD) Hb
occupancy achieved was 32.6% (9.4%), and the mean (SD) increase in Hb was 2.3 (0.9)
g/dL. Markers of hemolysis decreased, with a mean (SD) change in absolute reticulocytes
of –37.4% (17.3%), reticulocyte percentage of –54.0% (11.3%), indirect bilirubin of
–6.6% (119.3%), and lactate dehydrogenase of –33.1% (15.4%). The terminal elimination
half-life of GBT021601 from blood averaged about 10 days. Ektacytometry scans demonstrated
improvements in red blood cell (RBC) health, and blood smears showed increased counts
of RBCs with improved morphology (Figure); no changes in erythropoietin levels were
observed.
Image:
Summary/Conclusion: Multiple daily doses of GBT021601 were well tolerated in both
studies. In patients with SCD, the GBT021601 100 mg maintenance dose led to a mean
Hb occupancy >30%, increased Hb, reduced markers of hemolysis, and improved RBC health.
These data highlight the potential of GBT021601 and support its further development.
S269: MACROPHAGE FUNCTIONAL AND METABOLIC REWIRING THROUGH PPAR/PGC1 MODULATION IMPROVES
HEME/IRON-DRIVEN DEFECTIVE EFFEROCYTOSIS AND PROMOTES TISSUE DAMAGE RESOLUTION IN
SICKLE CELL DISEASE
R. Sharma1, S. Z. Vance1, A. Antypiuk1, D. Manwani2, F. Vinchi1,*
1Iron Research Laboratory, New York Blood Center; 2Pediatric Hematology, The Children’s
Hospital at Montefiore, New York, United States of America
Background: Sickle cell disease (SCD) is hallmarked by an underlying chronic inflammatory
status, which is contributed by pro-inflammatory macrophages. Macrophages are highly
plastic immune cells which integrate and respond to a variety of signals they are
exposed to in the surrounding microenvironment. One of these stimuli is represented
by heme, which is released from oxidized hemoglobin upon hemolysis and acts as damage-associated
molecular pattern to stimulate macrophage pro-inflammatory phenotypic switching through
TLR4 signaling activation and ROS production. Heme-induced M1 pro-inflammatory activation
program in macrophages promotes sterile inflammation and aggravates hepatic fibrosis
in SCD, contributing to organ damage typically associated with this disease.
Aims: While previous studies addressed heme ability to induce inflammatory cytokine
production in macrophages, how heme alters cell functional properties and whether
this exacerbates tissue injury remain unexplored. Macrophage functions as immune cell
recruitment ability and apoptotic cell clearance are relevant in the context of SCD,
where tissue damage and cell apoptosis occur frequently due to vaso-occlusive episodes,
hypoxia and ischemic injury.
Methods: Here we analyzed macrophage response to apoptotic stimuli in vivo, in mouse
models of heme overload and SCD, as well as in vitro, in bone-marrow-derived macrophages,
to unveil the impact of hemolysis on macrophage functional properties.
Results: Our results demonstrate that, in addition to inflammatory activation, heme
strongly alters macrophage functional response to apoptotic cell damage by exacerbating
their immune cell recruitment ability and impairing their efferocytic capacity. We
show that exposure to heme and iron excess drives defective efferocytosis of apoptotic
neutrophils in vitro, in cultured bone-marrow-derived macrophages, and in vivo, in
mouse model of heme overload. This is fully recapitulated in SCD mice, where limited
efferocytosis contributes to impaired apoptotic cell clearance and exacerbated tissue
damage. Mechanistically, altered efferocytosis depends on heme-driven activation of
TLR4 signaling pathway and the suppression of the transcription factor PPARγ and its
coactivator PGC1α. These changes lead to reduced expression of efferocytic receptors
and impaired mitochondrial biogenesis as well as mitochondrial dysfunction, and is
associated with metabolic skewing. This results in limited recognition and engulfment
of apoptotic cells and decreased shift to aerobic mitochondrial fatty acid β-oxidation
and anti-inflammatory IL-4/10 release, with consequent inhibition of continual efferocytosis,
inflammation resolution and tissue repair. We further demonstrate that impaired phagocytic
capacity and tissue damage are improved by hemopexin-mediated heme scavenging and
anti-inflammatory IL-4 treatment in SCD mice through phenotypic and functional macrophage
rewiring. Interestingly, defective efferocytosis is reproduced in vitro by macrophage
exposure to SCD patients’ plasma and rescued by hemoglobin/heme scavenging via haptoglobin
and hemopexin and PPARγ/PGC1α modulation via PPARγ agonist or IL-4.
Summary/Conclusion: Our data indicate that the therapeutic improvement of heme-altered
macrophage functional properties via heme scavenging or PPARγ/PGC1α modulation promotes
the resolution of inflammation and ameliorates tissue damage associated with SCD pathophysiology.
Thus, macrophage functional rewiring offers potentially valuable therapeutic strategies
for SCD patients to improve tissue damage resolution upon vaso-occlusive crisis and
ischemic events.
S270: LONGER-TERM ANALYSIS OF EFFICACY OF LUSPATERCEPT VERSUS PLACEBO IN PATIENTS
WITH TRANSFUSION-DEPENDENT BETA-THALASSEMIA ENROLLED IN THE BELIEVE STUDY
M. D. Cappellini1,*, A. T. Taher2, J. B. Porter3, K. H. Kuo4, T. D. Coates5 6, E.
Voskaridou7, G. L. Forni8, S. Perrotta9, A. Khelif10, A. Lal11, A. Kattamis12, A.
Piga13, O. Hermine14 15, N. Holot16, F. Lersch17, J. K. Shetty17, S. Vodala16, J.
Zhang16, D. Miteva17, V. Viprakasit18
1Fondazione IRCCS Ca’ Granda Policlinico Hospital, University of Milan, Milan, Italy;
2Department of Internal Medicine, American University of Beirut Medical Center, Beirut,
Lebanon; 3University College London, University College London Hospitals, London,
United Kingdom; 4Division of Medical Oncology and Hematology, Department of Medicine,
University Health Network, Division of Hematology, Department of Medicine, University
of Toronto, Toronto, ON, Canada; 5Children’s Center for Cancer and Blood Diseases,
Children’s Hospital Los Angeles; 6USC Keck School of Medicine, Los Angeles, CA, United
States of America; 7Thalassemia and Sickle Cell Center, Laiko General Hospital, Athens,
Greece; 8Centro della Microcitemia e Anemie Congenite e del Dismetabolismo del Ferro,
Ospedale Galliera, Genoa; 9Dipartimento della Donna, del Bambino e di Chirurgia Generale
e Specialistica, Università della Campania, Luigi Vanvitelli, Naples, Italy; 10Farhat
Hached Teaching Hospital, Sousse University, Sousse, Tunisia; 11University of California
San Francisco Benioff Children’s Hospital, Oakland, CA, United States of America;
12First Department of Pediatrics, National and Kapodistrian University of Athens,
Athens, Greece; 13University of Turin, Turin, Italy; 14Department of Hematology, Hôpital
Necker, Assistance Publique Hôpitaux de Paris, University Paris Cité; 15INSERM U1163
and CNRS 8254, Imagine Institute, Université Sorbonne Paris Cité, Paris, France; 16Bristol
Myers Squibb, Princeton, NJ, United States of America; 17Celgene International Sàrl,
a Bristol-Myers Squibb Company, Boudry, Switzerland; 18Siriraj Hospital, Mahidol University,
Bangkok, Thailand
Background: Ineffective erythropoiesis and anemia are hallmarks of β-thalassemia.
Luspatercept, a first-in-class erythroid maturation agent, has shown efficacy in the
phase 3 BELIEVE (NCT02604433) trial in patients (pts) with β-thalassemia requiring
regular red blood cell transfusions (RBCT). However, longer-term analyses of efficacy
are yet to be reported.
Aims: To observe and report the longer-term effect of luspatercept treatment on transfusion
burden and liver iron concentration (LIC).
Methods: Pts ≥18 years of age with β-thalassemia or hemoglobin E/β-thalassemia (compound
β-thalassemia mutation and/or multiplication of α-globin genes allowed) requiring
regular RBCT (6–20 RBC units/24 wk before randomization with no transfusion-free period
>35 d) were randomized 2:1 to luspatercept 1.0 mg/kg (titration up to 1.25 mg/kg allowed)
or placebo subcutaneously every 3 wk. Response was defined as RBCT burden reduction
of ≥33% (≥33% response) or ≥50% (≥50% response) from baseline (BL; ≥2 units) during
a rolling 12- or 24-wk interval. LIC was determined by MRI.
Results: As of Jan 5, 2021, 125/224 (55.8%) pts randomized to luspatercept completed
144 wk of treatment and 6 (2.7%) pts completed 192 wk. The main reasons for treatment
discontinuation were pt withdrawal (23.7% luspatercept vs 11.6% placebo), adverse
events (10.3% vs 1.8%), and lack of efficacy (1.8% vs 7.1%). More pts receiving luspatercept
vs placebo had ≥33% or ≥50% response during any rolling 12- or 24-wk interval up to
the cutoff date (all P<0.0001); the number of responders receiving luspatercept was
higher compared with previous data cuts (Table). The median (95% CI) longest duration
of response for ≥33% and ≥50% responders (rolling 12-wk interval) in the luspatercept
group increased from the primary data cut to 114.0 (107.0–137.0) and 99.0 (95.0–104.0)
d, respectively (Table); median (95% CI) longest duration of response for placebo
pts was 90.0 (86.0–94.0) and 86.0 (84.0–103.0) d, respectively. The median (range)
total duration of response for ≥33% and ≥50% responders (rolling 12-wk interval) in
the luspatercept group was 586.0 (84–1300) and 357.5 (84–1267) d, respectively, and
171.0 (84–627) and 169.0 (84–485) d in the placebo group. The 6 pts who completed
192 wk of luspatercept required on average 6.41 (SD 4.32) fewer RBC units during wk
145–192 compared with BL. Overall mean change from BL in transfusion window for ≥50%
responders (any 24-wk interval) was increased by 9.88 d (SD 22.04, n=51). More pts
receiving luspatercept (27/224 [12.1%]) than placebo (2/112 [1.8%]) achieved RBCT
independence (RBC-TI) ≥8 wk (P=0.0015), an increase from previous data cuts (Table).
Median (95% CI) longest durations of RBC-TI were 72.0 (62.0–103.0) d for luspatercept
and 71.5 (62.0–NA) for placebo, though the placebo result was driven by 2 pts. Longer-term
treatment with luspatercept resulted in a decreasing trend in LIC compared with BL
for ≥33% responders (rolling 12-wk interval) with a mean (SD) change from BL in LIC
at wk 144 (n=25) of −3.73 (9.08) mg/g dry weight.
Image:
Summary/Conclusion: Continued treatment with luspatercept resulted in more pts experiencing
reduced RBCT burden and longer duration of response compared with previous data cuts.
RBC units transfused decreased over the longer-term treatment period and the time
between transfusions increased compared with BL for ≥50% responders. Longer-term luspatercept
treatment also resulted in more pts experiencing RBC-TI ≥8 wk and a potential decreasing
trend in LIC; LIC analysis is ongoing. These data indicate that pts continue to benefit
from longer-term luspatercept treatment.
S271: BONE MARROW TFR2 DELETION IMPROVES THE THERAPEUTIC EFFECT OF ACTIVIN LIGAND
TRAP RAP-536 IN Β-THALASSEMIC MICE
E. Tanzi1 2, S. M Di Modica1, J. Bordini3, V. Olivari1 4, M. Pettinato1, A. Pagani1,
A. Campanella3 4, L. Silvestri1 4, A. Nai1 4,*
1Division of Genetics and Cell Biology, Ospedale San Raffaele; 2University of Milan
Bicocca; 3Division of Experimental Oncology, Ospedale San Raffaele; 4Vita-Salute San
Raffaele University, Milan, Italy
Background: β-thalassemia is a disorder due to mutations in the β-globin gene, characterized
by ineffective erythropoiesis (IE), anemia, and iron overload, whose treatment still
requires improvement. Luspatercept, an activin receptor-ligand trap that promotes
terminal erythroid differentiation downregulating the TGF-β pathway is a recently
approved drug for treatment of transfusion dependent β-thalassemia (TDT) and in phase
3 trial for non TDT (NTDT). However, its precise mechanism of action remains to be
elucidated and the possible connection with the erythropoiesis driving cytokine erythropoietin
(EPO) is still controversial. In addition, Luspatercept is only partially effective
in correcting anemia, thus the identification of strategies for enhancing its efficacy
would be of clinical benefit.
The second transferrin receptor (TFR2) balances red blood cells (RBC) production with
iron availability, contributing to the activation of the iron hormone hepcidin in
the liver and acting as an EPO receptor partner in erythroid cells. We have demonstrated
that bone marrow (BM) Tfr2 deletion increases erythroblast EPO sensitivity causing
erythrocytosis in wild-type mice (Nai et al, Blood 2015) and improving IE, anemia,
RBCs morphology and iron-overload in NTDT mice (Artuso et al, Blood 2018).
Aims: As we have previously demonstrated that the combined targeting of different
pathophysiologic cues of the disease results in a synergistic benefit on thalassemic
erythropoiesis and anemia (Casu, Pettinato, et al, Blood 2020), here we investigate
whether BM Tfr2 genetic inactivation improves the efficacy of Luspatercept in a NTDT
model (Hbb
th3/+
).
Methods:
Hbb
th3/+
mice with BM Tfr2 deletion (Tfr2
BMKO
/Hbb
th3/+
) and Hbb
th3/+
controls were treated with RAP-536 (Celgene, BMS group), a murine analogue of Luspatercept,
10mg/kg, twice a week for 8 weeks. Complete blood count was evaluated every two weeks.
At sacrifice, blood and organs were collected for full phenotypic analysis.
Results: As expected, BM Tfr2 deficiency strongly ameliorated anemia and IE of Hbb
th3/+
mice (hemoglobin, Hb=13.48±0.19g/dL vs 8.65±0.24g/dL in controls, P<0.000001). RAP-536
moderately increased RBC count, Hb and hematocrit after 2 weeks of treatment, and
the beneficial effect was maintained until the end of the protocol with a similar
efficacy in Hbb
th3/+
(Hb=9.54±0.33g/dL) and Tfr2
BMKO
/Hbb
th3/+
mice (Hb=14.48±0.50g/dL), without significant changes in MCV, MCH and reticulocyte
count. Splenomegaly was not affected by RAP-536 in Hbb
th3/+
mice, despite an amelioration of both spleen and BM IE, with reduced proportion of
immature erythroblasts and increased mature RBCs. Erythroid differentiation was further
improved in RAP-536-treated Tfr2
BMKO
/Hbb
th3/+
mice both in the spleen and in the BM, leading to a significant reduction of spleen
size, to values of wt animals.
Hepatic and serum iron levels were reduced in mice lacking Tfr2, likely because of
high erythroid consumption, while RAP-536 did not affect iron homeostasis in both
genotypes.
Summary/Conclusion: BM Tfr2 deletion and RAP-536 treatment had additive effect in
correcting IE and anemia in β-thalassemic mice. Since RAP-536 has comparable therapeutic
efficacy in Hbb
th3/+
and Tfr2
BMKO
/Hbb
th3/+
animals, it improves erythropoiesis through mechanism(s) other than Tfr2 deletion,
thus independent of EPO stimulation. Overall, our results add a piece of information
on the mechanism of action of Luspatercept and suggest that the concomitant TFR2 targeting
might represent a promising option for boosting the therapeutic efficacy of activin
receptor ligand-traps.
S272: SAFETY AND PRELIMINARY PHARMACODYNAMIC EFFECTS OF THE FERROPORTIN INHIBITOR
VAMIFEPORT (VIT-2763) IN PATIENTS WITH NON-TRANSFUSION-DEPENDENT BETA THALASSEMIA
(NTDT): RESULTS FROM A PHASE 2A STUDY
A. Taher1,*, A. Kourakli-Symeonidis2, A. Tantiworawit3, P. Wong4, P. Szecsödy5
1American University of Beirut Medical Center, Beirut, Lebanon; 2University of Patras
– Rio Regional University Hospital, Rio, Greece; 3Chiang Mai University – Faculty
of Medicine, Chiang Mai; 4Naresuan University Hospital, Mueang Phitsanulok, Thailand;
5Vifor Pharma AG, Glattburg, Switzerland
Background: NTDT is characterized by an imbalanced α/β-globin chain ratio, ineffective
erythropoiesis, increased intestinal iron absorption and iron overload. Vamifeport
inhibits ferroportin-mediated iron export into plasma and has been shown to decrease
serum iron and transferrin saturation (TSAT) levels in relevant animal disease models
and healthy volunteers.
Aims: The primary objective of this multicenter, Phase 2a, double-blind, randomized,
placebo-controlled study (NCT04364269) was to assess the safety and tolerability of
vamifeport (given once [QD] or twice [BID] daily) versus placebo in patients with
NTDT over a 12-week treatment period. A secondary objective was to assess preliminary
efficacy of vamifeport versus placebo with respect to pharmacodynamic effects on iron
parameters (serum iron, ferritin, hepcidin and TSAT) in these patients.
Methods: Adults (18–65 years) with documented NTDT (including β-thalassemia intermedia)
who provided informed consent, were included. NTDT was defined as receipt of <5 units
of red blood cells during the 24 weeks prior to randomization. Patients also required
a mean baseline hemoglobin of ≤11 g/dL on two consecutive measurements ≥1 week apart
within 6 weeks prior to randomization. Use of iron chelation therapy (ICT) within
4 weeks prior to randomization was not permitted; patients with TSAT <30%, serum ferritin
<150 ng/mL and/or liver iron content ≤1 mg/g (<300 ng/mL/3 mg/g, respectively, for
those recently on ICT) were also excluded. Patients received vamifeport at a dose
of 60 mg if their body weight was 40–59 kg or 120 mg if their weight was 60–100 kg.
Results: Twenty-five patients were included (vamifeport QD n=9, BID n=12, placebo
n=4; median age 42.0, 31.0 and 40.5 [range 18–61] years, respectively). Overall, 64%
were male, 56% had a body weight <60 kg and 16% had received prior ICT. At baseline,
mean (standard deviation [SD]) serum iron concentrations were 27.2 (11.6) µmol/L and
mean TSAT was 76.2 (27.6)% for the population as a whole. Mean (SD) serum ferritin
concentrations were 746.7 (1431.7) µg/L. There were no differences between groups
in treatment-emergent adverse event (TEAE) rates (vamifeport QD 66%, BID 58%; placebo
75%). All TEAEs were mild or moderate intensity, and each was reported only once.
There were no deaths or serious AEs (1 patient receiving vamifeport QD had an acute
hemolytic event); one TEAE (increased creatine kinase and transaminase levels, not
drug-related) led to drug discontinuation in the vamifeport BID group. There were
also no clinically relevant changes in any assessed safety parameter. Serum iron concentrations
decreased 3 h after first vamifeport dose (Day 1) in all vamifeport-treated patients
(mean [SD] decrease: QD -11.3 [7.2] µmol/L; BID -17.0 [9.6] µmol/L) and were maintained
below baseline levels at 3–4 h post vamifeport administration at each visit throughout
the remaining treatment period. Mean (SD) TSAT also decreased at Day 1 (3 h post dose;
QD -32.6 [19.6]%; BID -46.8 [22.6]%) and were below baseline levels 3–4 h post-dose
at each subsequent visit in the vamifeport groups (Figure). There were no clinically
meaningful changes from baseline in serum iron or TSAT levels in the placebo group,
or in ferritin or hepcidin levels in any treatment group.
Image:
Summary/Conclusion: In this 12-week Phase 2a study, vamifeport had a favorable safety
and tolerability profile and showed promising target engagement and pharmacodynamic
effects on serum iron and TSAT levels versus placebo in adults with NTDT.
S273: EVIDENCE OF NONINFERIORITY OF MITAPIVAT VERSUS SPLENECTOMY IN MURINE HEREDITARY
SPHEROCYTOSIS
A. Matte’1,*, A. Recchiuti2, E. Federti1, P. A. Kosinski3, V. Riccardi1, I. Iatcenko1,
L. Dang3, A. Iolascon4, R. Russo4, C. Brugnara5, A. Janin6, C. Leboeuf6, N. Mohandas7,
L. De Franceschi1
1Dept of Medicine, University of Verona, Verona; 2Dept of Medical, Oral, and Biotechnological
Science, “G. d’Annunzio” University of Chieti, Chieti, Italy; 3Agios Pharmaceuticals,
Cambridge, United States of America; 4Dept. of Molecular Medicine and Medical Biotechnology
and CEINGE, University of Naples Federico II, Naples, Italy; 5Dept of Laboratory Medicine,
Boston Children’s Hospital - Harvard Medical School, Boston, United States of America;
6University Diderot of Paris, Paris, France; 7Laboratory of Red Cell Physiology, New
York Blood Center, New York, United States of America
Background: Hereditary spherocytosis (HS) is the most common cause of inherited red
cell membranopathy, due to mutations in genes encoding membrane or cytoskeletal proteins,
including band 3, ankyrin, spectrin or band 4.2. Loss of membrane cohesion results
in surface area loss and generation of spherocytic red cells with decreased cellular
deformability and reduced red cell survival due to splenic sequestration. Relative
PK deficiency has been observed in HS red cells (Andreas O et al. BJH 187: 386, 2019).
Splenectomy is the gold standard treatment of anemia in patients with HS. However,
the benefits of splenectomy in mild HS is still controversial and its cost/effectiveness
might be underestimated. Using band 4.2-/- mice, a well-established model of HS (Peters
LL et al JCI 103: 1527, 1999), we recently show that mitapivat, an oral PK activator,
ameliorates anemia, reduces chronic hemolysis and improves RBC features in 4.2-/-
mice (Matte A et al. EHA 2021).
Aims: To understand whether the oral pyruvate kinase activator, mitapivat, is not
less efficacious than splenectomy in treatment of anemia in 4.2-/- mice.
Methods: Splenectomized 4.2-/- mice (2.5 months of age) were either treated with vehicle
or mitapivat at dosages of 100 mg/kg/day over 4 months. An unsplenectomized group
of 4.2-/- mice treated with and without mitapivat was also analyzed. The following
parameters were evaluated: hematologic parameters, markers of hemolysis, erythropoiesis
and iron homeostasis.
Results: In band 4.2-/- mice, splenectomy resulted in (i) amelioration of anemia with
reduced reticulocyte count (Hb 13.3 ±0.5 g/dL, n=3 vs 11.9±0.6 g/dL, n=8; P<0.05;
retics: 4.9±0.5%, n=3 vs 10.0±1.4%, n=8 P<0.05) and reduced hemolytic indices (LDH,
total bilirubin) when compared to non-splenectomized 4.2-/- mice. The decreased hemolysis
was associated with reduction in serum erythropoietin (EPO). Mitapivat was non-inferior
to splenectomy for amelioration of anemia (Hb 13.4±0.8 g/dL, n=11 vs 11.9±0.6 g/dL,
n=8), reduction in reticulocyte count (retics: 7.8±1.4%, n=11 vs 10.0±1.4%, n=8 P<0.05)
and EPO levels (401±123 U/L, n=3). Noteworthy, in splenectomized 4.2-/- mice, mitapivat
further improved Hb (Hb: 14.1±0.3 g/dL, n=3), reduced reticulocyte counts (retics:
3.6±0.2%, n=3) and EPO levels when compared to vehicle treated splenectomized 4.2-/-
mice.
Image:
Summary/Conclusion: Our data indicate that mitapivat was at least as effective as
splenectomy in treatment of anemia in a murine model of HS.
S274: A NOVEL SUBTYPE OF ANEMIA CAUSED BY MUTATIONS IN TFRC GENE
S. Colucci1 2,*, V. Venturi3, F. Nicole1 2, D. Jové Solavera3, M. Zimon4, P. Richter-Pechanska1,
G. Hernandez3 5, S. Unal6, F. Gumruk6, A. Diaz-Conradi7, L. Romero-Cortadellas3, X.
Ferrer-Cortès3 5, M. Olivella8, M. Erlacher9, C. Niemeyer9, T. Wiesel10, R. Pepperkok4,
M. D. Fleming11, A. E. Kulozik1 2, M. Sanchez3 5, M. U. Muckenthaler1 2
1Department of Pediatric Oncology, Hematology, and Immunology, Heidelberg University;
2Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany; 3Department of Basic
Sciences, Iron metabolism: Regulation and Diseases, Universitat Internacional de Catalunya
(UIC), Sant Cugat del Vallès, Spain; 4European Molecular Biology Laboratory (EMBL),
Heidelberg, Germany; 5BloodGenetics S.L. Diagnostics in Inherited Blood Diseases,
Esplugues de Llobregat, Spain; 6Department of Pediatrics, Hacettepe University Faculty
of Medicine, Ankara, Turkey; 7Hospital de Nens de Barcelona, Barcelona; 8Biosciences
Department, Faculty of Sciences and Technology (FCT), University of Vic - Central
University of Catalonia, Vic, Spain; 9Department of Pediatrics and Adolescent Medicine,
Medical Center, University of Freiburg, Freiburg; 10Vestische Kinder- und Jugendklinik
Datteln, University Witten/Herdecke, Datteln, Germany; 11Department of Pathology,
Boston Children’s Hospital, Harvard Medical School, Boston, United States of America
Background: Anemia affects ~25% of the world population and is frequently caused by
iron deficiency as a consequence of malnutrition or inflammation. Only in rare cases
anemia is due to mutations in genes responsible for balancing iron homeostasis. Among
these, loss of function variants of MT2 cause iron-refractory iron-deficiency anemia
(IRIDA); a disease characterized by inappropriately high levels of hepcidin. Hepcidin
is a hepatic hormone that limits dietary iron uptake and iron release from intracellular
stores and thus renders IRIDA patients resistant to iron therapies. In mice, a form
of anemia resistant to iron supplementation is further caused by mutations in Transferrin
Receptor 1 (TfR1; Tfrc gene). TfR1 internalizes iron-bound transferrin, a process
fundamental for erythropoiesis. However, up to date, the sole human report describing
a TfR1 mutation (p.Tyr20His) showed that it impairs the immunological compartment
rather than erythroblast maturation. Here, we report pediatric patients with previously
undescribed mutations in TFRC, diagnosed with microcytic hypochromic anemia, partially
resistant to iron therapies.
Aims: Identification of novel disease alleles that cause anemia.
Methods: Whole exome sequencing (WES) of DNA of patients with anemia. Functional characterization
of novel mutations identified in TFRC gene was assessed in HeLa cells overexpressing
TfR1 wild-type and mutated by western blotting and wide-field microscopy.
Results: A 4-year old boy manifested symptoms of anemia and the analysis of hematological
parameters revealed low levels of hemoglobin, MCV and MCH. Plasma iron, transferrin
saturation and hepcidin were within physiological ranges. Unexpectedly, soluble TfR1
(sTfR1) levels were almost undetectable and the bone marrow smear revealed increased
erythropoiesis and iron deficiency in this compartment. The condition of hypochromic
microcytic anemia was persistent over years and could not be corrected by oral iron
therapy. WES analysis identified a TFRC_c.941C>T homozygous mutation, encoding for
TfR1P314L. Relatives carrying the same mutation in heterozygosity were not anemic.
This indicates that TFRC_c.941C>T mutation causes a recessive form of anemia. Overexpression
of a construct expressing the TfR1P314L mutant in HeLa cells showed a reduction in
total and secreted TfR1, thus reflecting upon the observation of very low sTfR1 levels
in the blood of the patient. We further show that lower TfR1 levels are likely due
to a faster degradation rate of TfR1P314L compared to TfR1WT. In addition, the TfR1P314L
mutated protein showed impaired internalization of fluorescently labeled transferrin,
most likely explaining the reduced iron levels observed in the bone marrow.
In addition to this index patient, 3 additional children of 2 independent families
were identified with TFRC mutations and hypochromic microcytic anemia. The genetic
analysis detected a TFRC_c.967C>G heterozygous mutation encoding for TfR1P323A and
a TFRC_c.934G>A homozygous mutation, encoding for TfR1G312R. All three TFRC mutations
are highly conserved across species and are located in the same extracellular loop
of TfR1.
Summary/Conclusion: We identified a novel subtype of juvenile hereditary hypochromic
microcytic anemia that is partially resistant to iron therapies and caused by missense
mutations in the TFRC gene. Reduced endocytosis of iron-bound transferrin may contribute
to insufficient iron supply for erythropoiesis and resistance to iron supplementation.
In the clinical practice, our findings suggest to include TFRC sequencing analysis
for patients with unexplained anemia.
S275: ALTERATION OF IRON HOMEOSTASIS THROUGH GENETIC AND PHARMACOLOGIC MODULATION
OF FERROPORTIN MODIFIES MDS PATHOPHYSIOLOGY IN A PRECLINICAL MOUSE MODEL
S. Z. Vance1, A. Antypiuk1, R. Sharma1, F. Dürrenberger2, V. Manolova2, F. Vinchi1,*
1Iron Research Laboratory, New York Blood Center, New York, United States of America;
2Vifor (International) Ltd, St. Gallen, Switzerland
Background: Patients with myelodysplastic syndromes (MDS) are prone to develop iron
overload as a consequence of ineffective erythropoiesis and chronic transfusion therapy.
Although iron overload is a common feature in MDS, it remains unclear whether and
how iron excess is detrimental for MDS pathophysiology.
Aims: This study aimed at characterizing altered iron homeostasis in a preclinical
MDS mouse model, understanding the molecular mechanisms underlying iron toxicity and
unraveling the potential therapeutic value of pharmacologic iron restriction in this
disease condition.
Methods: To this end, we took advantage of complementary approaches and analyzed the
effect of iron overload obtained through genetic activation of the iron exporter ferroportin
(FPNC326S), and iron restriction achieved through the administration of the FPN inhibitor
Vamifeport for 3 months in NUP98-HOXD13 MDS mice. Mice were analyzed at 6 months of
age.
Results: MDS mice develop anemia and low white blood cell counts in the peripheral
blood associated with a significant iron overload phenotype, hallmarked by low hepcidin
levels, elevated serum iron and transferrin saturation, non-transferrin-bound iron
(NTBI) formation and tissue iron deposition. FPNC326S MDS mice show an aggravated
iron phenotype, with further elevated NTBI and tissue iron accumulation and still
inappropriately low hepcidin levels. Vamifeport administration in MDS mice reduced
serum iron and NTBI levels and prevented tissue iron loading. While iron excess in
FPNC326S MDS mice did not improve erythropoiesis and hematologic parameters in the
peripheral blood, iron restriction by Vamifeport significantly ameliorated red blood
cell maturation and hemoglobin levels in MDS mice. The improved ineffective erythropoiesis
was associated with reduced oxidative stress and apoptosis in erythroid progenitors.
Unexpectedly, we observed a major role of the iron status in myeloid expansion in
MDS. The number of immature myeloid blasts and myeloid progenitors as well as inflammation
markers in the bone marrow of MDS mice were aggravated by iron overload in FPNC326S
MDS mice and attenuated by iron restriction in Vamifeport-treated MDS mice compared
to control MDS animals. Myeloid bias, monitored as percentage bone marrow myeloid
cells, was exacerbated by iron overload and alleviated by iron restriction. Overall,
this translated into a faster and delayed transition to AML, respectively. Finally,
iron showed a profound impact on the stem cell pool in MDS. Iron overload aggravated
and iron restriction alleviated the formation of reactive oxygen species, accumulation
of DNA damage and levels of the apoptosis marker annexin V in hematopoietic stem and
progenitor cells (HSPCs). By reducing the intracellular labile iron pool, Vamifeport
improved HSPC quality and survival, partially rescuing the stem cell pool size in
MDS mice. The improved stem cell pool, reduced myeloid bias and attenuated anemia
resulted in a significant survival increment in iron-restricted compared to control
MDS mice.
Summary/Conclusion: Our results show for the first time in a preclinical mouse model
of MDS that iron excess driven by ineffective erythropoiesis has pathological implication
in transfusion-independent MDS. These effects are likely aggravated by transfusions
causing additional iron overload. Finally, iron restriction achieved through pharmacologic
FPN inhibition significantly improves MDS pathophysiology, uncovering the therapeutic
potential of early prevention of NTBI formation in MDS.
S276: TRANSFERRIN RECEPTOR 2 TARGETING AMELIORATES DIFFERENT FORMS OF ANEMIA CAUSED
BY CHRONIC INFLAMMATION
V. Olivari1 2,*, M. R. Lidonnici3, E. Tanzi1 4, S. M. Di Modica1, F. Tiboni3, L. Silvestri1
2, G. Ferrari2 3, A. Nai1 2
1Regulation of Iron Metabolism Unit - Division of Genetics and Cell Biology, IRCCS
San Raffaele Scientific Institute; 2Vita-Salute San Raffaele University; 3Gene Transfer
into Stem Cell Unit, SR-Tiget, IRCCS San Raffaele Scientific Institute; 4University
of Milan Bicocca, Milan, Italy
Background: Anemia is a common complication of chronic inflammation. It is caused
by an increased production of the iron-regulatory hormone hepcidin, that leads to
iron restriction, and by decreased erythrocytes sensitivity to erythropoietin (EPO)
stimulation. Anemia of Chronic Kidney Disease (CKD) is a form of anemia of inflammation,
in which progressive renal damage, besides causing systemic inflammation and iron
deficiency, impairs erythropoietin (EPO) production. Current EPO-based treatments
may cause off-target effects on long term and often require iron supplementation,
mainly for CKD.
Transferrin Receptor 2 (TFR2) modulates iron homeostasis in the liver, contributing
to the transcriptional activation of hepcidin, and acts as a brake of EPO signaling
in erythroid cells. Its selective deletion in the bone marrow (BM, Tfr2
BMKO
) enhances erythropoiesis both in wild-type and thalassemic mice.
Aims: We investigated whether Tfr2 targeting might represent a therapeutic option
for different forms of anemia, able to enhance erythropoiesis in chronic inflammation
and simultaneously increase iron availability in CKD.
Methods: Chronic sterile inflammation was induced by weekly subcutaneous injection
of turpentine oil (100ul/20g body weight for 3 weeks) in Tfr2
BMKO
mice. Mice were sacrificed 2 or 14 days after the last injection.
CKD was induced feeding Tfr2
BMKO
mice an adenine-rich diet for 8 weeks. The same protocol was applied also in germline
Tfr2 deficient (Tfr2-ko) animals to simultaneously inactivate erythroid and hepatic
Tfr2.
Complete blood count was periodically determined, and a complete phenotypic analysis
was performed at sacrifice.
Results: Chronically inflamed Tfr2
BMKO
mice maintained red blood cell count and hemoglobin levels higher than controls for
the entire 5-week-long timespan, despite comparable inflammation.
In CKD mice, BM-specific Tfr2 inactivation enhanced erythropoiesis and delayed anemia
until iron availability was sufficient: indeed, hemoglobin came back to control values
at the end of the protocol. The drop in hemoglobin levels was prevented in Tfr2-ko
mice, in which iron supply is increased. Renal damage was comparable among the experimental
groups, excluding a differential effect of the diet.
Of note, BM-specific Tfr2 deletion boosted erythropoiesis and activated EPO-EPOR pathway
without affecting EPO levels both in CKD and turpentine mice.
Summary/Conclusion: We proved that erythroid Tfr2 targeting ameliorates hematological
parameters in murine models of anemia of inflammation and anemia of CKD. The beneficial
effect is achieved enhancing erythroid EPO responsiveness, without affecting EPO levels
per se. For this reason, an approach based on erythroid TFR2 targeting would avoid
the side effects of the available therapies.
Moreover, we demonstrated that the concomitant inhibition of hepatic and erythroid
Tfr2 in anemia of CKD would represent a therapeutic opportunity to simultaneously
increase iron availability and erythroid responsiveness in a balanced manner.
Overall, our results prove that TFR2 targeting may represent a tunable therapeutic
approach for different forms of anemia.
S277: HYPOFERREMIA CAUSED BY FERROPORTIN DOWNREGULATION IS REGULATED BY NFКB THROUGH
HDAC-DEPENDENT MECHANISMS
O. Marques1,*, N. Horvat1, M. C. Lai2, L. Zechner1, S. Colucci1, R. Sparla1, S. Zimmermann3,
C. J. Neufeldt4, S. Altamura1, J. Zaugg2, K. Mudder1, J. D. Knopf5, M. K. Lemberg5,
M. Pasparakis6, M. W. Hentze2, M. U. Muckenthaler1
1Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Universitatsklinikum
Heidelberg; 2European Molecular Biology Laboratory; 3Department of Medical Microbiology
and Hygiene; 4Department of Molecular Virology, Universitatsklinikum Heidelberg; 5ZMBH,
University of Heidelberg; 6CECAD, University of Cologne, Heidelberg, Germany
Background: Anemia of Inflammation (AI) is a highly prevalent comorbidity in patients
with chronic inflammatory disorders. The underlying pathophysiology is partly explained
by hypoferremia that results from iron sequestration in macrophages and the reduction
of dietary iron uptake due to decreased iron export mediated by Ferroportin (Fpn).
In response to inflammation, Fpn levels are decreased by hepcidin-mediated post-translational
and/or transcriptional mechanisms, depending on the type of inflammatory pathway activated.
Despite the fact that Fpn transcriptional repression is frequently observed in inflammatory
conditions, the regulatory mechanisms involved remained unidentified.
Aims: To gain mechanistic insight how Fpn mRNA levels are regulated in response to
inflammation.
Methods: We conducted a pharmacological and RNAi screen, targeting mediators of TLR2/6
signalling, in bone marrow-derived macrophages (BMDMs) stimulated with the TLR2/6-specific
ligand FSL1. Changes in chromatin accessibility were evaluated by chromatin-accessibility
Real-Time Polymerase Chain Reaction (CHART-PCR) and Assay for Transposase-Accessible
Chromatin using sequencing (ATACseq). Data obtained in cell-based assays were validated
in mice injected with FSL1.
Results: The screen in BMDMs together with validation experiments demonstrated that
the FSL1 induced TLR2 response represses Fpn mRNA levels via MYD88 and TRAF6 signaling
transducers. While FSL1 can activate independent JNK, p38 and NFκB signaling branches,
we show that Fpn transcriptional repression is specifically dependent on alternative
NFκB signaling involving the transcription factor RELB. This contrasts our findings
for TLR4 stimulation by LPS, where reduced Fpn mRNA expression depends on classical
NFκB signaling, involving the NFкB subunits RELA and p50.
Using ATACseq we identified Fpn genomic regions that are altered by inflammatory stimuli.
These data show chromatin condensation around the Fpn transcription start site (TSS)
upon FSL1 or LPS treatment. Since chromatin condensation is mainly promoted by histone
deacetylases (HDACs), we next tested whether Fpn mRNA reduction upon inflammatory
cues could be counteracted by treatment with HDAC inhibitors (HDACi). We demonstrate
that pre-treatment with the pan-HDACi SAHA (Suberoylanilide Hydroxamic Acid) prevented
chromatin remodelling at the Fpn locus and Fpn transcriptional repression in response
to inflammation and RELB overexpression, suggesting that the effect of HDACs on Fpn
responses occurs downstream of NFкB. In line with these data, NFκB pharmacological
inhibition also prevented Fpn chromatin remodeling in the vicinity of the TSS. The
findings obtained in BMDMs were extended in mice treated with SAHA prior to FSL1 injection,
showing reduced hypoferremia and attenuation of splenic Fpn mRNA repression, while
serum hepcidin levels remained unaltered.
Summary/Conclusion: These results identify NFκB as an important repressor of Fpn transcription
in response to inflammatory signals involving chromatin remodeling in a genomic region
near Fpn’s TSS. Importantly, HDAC inhibition attenuates Fpn mRNA repression as well
as the resulting hypoferremia in mice. Our findings generate molecular insight about
how hypoferremia is generated and demonstrate that Fpn, like hepcidin, is controlled
by HDACs.
S278: EVALUATION OF THE MAIN REGULATORS OF SYSTEMIC IRON HOMEOSTASIS IN PYRUVATE KINASE
DEFICIENCY
A. Zaninoni1, R. Russo2 3, R. Marra2 3, I. Andolfo2 3, E. Fermo1, A. P. Marcello1,
D. Consonni4, B. Eleni Rosato2 3, S. Martone2 3, B. Fattizzo1, W. Barcellini1, A.
Iolascon2 3, P. Bianchi1,*
1Hematology Unit - Physiopathology of Anemias Unit, Foundation IRCCS Cà Granda Policlinico
Milan, Milan; 2Department of Molecular Medicine and Medical Biotechnology; 3CEINGE
– Advanced Biotechnologies, University Federico II, Naples; 4Epidemiology Unit, Foundation
IRCCS Cà Granda Policlinico Milan, Milan, Italy
Background: Iron loading anemias are characterized by ineffective erythropoiesis and
iron overload. This group of anemias includes thalassemia syndromes, congenital dyserythropoietic
anemias (CDA), and some forms of congenital hemolytic anemias. Among them pyruvate
kinase deficiency (PKD) has been shown to develop iron overload also in absence of
transfusions suggesting dyserythropoietic features. Moreover, severe forms can be
misdiagnosed as CDA due to bone marrow abnormalities and ineffective erythropoiesis
further supporting these evidences. The hormone erythroferrone (hERFE) is produced
by erythroblasts in response to erythropoietin (EPO), and acts by suppressing hepcidin,
thereby increasing iron absorption and mobilisation for erythropoiesis demand. The
ERFE-hepcidin axis seems to play a crucial role in the pathogenesis of these disorders;
an increased erythroferrone release by immature erythroid cells results in hepcidin
suppression and secondary iron overload that could finally results in ineffective
erythropoiesis and anemia.
Aims: To investigate the pathophysiological basis of iron overload in PKD.
Methods: We analysed the levels of hERFE, EPO, hepcidin, and soluble transferrin receptor
(sTFR) in a large group of 41 PKD patients equally distributed by gender, age and
severity. The results were analysed in comparison with two groups of patients affected
by hemolytic anemia with overt dyserythropoiesis (42 patients with CDA type II) and
with congenital hemolytic anemia due to RBC membrane defects (51 patients with hereditary
spherocytosis [HS]), respectively.
Results: Demographic, hematologic, and biochemical features of the three groups of
patients are reported in the Figure 1A. Among the PKD patients, 18/41 were <18 yrs,
median Hb level at the time of the study was 9.05g/dL (range 5.5-14.5), 12 underwent
splenectomy, 28 ever received at least three transfusions their life, 14 of them transfusion
dependent (>6 tx/yrs). Mean ferritin levels at the time of the study were 546 ng/ml
(range 59-4990), 15/41 patients requiring chelation therapy for iron overload developed
also in absence of transfusions.
As expected, CDAII patients showed decreased hepcidin levels (3.74 ng/mL; n.v. 17.25,
P<0.001) associated with increased erythropoietin (62.7 IU/L, n.v. 6.5, P=0.01) and
hERFE (24.8 ng/mL, n.v. 1, P<0.0001). On the contrary, HS showed increased hepcidin,
with less marked increased of ERFE (9.9 ng/mL, P=0.02) and EPO (36.4 IU/L, P=0.005).
In PKD patients we observed decreased hepcidin levels (7.15 ng/mL, P=0.03)), increased
hERFE (18 ng/mL, P<0.0001) and EPO (75.6 IU/L, P=0.009). Instead, sTFR was equally
increased in the three groups of patients (Figure 1B).
Interestingly, by comparing the three groups of patients, PKD showed dyserythropoietic
features as evidenced by the observation of intermediate values between HS and CDAII
of hepcidin (P=0.007 PKD v CDAII and P=0.0002 PKD vs HS), hEFRE, and sTFR (Figure
1C).
Image:
Summary/Conclusion: This study provides the first analysis of the main regulators
of systemic iron homeostasis in PK deficiency compared either with the model of a
structural RBC defect (HS) or with the typical model of dyserythropoietic anemia with
ineffective erythropoiesis, such as CDAII. These data provide evidence of the dyserythropoietic
features of PK deficiency, underlining the need of accurate diagnosis and paving the
way of novel therapeutic approaches in PK deficiency.
S279: INCREASED RISK OF INFECTIONS IN INDIVIDUALS WITH HEMOCHROMATOSIS C282Y HOMOZYGOSITY
AND IN INDIVIDUALS WITH HIGH OR LOW PLASMA IRON OR TRANSFERRIN SATURATION: A PROSPECTIVE
STUDY OF 138252 INDIVIDUALS
M. Mottelson1 2 3,*, A. Glenthøj1 2, B. Nordestgaard2 3 4 5, C. Ellervik4 6 7 8, S.
Bojesen2 3 4 5, J. Helby1 3
1Department of Hematology, Copenhagen University Hospital - Rigshospitalet; 2Department
of Clinical Medicine, University of Copenhagen; 3The Copenhagen General Population
Study; 4Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev
and Gentofte; 5The Copenhagen City Heart Study, Copenhagen University Hospital – Bispebjerg
and Frederiksberg, Copenhagen; 6Department of Production, Researh, and Innovation,
Region Zealand, Sorø, Denmark; 7Department of Laboratory Medicine, Boston Children’s
Hospital; 8Department of Pathology, Harvard Medical School, Boston, MA, United States
of America
Background: Plasma iron and transferrin saturation are measures of iron available
in the blood stream. Plasma ferritin indicate body iron stores. Levels of plasma iron,
transferrin saturation and ferritin are increased in hemochromatosis C282Y homozygous
individuals.
Aims: We tested the hypothesis that high and low plasma iron and transferrin saturation
are associated with increased risk of infections observationally and genetically through
hemochromatosis C282Y homozygosity.
Methods: We studied 138,252 general population individuals. Plasma iron and transferrin
saturation were measured in 136,656 and 136,599 individuals, respectively. 132,542
individuals were genotyped for the C282Y hemochromatosis variant. Individuals were
followed prospectively for up to 28 years for hospital admissions and emergency room
visits due to infectious diseases using the National Patient Register which covers
all danish hospitals.
Results: During follow-up 31,199 individuals were hospitalized due to an infection.
After multivariable adjustment including other risk factors for infections, individuals
with the 5% lowest and 5% highest plasma iron levels had hazard ratios for any infection
of 1.20 (95% CI:1.12-1.28;P<0.001) and 1.14 (95% CI:1.07-1.22;P<0.001) respectively,
when compared to individuals with plasma iron levels between the 25th-75th percentile.
Findings for transferrin saturation were similar. Hazard ratio for any infection in
hemochromatosis C282Y homozygous individuals was 1.40 (95% CI:1.16-1.69; P<0.001)
when compared to wildtype individuals. Risk of sepsis was especially high in C282Y
homozygotes (hazard ratio 1.69;95% CI:1.05-2.73; P=0.03). Risk of any infection was
increased even in C282Y homozygotes with normal levels of plasma iron, transferrin
saturation or ferritin.
Summary/Conclusion: High and low plasma iron and transferrin saturation were associated
with increased risk of infections. Hemochromatosis C282Y homozygotes had increased
risk of infection even when they had normal levels of plasma iron, transferrin saturation
or ferritin.
Current hemochromatosis treatment strategy centralized around ferritin normalization
may not reduce risk of infection satisfactorily in patients with hereditary hemochromatosis
genotype.
S280: B-CELL MALIGNANCIES TREATED WITH TARGETED DRUGS AND SARS-COV-2 INFECTION. A
EUROPEAN HEMATOLOGY ASSOCIATION SURVEY (EPICOVIDEHA)
M. S. Infante1, J. Salmanton-García2,*, A. Fernandez-Cruz3, F. Marchesi4, Z. RÁČIL5,
M. Hanakova6, O. Jaksic7, B. Weinbergerova8, C. Besson9, R. Duarte10, F. Itri11, T.
VALKOVIĆ12, A. Busca13, A. Guidetti14, A. GLENTHØJ15, G. Collins16, V. Bonuomo17,
U. Sili18, G. Seval19, M. Machado20, A. Lopez-Garcia21, R. Cordoba22, O. Blennow23,
G. Abu-zeinah24, S. Lemure25, A. Kulasekararaj26, I. Falces-Romero27, C. Cattaneo28,
O. A. Cornely29, J.-A. Hernandez-Rivas30, L. Pagano31
1Hematology Department, University Hospital Infanta Leonor, MADRID, Spain; 2Internal
Medicine, University of Cologne, Excellence center for Medical Mycology, Cologne,
Germany; 3Internal Medicine, Hospital Universitario Puerta de Hierro, Majadahonda,
Spain; 4hematology and stem cell transplant Unit, IRCCS Regina Elena National Cancer
Institute, Rome, Italy; 5Institute of Hematology and Blood transfusion, Prague, Czechia;
6Hematology Department, Institute of Hematology and Blood transfusion, Prague, Czechia;
7Hematology Department, University Hospital Dubrava, Zagreb, Croatia; 8Internal Medicine,
Masaryk University and University Hospital Brno, Brno, Czechia; 9Hematology Department,
Centre Hospitalier de Versailles, Versailles, France; 10hematology and stem cell transplant
Unit, Hospital Universitario Puerta de Hierro, MADRID, Spain; 11Hematology Department,
San Luigi Gonzaga Hospital, Orbassano, Italy; 12Croatian Cooperative Group for Hematological
Diseases (CROHEM), University Hospital Centre, Rijeka, Croatia; 13Stem Cell Transplant
Center, AOU Cittá della Salute e della Scienza, Turin; 14Hematology Department, Fondazione
IRCCS Istituto Nazionale, Milan, Italy; 15Hematology Department, Rigshospitalet University
Hospital, Copenhagen, Denmark; 16Hematology Department, NIHR Oxford Biomedical Research
Centre, Oxford, United Kingdom; 17Hematology Department, University Hospital of Verona,
Verona, Italy; 18Hematology Department, Marmara University Hospital, Istanbul; 19Hematology
Department, Ankara University Hospital, Ankara, Turkey; 20Clinical Microbiology and
Infectious diseases, Hospital Universitario Gregorio Marañon; 21Hematology Department,
Fundación Jimenez Diaz University Hospital; 22Hematology, Fundacíon Jimenez Diaz,
Madrid, Spain; 23Infectious diseases, Karolinska University Hospital, Stockolm, Sweden;
24Hematology Department, Weil Cornell Medicine, New York, United States of America;
25Hematology Department, CHU de Montpellier, Montpellier, France; 26Hematology Department,
King’s College Hospital, London, United Kingdom; 27Hematology, University Hospital
La Paz, MADRID, Spain; 28Hematology, ASST-Ospedali Civili, Brescia, Italy; 29Internal
Medicine, University of Cologne, Cologne, Germany; 30Hematology, University Hospital
Infanta Leonor, Madrid, Spain; 31Hematology, Fondazione Policlinico Universitario
Agostino Gemelli IRCCS, Roma, Italy
Background: Patients with lymphoproliferative diseases (LPD) appear particularly vulnerable
to SARS-CoV-2 infection, partly because of the effects of the anti-neoplastic regimens
(chemotherapy, signaling pathway inhibitors, and monoclonal antibodies) on the immune
system. The real impact of COVID-19 on the life expectancy of patients with different
subtypes of lymphoma and targeted treatment is still unknown.
Aims: The aim of this study is to describe and analyse the outcome of COVID-19 patients
with underlying LPD treated with targeted drugs such as monoclonal antibodies (obinutuzumab,
ofatumumab, brentuximab, nivolumab or pembrolizumab), BTK inhibitors (ibrutinib, acalabrutinib),
PI3K inhibitors (idelalisib), BCL2 inhibitors (venetoclax) and IMIDs, (lenalidomide).
Methods: The survey was supported by EPICOVIDEHA registry. Adult patients with baseline
CLL or non-Hodgkin Lymphoma (NHL) treated with targeted drugs and laboratory-confirmed
COVID-19 diagnosed between January 2020 and January 2022 were selected.
Results: The study included 368 patients (CLL n=205, 55.7%; NHL n=163, 44.3%) treated
with targeted drugs (Table 1). Median follow-up was 70.5 days (range 19-159). Most
used targeted drugs were ITKs (51.1%), anti-CD20 other than rituximab (16%), BCL2
inhibitors (7.3%) and lenalidomide (7.9%). Of note, only 16.0% of the patients were
vaccinated with 2 or more doses of vaccine at the onset of COVID-19.
Pulmonary symptoms were present at diagnosis in 244 patients (66.2%). Severe COVID-19
was observed in 47.8 % patients while 21.7% were admitted to to intensive care unit
(ICU), being 55 (26.8%) CLL patients and 25 (15.3%) NHL patients. More comorbidities
were reported in patients with severe-critical COVID-19 compared to those with mild-
asymptomatic infection (p=0.002). This difference was relevant in patients with chronic
heart diseases (p=0.005). Overall, 134 patients (36.4%) died. Primary cause of death
was COVID-19 in 92 patients (68.7%), LPD in 14 patients (10.4%), and a combination
of both in 28 patients (20.9%).Mortality was 24.2% (89/368) at day 30 and 34.5%(127/368)
at day 200. After a Cox multivariable regression age >75 years (p<0.001, HR 1.030),
active malignancy (p=0.011, HR 1.574) and admission to ICU (p<0.00, HR 4.624) were
observed as risk factors. Survival in patients admitted to ICU was 33.7% (LLC 38.1%,
NHL 24%). Mortality rate decreased depending on vaccination status, being 34.2% in
not vaccinated patients, 15.9-18% with one or two doses, decreasing to 9.7% in patients
with booster dose (p<0.001). There was no difference in OS in NLH vs CLL patients
(p=0.344), nor in ITKs vs no ITKs treated patients (p=0.987). Additionally, mortality
rate dropped from the first semester 2020 (41.3%) to last semester 2021 (25%).
Image:
Summary/Conclusion: - Our results confirm that patients with B-malignancies treated
with targeted drugs have a high risk of severe infection (47.8%) and mortality (36.4%)
from COVID-19.
- Presence of comorbidities, especially heart disease, is a risk factor for severe
COVID-19 infection in our series.
- Age >75 years, active malignancy at COVID-19 onset and ICU admission were mortality
risk factors.
- COVID-19 vaccination was a protective factor for mortality, even in this population
with humoral immunity impairment.
- The learning curve in the management of the infection throughout the pandemic and
the development of COVID-19 treatments showed benefit in this particularly vulnerable
population.
S281: PRIOR EXPOSURE TO IMMUNOSUPPRESSIVE AGENTS AND COMORBIDITIES ARE ASSOCIATED
WITH WORSE OUTCOMES OF SARS-COV2 INFECTION IN PH-NEG CHRONIC MYELOPROLIFERATIVE NEOPLASMS:
RESULTS OF EPICOVIDEHA SURVEY
M. Marchetti1,*, J. Salmanton-García2, S. El-Ashwah3, M. V. Sacchi1, F. Marchesi4,
Z. Ráčil5, M. Hanakova6, G. Zambrotta7, L. Verga7, F. Passamonti8, F. Itri9, J. Van
Doesum10, S. Martìn-Pérez11, A. López-García12, J. Dávila-Valls11, R. Cordoba13, G.
Abu-Zeinah14, G. Dragonetti15, C. Cattaneo16, V. Bonuomo17, L. Prezioso18, A. Glenthøj19,
F. Farina20, R. Duarte21, O. Blennow22, O Cornely23, L. Pagano24
1Hematology & TMO Unit, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria,
Italy; 2Department I of Internal Medicine, Excellence Center for Medical Mycology
(ECMM), University of Cologne and University Hospital, Faculty of Medicine, Cologne,
Germany; 3Oncology Center, Mansoura University, Mansoura, Egypt; 4Hematology and Stem
Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy; 5Institute
of Hematology and Blood Transfusion, Prague and Brno Hospital; 6Institute of Hematology
and Blood Transfusion, Prague Hospital, Prague, Czechia; 7Hematology Unit, Azienda
Ospedaliera San Gerardo; University Bicocca-Milan, Monza; 8Division of Hematology,
Department of Medicine and Surgery, University of Insubria, Varese; 9Hematology Unit,
Hospital San Luigi Gonzaga, Orbassano, Italy; 10University Medical Center Groningen,
University of Groningen, Groningen, Netherlands; 11Hospital Nuestra Señora de Sonsoles,
Ávila; 12Health Research Institute IIS-FJD; 13Fundacion Jimenez Diaz University Hospital,
Madrid, Spain; 14Division of Hematology and Oncology, Weill Cornell Medicine, New
York, United States of America; 15Hematology Unit, Fondazione Policlinico Universitario
Agostino Gemelli - IRCCS, Rome; 16Hematology Unit, ASST-Spedali Civili, Brescia; 17Department
of Medicine, Section of Hematology, Azienda Ospedaliera Universitaria Integrata, Verona;
18Hematology and Bone Marrow Unit, University Hospital, Parma, Italy; 19Department
of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark;
20Hematology & TMO unit, IRCCS Ospedale San Raffaele, Milan, Italy; 21Hospital Universitario
Puerta de Hierro, Majadahonda, Madrid, Spain; 22Department of Infectious Diseases,
Karolinska University Hospital, Stockholm, Sweden; 23Dpt I of Internal Medicine, ECMM,
CECAD, Clinical Trials Centre Cologne (ZKS Köln), Center for Molecular Medicine Cologne
(CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne,
German Centre for Infection Research (DZIF), Cologne, Germany; 24Hematology Unit,
Fondazione Policlinico Universitario Agostino Gemelli - IRCCS, Università Cattolica
del Sacro Cuore, Rome, Italy
Background: Philadelphia-negative chronic myeloproliferative neoplasms (MPN) typically
incur high rates of thrombosis and infections and cytoreductive drugs may modulate
such risks.
Aims: The present analysis aims at assessing the severity and outcomes of MPN facing
coronavirus disease 2019 (COVID-19). Hence, we aimed to assess the impact of immunosuppressive
agents and comorbidity burden in COVID-19 outcome.
Methods: The EPICOVIDEHA registry is an online survey (www.clinicalsurveys.net) that
has collected since April 2020 until January 2022 5,445 cases of COVID-19 in individuals
with baseline haematological malignancies (Salmanton-García et al, 2021 Hemasphere)
The survey is promoted by the European Hematology Association - Infectious Diseases
Working Party (EHA-IDWP) and has been approved centrally by the Institutional Review
Board and Ethics Committee of Fondazione Policlinico Universitario A. Gemelli – IRCCS
– Università Cattolica del Sacro Cuore, Rome, Italy (Study ID: 3226).
Results: Overall, 308 patients (5.6%) with MPN were observed for a median of 102 days
(IQR: 21-223, range 22-97) after COVID-19 diagnosis. Median age at infection was 69
years (IQR: 58-77, range 22-97) and at least one comorbidity was reported from most
of the individuals (62.6%, n = 193). A large portion of patients had a history of
cardiopathy (n=109, 35.4%), diabetes (n=40, 15.9%), or chronic pulmonary disease (n=44,
14.3%). Myelofibrosis (MF) (n=140, 45.4%) was the most prevalent baseline malignancy,
with 18 MF patients (12.9%) reporting 3 or more comorbidities. Out of the whole cohort,
72 patients (42.8% of MF) received immunosuppressige therapies including steroids,
immunomodulatory drugs (IMiDs) or JAK-inhibitors.
Hospitalization and consecutive admission to intensive care unit was required for
187 (60.7%) and 45 (24%) patients, respectively. At multivariate logistic regression,
hospital admission was predicted by age ≥70 years (OR 2.809; 95% CI 1.651-4.779),
exposure to immunosuppressive therapies (OR 2.802; 95% CI 1.5380-5.103) and comorbidity
burden.
During the study follow-up (median 101 days; range 21-222) 84 patients deceased after
a median time of 14 days (IQR: 8-49, range 0-457) since COVID-19 diagnosis. The fatality
rate (FR) decreased from 40.3% (50 out of 124) in the first two quarters of year 2020
to 15.8% (3 out of 19) in the first two quarters of year 2021 (p<0.05). Death was
principally attributable to COVID-19 in 58 patients (69.0%) and contributable by COVID-19
in 15 (17.9%). FR was particularly high (54 out of 140, 38.6%) in MF patients and
in patients receiving immunosuppressive agents (32 out of 86, 37%). Moreover, FR increased
from 13.0% in individuals with no comorbidity to 36.0% and 62.1% in those with ≥2
or ≥3 comorbidities, respectively. More specifically, three comorbidities independently
increased the FR: chronic cardiopathy (HR 1.653; 95%CI 1.017-2.687), chronic pulmonary
disease (HR 1.847; 95% CI 1.097-3.109), and diabetes mellitus (HR 1.712; 95% CI 1.006-2.914).
A heavy comorbidity burden, namely 3 or more comorbidities (HR 2.956; 95% CI 1.403-6.227),
advanced age, namely ≥70 years (HR .809; 95% CI 1.651-4.779), myelofibrosis (HR 2.501;
95% CI 1.384-4.519), and ICU admission (HR 2.669; 95% CI 1.641-4.342) independently
predicted FR.
Image:
Summary/Conclusion: COVID-19 infection led to a particularly dismal outcome in patients
exposed to immunosuppressive agents and in those with chronic heart or pulmonary diseases,
or diabetes. These data allow to tailor future strategies for preventing severe COVID-19
in MPN patients.
S282: A RANDOMIZED CONTROLLED CLINICAL TRIAL DEMONSTRATES THAT PLASMA FROM CONVALESCENT
AND VACCINATED DONORS IMPROVES OUTCOME OF COVID-19 IN PATIENTS WITH HEMATOLOGICAL
DISEASE, CANCER OR IMMUNOSUPPRESSION
C. Müller-Tidow1,*, M. Janssen1, U. Schäkel1, J. Gall2 3, A. Leo4, P. Stelmach1, J.
Krisam5, L. Baumann5, J. Stermann5, U. Merle6, M. Zeier7, M. A. Weigand8, L. Bullinger9,
J.-F. Schrezenmeier9, M. Bornhäuser10, N. Alakel10, O. Witzke11, T. Wolf12, M. J.
Vehreschild12, S. Schmiedel13, M. M. Addo13, F. Herth14, M Kreutner15, P.-R. Tepasse16,
B. Hertenstein17, M. Hänel18, A. Morgner18, M. Kiehl19, O. Hopfer19, M.-A. Wattad20,
C. C. Schimanski21, C Celik21, T. Pohle22, M. Ruhe22, W. V. Kern23, A Schmitt1, M.
Schmitt1, P. Dreger1, H.-M. Lorenz1, M. Souto-Carneiro1, N. Halama24, S. Meuer4, H.-G.
Kräusslich25, B. Müller25, R. Bartenschlager26, J. Klemmer1, K. Kriegsmann1, R. F.
Schlenk1 2 3, C. M. Denkinger27
1Department of Internal Medicine V, Heidelberg University Hospital; 2NCT-Trial Center,
National Center of Tumor Diseases, Heidelberg University Hospital; 3German Cancer
Research Center; 4Institute for Clinical Transfusion Medicine and Cell Therapy Heidelberg;
5Institute for Medical Biometry and Informatics, Ruprecht-Karls University Heidelberg;
6Department of Internal Medicine IV, Heidelberg University Hospital,; 7Department
of Nephrology, University of Heidelberg, Heidelberg; 8Department of Anaesthesiology,
Heidelberg University Hospital, Heidelberg, Heidelberg; 9Department of Hematology,
Oncology and Tumor Immunology, Charité University Medicine, Campus Virchow Clinic,
Berlin; 10Department of Internal Medicine I, University Hospital and Faculty of Medicine
Carl Gustav Carus of TU Dresden, Dresden; 11Department of Infectious Diseases, West
German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen,
Essen; 12Department of Internal Medicine, Infectious Diseases, University Hospital
Frankfurt, Goethe University Frankfurt, Frankfurt am Main; 13I. Department of Medicine,
University Medical Center Hamburg-Eppendorf, Hamburg; 14Pneumology and Critical Care
Medicine, Thoraxklinik, University of Heidelberg; 15Center for Interstitial and Rare
Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research
(DZL), Heidelberg; 16Department of Medicine B, Gastroenterology and Hepatology, University
Hospital Münster, Münster; 17Medical Department I, Klinikum Bremen-Mitte, Bremen;
18Department of Internal Medicine III, Klinikum Chemnitz gGmbH, Chemnitz; 19Department
I of Internal Medicine, Frankfurt (Oder) General Hospital, Frankfurt (Oder); 20Department
of Hematology, Oncology, Palliative Care and Stem Cell Transplantation, Klinikum Hochsauerland
GmbH, Meschede; 21Department of Internal Medicine II, Klinikum Darmstadt GmbH, Darmstadt;
22Department of Internal Medicine I, Klinikum Herford, Herford; 23Department of Medicine
II, Division of Infectious Diseases and Travel Medicine, University Medical Centre
Freiburg, Freiburg; 24Department of Medical Oncology, National Center for Tumor Diseases,
Heidelberg University Hospital; 25Department of Infectious Diseases, Virology, Heidelberg
University Hospital; 26Department of Infectious Diseases, Molecular Virology, Heidelberg
University Hospital; 27Division of Tropical Medicine, Department of Infectious Diseases,
Heidelberg University Hospital, Heidelberg, Germany
Background: Therapy options are limited for COVID-19 patients with hematological disease,
cancer, immunosuppression or advanced age. Even though no benefit was observed for
convalescent plasma in unselected patients with COVID-19, retrospective data suggest
that it could be effective in patients unable to mount a sufficient immune response
upon SARS-CoV-2 infection. Plasma from vaccinated donors has not been systematically
assessed for COVID-19 treatment.
Aims: We conducted a randomized clinical trial to address plasma efficacy in patients
at high risk for an adverse outcome.
Methods: COVID-19 patients with confirmed SARS-CoV-2 infections and oxygen saturation
<=94% were randomized (NCT05200754). Patients received convalescent or vaccinated
SARS-CoV-2 plasma in two bags (238 - 337 ml plasma each) from different donors on
day 1 and 2 (PLASMA) or standard of care (CONTROL). Randomization was stratified according
to four clinical patient groups, hematological/solid cancer (group-1), treatment or
disease associated immunosuppression (group 2), high risk disease by standard parameters
(group-3) or age >=75 years (group-4). Mechanically ventilated patients were not eligible.
Plasma was obtained from donors with high level neutralizing activity (titer >=1:80)
either after SARS-CoV-2 infection (convalescent) or after vaccination with at least
two doses of mRNA vaccines (vaccinated). Crossover for the control group was allowed
at day 10. The primary endpoint was time to improvement as two points on a seven-point
ordinal scale or live discharge from the hospital (IMPROVEMENT) with prespecified
analyses of subgroups (Janssen M, et al. Trials 2020 Oct 6;21(1):828).
Results: A total of 133 patients were randomized with 68 receiving PLASMA with a median
age of 68 years (range 36-95) or CONTROL (n=65, of which n=10 (15.4%) crossed over
at day 10) with a median age of 70 years (range 38-90). The distribution of the four
predefined groups was group-1, n=53; group-2, n=18; group-3, n=35; and group-4, n=27.
The intention to treat analysis revealed a non-significant shorter time to IMPROVEMENT
for patients in PLASMA (median 12.5 days, 95%-CI [10; 16]) compared to patients in
CONTROL (median 18 days, 95%-CI [11; 28]), hazard ratio 1.24, 95% confidence interval
[0.83; 1.85], p=0.29). Overall, 27 patients died (PLASMA, n=12; CONTROL, n=15; p=0.80).
Predefined subgroup analysis revealed a clinically significant benefit in patients
with hematological malignancies, other cancers or immunosuppression (group-1, group-2,
n=71). With a median time to improvement of 13 days (95%-CI [9; 19]) for PLASMA and
32 days (95%-CI [17; 57]) for CONTROL(HR 2.03, 95%-CI [1.17; 3.6], p=0.01). A sensitivity
analysis revealed that IMPROVEMENT appeared to be seen even earlier with vaccinated
(median 10 days, 95%-CI [8; 14]) compared to convalescent SARS-CoV-2 plasma (median
13 days, 95%-CI [6; 38]) and CONTROL. Within group-1 and group-2, six patients in
PLASMA (18.2%) and 10 in CONTROL (28.6%) died. No significant differences in improvement
were observed in group-3 and group-4 with a HR of 0.72 (95%-CI [0.41; 1.28], p=0.26).
Within group-3 and group-4, six patients in PLASMA (18.8%) and five in CONTROL (16.7%)
died. No previously unknown side effects of plasma therapy emerged within the trial.
Image:
Summary/Conclusion: Plasma from convalescent and particularly vaccinated donors improved
outcome of COVID-19 patients with an underlying hematological disease /cancer or other
reasons of impaired immune response. Plasma did not improve outcome in immune-competent
patients with other risk factors and/or older age.
S283: ERN-EUROBLOODNET EUROPEAN REGISTRY OF PATIENTS AFFECTED BY RED BLOOD CELL DISORDERS
AND COVID-19
P. Velasco1,*, F. Longo2, A. Piolatto2, E. J. Bardón-Cancho3 4 5, B. Ponce-Salas6,
P. Flevari7, E. Voskaridou8, B. J. Biemond9, E Nur9 10, P. Delaporta11, T. Besse-Hammer12,
A. Ruiz-Llobet13, S. Raso14, A. Spasiano15, M. E. Guerzoni16, D. Beneitez-Pastor17
18 19 20, L. Dedeken21, A. Pepe22, R. Rosso23, J. B. Kunz24, M. de Montalembert25,
S. Campisi26, A. Glenthøj27, P. Gonzalez Urdiales28 29, F. S. Benghiat30, M.-A. Azerad31,
C. J. Saunders32, T. Ferreira Faria33, T. Casini34, S. Bagnato35, A. Van de Velde36,
V. Labarque37, E. Bertoni38, A. Van Damme39, M. D. Diamantidis40, R. Russo41 42, E.
Stiakaki43, A Quota44, S. Christou45, M. J. Teles46 47 48 49 50, I. Lafiatis51, J.-L.
Kerkhoffs52, M. Argüello Marina53, M. Lorite54, A. Rodriguez55 56, A. Iolascon41 42,
A. T. Taher57, R. Colombatti58, N. Roy59, M. D. M. Mañú Pereira60
1pediatric oncology and hematology, Hospital Vall d’Hebron, Barcelona, Spain; 2Department
of Clinical and Biological Sciences, University of Torino, Orbassano (To), Italy;
3Pediatric Hematology Unit, Hospital General Universitario Gregorio Marañón; 4Facultad
de Medicina, Universidad Complutense de Madrid; 5Instituto Investigación Sanitaria
Gregorio Marañón; 6Onco-Hematología Infantil, Hospital General Universitario Gregorio
Marañón, Madrid, Spain; 7Center of Excellence in Rare Haematological Diseases-Haemoglobinopathies,
Laiko General Hospital; 8Center of Excellence in Rare Haematological Diseases-Haemoglobinopathies,
Laiko General Hospital, Athens, Greece; 9Department of Hematology, Amsterdam University
Medical Centers; 10Department or Blood Cell Research, Sanquin Research, Amsterdam,
Netherlands; 11First Department of Pediatrics, National and Kapodistrian University
of Athens, ‘AghiaSophia’ Children’s Hospital, Athens, Greece; 12URC, CHU Brugmann,
Brussels, Belgium; 13Paediatric Haematology Department, Hospital Sant Joan de Déu,
Universitat de Barcelona, Barcelona, Spain; 14Department of Hematology and Rare Diseases,
V Cervello, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo; 15Malalttie
Rare del Globulo Rosso, AORN A. Cardarelli, Naples; 16Pediatric Unit,Department of
Medical and Surgical Sciences of Mothers, Pediatric Unit,Department of Medical and
Surgical Sciences of Mothers, Modena, Italy; 17Translational ResearchGroup in Rare
Anemia Disorders, Vall d’Hebron Institut de Recerca; 18Hematology Department, Vall
d’Hebron Hospital Universitari; 19Experimental Hematology Group, Vall d’Hebron Institute
of Oncology (VHIO); 20Vall d’Hebron Hospital Universitari, ERN-EuroBloodNet, Barcelona,
Spain; 21Hôpital Universitaire des Enfants Reine Fabiola (ULB), Brussels, Belgium;
22Fondazione Toscana G. Monasterio per la ricerca medica e di sanità pubblica, Pisa;
23Thalassemia and Heamoglobinophaties, Policlinico Universitario G. Rodolico, Catania,
Italy; 24University Hospital, Heidelberg, Germany; 25Hôpital Necker, Paris, France;
26Ospedale Umberto I, Siracusa, Italy; 27Department of Hematology, Rigshospitalet,
Copenhagen, Denmark; 28Pediatric oncology and hematology department, Hospital Universitario
Cruces; 29Instituto de investigaciónBiocruces Bizkaia, Barakaldo, Spain; 30CUB Erasmus
Hospital, Brussels; 31Hematology, CHU Liège Citadelle site, Liège, Belgium; 32Hospital
dos Capuchos, Centro Hospitalar e Universitário Lisboa Central, Lisbon; 33Pediatric
Department, Hospital Fernando Fonseca, Amadora, Portugal; 34Pediatric OncoHematology,
Meyer Children Hospital, Florence; 35Thalassemia, Ospedale Civile, Lentini, Italy;
36Hematology, University Hospital Antwerp, Edegem; 37Pediatric Hemato-Oncology, University
Hospitals Leuven, Leuven, Belgium; 38Oncohaematology and BMT Unit, ASST Spedali Civili
di Brescia, Brescia, Italy; 39Paediatric Hémato-oncology, Cliniques Universitaires
Saint-Luc, Brussels, Belgium; 40Thalassemia and Sickle Cell Disease Unit, Department
of Hematology, General Hospital of Larissa, Larissa, Greece; 41Department of Molecular
Medicine and Medical Biotechnology, University Federico II; 42CEINGE - Advanced Biotechnologies,
Naples, Italy; 43Pediatric Hematology-Oncology Department, Universityof Crete, University
Hospital of Heraklion, Heraklion, Greece; 44UOSD Thalassemia, V.Emanuele Hospital,
Gela, Italy; 45Thalassemia Clinic, Archbishop Makarios III Hospital, Nicosia, Cyprus;
46Clinical Pathology, Centro Hospitalar e Universitário de São João; 47Clinical Hematology,
Centro Hospitalar e Universitário do Porto; 48Anemia Working Group Portugal; 49Institute
for the HealthResearch and Innovation (i3S); 50Hematology Working Group of the Portuguese
Society of Clinical Pathology, Porto, Portugal; 51General Hospital of Mytilini, Mytilini,
Greece; 52Hematology, HagaZiekenhuis, The Hague, Netherlands; 53Hematology, Hospital
Príncipe de Asturias, Madrid; 54Department of pediatric Hematology, Hospital Son Espases,
Palma de Mallorca; 55Clinical Pharmacology Service, Hospital Universitari Vall d’Hebron;
56Vall d’Hebron Institut de Recerca, Barcelona, Spain; 57Division of Hematology and
Oncology, Department of Internal Medicine, American University of Beirut Medical Center,
Beirut, Lebanon; 58UOC Pediatric Hematology Oncology, University of Padova, Padova,
Italy; 59Department of Haematology, Oxford University Hospitals NHS Foundation Trust,
Oxford, United Kingdom; 60Translational Research in Child and Adolescent Cancer, Vall
d’Hebron Institut de Recerca, Barcelona, Spain
Background: Patients with red blood cell disorders (RBCD), are likely to be at increased
risk of complications from SARS-Cov-2 (Covid-19), but evidence in this population
is scarce due to its low frequency and heterogeneous distribution.
Aims: ERN-EuroBloodNet, the European Reference Network in rare hematological disorders,
established a European registry to determine the impact of COVID-19 on RBCD patients
and identify risk factors predicting severe outcomes.
Methods: The ERN-EuroBloodNet registry was established in March 2020 by VHIR based
on Redcap software in accordance with the Regulation (EU) 2016/679 on personal data.
The local Research Ethics Committee confirmed that the exceptional case of the pandemic
justifies the waiver of informed consent.
Eligible patients had confirmed RBCD and COVID-19. Data collected included demographics,
diagnosis, comorbidities, treatments, and COVID-19 symptoms and management.
For analysis of COVID-19 severity, two groups were established 1) Mild: asymptomatic
or mild symptoms without clinical pneumonia and 2) Severe: pneumonia requiring oxygen/respiratory
support and/or admission to intensive care unit. Continuous variables were compared
using the Wilcoxon rank-sum test or Kruskall Wallis test, while categorical variables
were analyzed using the Chi-square test or Fisher’s Exact test. Relevant factors influencing
disease or severity were examined by the logistic regression adjusted for age.
Results: As of February 25, 2022, 42 medical centers from 10 EU countries had registered
428 patients: 212 Sickle cell disease (SCD), 186 Thalassemia major and intermedia
(THAL).
The mean age of SCD was lower (22y) than of THAL (39.4y). Splenectomy and comorbidities
were higher in THAL (51.4% and 61,3%) than in SCD (16,3% and 46,8%) (p<0.001, p=0.004).
Age and BMI correlated with COVID-19 severity, as described in the general population
(p=0.003, p<0.001). Fig 1 shows age distribution and COVID-19 severity by disease
severity groups. The mean age for severe COVID-19 was lower in patients with severe
SCD (SS/SB0 vs SC/SB+: 23y vs 67.5y) and THAL (major vs intermedia: 43.5 vs 51.3y)
(p<0.001).
Potential risk factors such as elevated ferritin, current chelation or history of
splenectomy did not confer additional risk for developing severe COVID-19 in any patient
group. Only diabetes as a comorbidity correlated with severity grade in SCD (p=0.01)
and hypertension in THAL (p=0.009). While severe COVID-19 infection in SCD was associated
with both ACS (p<0.001) and kidney failure requiring treatment (p<0.001), this was
not predicted by a history of previous ACS or kidney disease in steady state.
Overall, 14,6% RBC patients needed oxygen/respiratory support, 4% were admitted to
ICU with an overall mortality rate of 1%, much lower than reported in other similar
cohorts.
Image:
Summary/Conclusion: Results obtained so far show that severe COVID-19 occurs at younger
ages in more aggressive forms of SCD and THAL. Current preventive approaches focus
on age over disease severity. Our data highlights the risk of severe COVID-19 infection
in some young patients, particularly those with SS/SB0 SCD, suggesting that immunization
should be considered in this pediatric group as well.
Results between similar sized cohorts of RBCD patients vary between each other and
those presented here, highlighting the importance of collecting all of these small
cohorts together to ensure adequate statistical power so that definitive risk factors
can be reliably identified and used to guide management of patients with these rare
disorders in the light of the ongoing pandemic.
S284: RISK FACTORS AND OUTCOMES OF PATIENTS WITH HAEMATOLOGICAL MALIGNANCY AND COVID-19:
ONGOING REDUCTION IN MORTALITY DURING THE 3RD WAVE OF THE PANDEMIC
J. Maddox1,*
1Haematology, South Tees NHS Trust, Middlesborough, United Kingdom
Background: It was established early in the COVID-19 pandemic that patients with cancer,
in particular haematological malignancy, had worse outcomes than non-cancer patients.
There is a lack of recent data to describe how mortality in patients with haematology
malignancy has changed with widespread vaccination and several effective treatments
now being available.
Aims: Based in a large NHS Trust in the North-East of England, our aim was to identify
all local patients with haematological malignancy who had contracted COVID-19 since
the start of the pandemic. We then examined these patients in more detail to ascertain
risk factors for mortality, and how this has changed over the course of the pandemic.
Methods: We included patients with an active diagnosis of haematological malignancy,
or those who had received potentially curative treatment within the past 3 years.
We excluded patients with pre-malignant conditions. Data up to the end of November
2021 found 213 eligible patients. Nearly all patients identified in the 1st wave of
the pandemic were identified from hospital testing, reflecting the lack of availability
of widespread testing in the community. More recently, patients identified in the
community have predominated.
Results: Overall mortality following COVID-19 infection was 21.6% at 4 weeks and 27.7%
at 8 weeks after COVID-19 diagnosis. Mortality was highest in wave 1 (March – June
2020), decreasing in wave 2 (Sept 20 – March 2021) and again in wave 3 (May 2021 –
current), with 4-week mortality figures of 44%, 26% and 8% respectively. It should
be noted that widespread community testing was not available early in the pandemic
so the recorded cases in wave 1 were sicker patients needing hospital care. Removing
pillar 2 data still shows a reduction in 4-week mortality over the course of the pandemic,
from 46% (wave 1) to 39% (wave 2) and 8% (wave 3).
Although official COVID-19 mortality figures only include deaths within 4 weeks of
a confirmed infection, we note that the Kaplan-Myer mortality curve did not level
out until 6-8 weeks after initial infection (figure 1). Overall mortality at 8 weeks
was therefore higher at 61% (wave 1), 31% (wave 2) and 11% (wave 3).
The main risk factor for mortality was patient age, with 8-week mortality in age groups
<60yr, 60-69yr, 70-79yr, 80-89yr and 90+yr being 2%, 26%, 30%, 51% and 86% respectively.
Although men comprised the majority of detected cases (62%), mortality at four and
eight weeks was near identical between sexes.
Although disease subgroups were relatively small, we found the highest COVID-related
mortality in patients with CLL (43%) and MDS (42%). It is perhaps not surprising as
these two disease groups also had the highest mean patient ages.
Patients receiving chemotherapy had no significant increase in 8-week mortality compared
to those not receiving chemotherapy, 30% vs. 27% (p=ns). The presence of neutropenia
was however a risk factor for mortality. Patients who were neutropenic at the time
of infection had an 8-week mortality of 43%, compared to 25% in those who were not
neutropenic (P=0.04).
Image:
Summary/Conclusion: Overall, COVID-19 related mortality in patients with haematological
malignancy has significantly declined over the course of the pandemic. The main risk
factor for death is increased patient age, with neutropenia also being a risk factor.
S285: INHIBITION OF COMPLEMENT C1S WITH SUTIMLIMAB IN PATIENTS WITH COLD AGGLUTININ
DISEASE (CAD): 2-YEAR FOLLOW-UP FROM THE CARDINAL STUDY
A. Röth1,*, W. Barcellini2, S. D’Sa3, Y. Miyakawa4, C. M. Broome5, M. Michel6, D.
J. Kuter7, B. Jilma8, T. H. A. Tvedt9, I. C. Weitz10, T. Sourdille11, J. Wang11, D.
S. Vagge12, K. Kralova13, F. Shafer14, M. Wardecki15, M. Lee14, S. Berentsen16
1Department of Hematology and Stem Cell Transplantation, West German Cancer Center,
University Hospital Essen, University of Duisburg-Essen, Essen, Germany; 2Fondazione
IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy; 3UCLH Centre for Waldenström’s
Macroglobulinemia and Related Conditions, University College London Hospitals NHS
Foundation Trust, London, United Kingdom; 4Thrombosis and Hemostasis Center, Saitama
Medical University Hospital, Saitama, Japan; 5Division of Hematology, MedStar Georgetown
University Hospital, Washington DC, United States of America; 6Henri-Mondor University
Hospital, Assistance Publique-Hôpitaux de Paris, UPEC, Créteil, France; 7Division
of Hematology, Massachusetts General Hospital, Harvard Medical School, Boston, MA,
United States of America; 8Department of Clinical Pharmacology, Medical University
of Vienna, Vienna, Austria; 9Section for Hematology, Department of Medicine, Haukeland
University Hospital, Bergen, Norway; 10Keck School of Medicine of USC, Los Angeles,
CA; 11Sanofi, Cambridge, MA, United States of America; 12IQVIA, Bangalore, India;
13Sanofi, Paris, France; 14Sanofi, Bridgewater, NJ, United States of America; 15Sanofi,
Warsaw, Poland; 16Department of Research and Innovation, Haugesund Hospital, Haugesund,
Norway
Background: CAD is a rare chronic autoimmune hemolytic anemia characterized by classical
complement pathway (CP)-mediated hemolysis. Sutimlimab is a first-in-class humanized
monoclonal antibody that selectively inhibits C1s of the C1 complex, preventing CP
activation, while leaving the alternative and lectin pathways intact. One-year interim
follow-up from the CARDINAL study (NCT03347396) have previously demonstrated that
sutimlimab resulted in sustained improvements in hemolytic markers and quality of
life.
Aims: To report 2-year sutimlimab efficacy and safety from the CARDINAL Part B extension.
Methods: CARDINAL was a Phase 3, open-label, single-arm study with a 26-week treatment
period (Part A) and a 2-year extension (Part B) after the last patient (pt) finishes
Part A. Sutimlimab was administered through intravenous infusions on Days 0 and 7,
followed by biweekly dosing. Efficacy data through Week 131, the last data recording
within the 2-year Part B period, are reported here. Efficacy endpoints included change
from baseline in hemolytic markers, pharmacodynamic (PD) markers and blood transfusions.
Quality of life (QOL) was assessed using the Functional Assessment of Chronic Illness
Therapy (FACIT)-Fatigue Scale. Safety was recorded until end-of-study visit 9 weeks
after their last dose; endpoints included incidence of treatment-emergent adverse
event (TEAE) and serious TEAE (TESAE). Descriptive statistics, frequency, and percentage
were used to analyze outcomes.
Results: Of the 24 pts enrolled in Part A, 22 completed Part A and entered Part B,
with 19 (86.4%) pts completing Part B. Sutimlimab treatment improved mean (SD) hemoglobin
(Hb) levels within one week; mean Hb remained >11 g/dL from Week 5–131 (baseline:
8.64 (1.67) (Figure). Mean total bilirubin was normalized from Week 3–131 (Figure).
Mean FACIT-Fatigue scores improved within 1 week and remained ≥5 from Week 1–123 (Figure),
consistent with a clinically meaningful change. Improvements in Hb, bilirubin, and
FACIT-Fatigue correlated with normalization of C4 and near-complete inhibition of
CP activity. Normalization of mean absolute reticulocyte count was observed alongside
normalized haptoglobin levels and reductions in LDH. From Week 26–131, 15 (68.2%)
pts remained transfusion-independent. All 22 pts experienced ≥1 TEAE; 12 (54.5%) pts
experienced ≥1 TESAE. Serious infections were reported in 7 (31.8%) pts, including
one pt with sepsis due to streptococcus pneumoniae. No meningococcal infections were
reported. Three pts discontinued the study due to AEs (cyanosis and klebsiella pneumoniae
(n=1); vitreous hemorrhage (n=1); cyanosis and gastrointestinal symptoms including
erosive gastritis (n=1)). No pts developed systemic lupus erythematosus, serious hypersensitivity
or anaphylaxis. Two pts died during the study (klebsiella pneumoniae (n=1); exacerbation
of CAD (n=1) in a patient with a femoral neck fracture and complex medical history
including myelodysplastic syndrome, approximately 1.5 months after receiving the last
dose of sutimlimab).
Image:
Summary/Conclusion: Sutimlimab, a first-in-class selective anti-C1s classical complement
pathway inhibitor, maintained mean Hb levels >11g/dL, achieved sustained normalization
of mean bilirubin, haptoglobin and reticulocyte count. Sutimlimab continued to improve
FACIT-Fatigue scores, with no newly identified safety concerns at 2 years of treatment.
This study has demonstrated that sutimlimab is an effective and well-tolerated long-term
therapy for the management of chronic CAD through continued upstream inhibition of
the classical CP.
S286: LONG-TERM EFFICACY AND SAFETY RESULTS FROM AN ONGOING OPEN-LABEL PHASE 2 STUDY
OF PARSACLISIB FOR THE TREATMENT OF AUTOIMMUNE HEMOLYTIC ANEMIA (AIHA)
W. Barcellini1,*, I. Murakhovskaya2, L. Terriou3, F. Pane4, A. Patriarca5, K. Butler6,
S. Moran6, S. Wei6, U. Jäger7
1Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy; 2Albert
Einstein College of Medicine/Montefiore Medical Center Bronx, New York, United States
of America; 3Univ. Lille, Inserm, CHU Lille, Centre de Référence des Maladies Autoimmunes
Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), INFINITE – Institute
for Translational Research in Inflammation, Lille, France; 4University of Naples “Federico
II”, Naples; 5University of Eastern Piedmont and AOU “Maggiore della Carità”, Novara,
Italy; 6Incyte Corporation, Wilmington, United States of America; 7Medical University
of Vienna, Vienna, Austria
Background: AIHA is a rare condition caused by autoantibody-mediated hemolysis of
red blood cells. Few therapies beyond steroids and rituximab are available.
Aims: To report updated results from an ongoing multicenter, phase 2, open-label study
of the phosphoinositide 3-kinase-δ inhibitor parsaclisib in patients (pts) with AIHA
(NCT03538041).
Methods: Pts ≥18 years old with warm AIHA (wAIHA), cold agglutinin disease (CAD),
or mixed-type AIHA; hemoglobin (Hgb) 7–10 g/dL; and failure of ≥1 standard therapy
were eligible. After informed consent, pts were treated with oral parsaclisib for
12 wk at a starting dose of 1.0 mg once daily (QD; cohort 1) or 2.5 mg QD (cohort
2). Increases to 2.5 mg QD were allowed in cohort 1 after 6 wk if no clinical response
was achieved and/or transfusion was required; reductions to 1.0 mg QD were permitted
in cohort 2 for tolerability issues. Concomitant corticosteroids (≤20 mg/d prednisone)
were allowed. After 12 wk of treatment, pts responding on parsaclisib could continue
into an extension period. Primary endpoints were efficacy (proportion of pts with
complete response [CR; Hgb ≥12 g/dL] or partial response [PR; Hgb 10–12 g/dL or ≥2 g/dL
increase from baseline, inclusive of CRs] at any visit from Wk 6–12) and safety (treatment-emergent
adverse events [TEAEs]).
Results: As of Aug 5, 2021, 25 pts enrolled and received parsaclisib (cohort 1, n=10
[8 with dose increase]; cohort 2, n=15); 20 pts (80%) completed 12 wk of treatment.
Sixteen (64%), 6 (24%), and 3 (12%) pts had wAIHA, CAD, and mixed AIHA, respectively.
Mean (SD) age was 61.6 (17.0) years; 14 pts (56%) were female and 23 (92%) were White.
Mean (SD) Hgb at baseline was 8.9 (0.8) g/dL, and 9 pts (36%) had transfusions in
the past year. Mean parsaclisib exposure was 334 (range, 7–819) days. Overall, 8 pts
(32%) achieved CR and 16 (64%) had PR at any visit from Wk 6–12. Among pts with wAIHA
(n=16), 14 pts (88%) completed 12 wk of treatment; 8 (50%) and 12 (75%) achieved CR
and PR, respectively, at any visit from Wk 6–12. Seventeen pts entered the extension
period (12 with wAIHA). Increase in Hgb was sustained in the total cohort and among
pts with wAIHA during the initial 12-wk treatment and extension periods (Figure).
During the 12-wk treatment period, TEAEs occurred in 21 pts (84%) overall. Six pts
(24%) had grade (Gr) ≥3 TEAEs, with only neutropenia occurring in >1 pt (n=2 [8%]);
2 pts (8%) had serious AEs (SAEs). Seven pts (28%) had treatment-related AEs, with
only pruritic rash reported in >1 pt (n=2 [8%]). Three pts discontinued during the
12-wk treatment period (AE [n=1; thrombocytopenia], lack of efficacy [n=1], withdrawal
by subject [n=1]), and 2 pts were of unknown status.
During the extension period, 15/17 pts (88%) experienced ≥1 TEAE. Gr ≥3 AEs and SAEs
were each reported in 9 pts (53%). Six pts (35%) had treatment-related AEs, with diarrhea
and rash reported in >1 pt (n=2 [12%] each). One Gr 3 TEAE (psoriasis) and 3 SAEs
(diarrhea [Gr 2], cytomegalovirus reactivation [Gr 2], psoriasis [Gr 3]) were deemed
treatment related. Two (12%) pts had TEAEs leading to parsaclisib discontinuation.
One fatal TEAE (acute respiratory failure in the extension period) was deemed unrelated
to parsaclisib.
Image:
Summary/Conclusion: Parsaclisib was generally well tolerated and resulted in Hgb improvements
as early as Wk 2 that increased over 12 wk of treatment and were sustained through
the extension period. Parsaclisib may be an effective oral treatment for AIHA, and
a randomized, controlled phase 3 trial in wAIHA is now recruiting (NCT05073458).
S287: FACTORS INFLUENCING AUTOLOGOUS LYMPHOCYTE COLLECTIONS FOR CHIMERIC ANTIGEN RECEPTOR
(CAR) T-CELLS – THE ROLE OF T-CELL SENESCENCE
V. Vucinic1,*, T. Tumewu1, M. Brückner1, M. Jentzsch1, F. Ramdohr1, R. Buhmann2, Y.
Remane3, S. Hoffmann1, M. Janz4, O. Penack5, G. Vogtmann1, E. Ruschpler1, L. Bullinger5,
U. Keller4, M. Cross1, S. Schwind1, M. Herling1, G.-N. Franke1, E. Bach1, H. Reinhard2,
U. Platzbecker1
1Medical Clinic and Policlinic for Hematology and Celltherapy; 2Institute for Transfusion
Medicine; 3Pharmacy, University Leipzig, Leipzig Medical Center, Leipzig; 4University
of Berlin, Campus Benjamin-Franklin; 5University of Berlin, Campus Virchow-Klinikum,
Berlin, Germany
Background: The apheresis of autologous CD3+ lymphocytes is the first pivotal step
in the production process of chimeric antigen receptor T-cells. A range of factors
like previous cytotoxic therapies or disease status can influence the quality of collection
but may also impact the fitness of T-lymphocytes. Repeated T-cell activation leads
to progressive loss of expression of CD27 and CD28, receptors shown to be uniformly
present on naïve CD4+ cells (van Leewen et al, J. Immunol, 2004).
Aims: To evaluate factors influencing both collection of autologous lymphocytes and
the subsequent manufacturing of tisagenlecleucel with a special focus on T-cell senescence,
defined as loss of CD27 and CD28.
Methods: Between February 2019 and October 2021, 59 collections were performed for
subsequent CAR-T cell therapy with tisagenlecleucel in 51 patients with relapse/refractory
(r/r) diffuse large B-cell lymphoma and one patient with refractory acute lymphoblastic
leukemia.
The median age was 60.5 (range 17-80) years and 40 (77%) patients were male. The collections
were performed on a Spectra Optia cell-separator by processing a median 3x total blood
volume.
Results: The target numbers of CD3+ cells (>0.55 x109) were successfully collected
within a single collection day in all patients but one. The median yield of CD3+ cells
was 5.0 (0.4-31.9 x109, with yields for CD3+CD4+ and CD3+C8 + 20.1 (range 2.4-102.8)
x108 and 27.4 (range 2.41-266.8) x108, respectively. The median CD4:CD8 ratio was
0.7 (0.08-5.3).
The yields for CD3+CD27+CD28+ and CD3+CD27-CD28- cells were 25.9 (range 1.1-93.0)
x108 and 10.9 (range 0.2-183.8) x108, respectively. We observed no difference in median
collected CD3+ cell yields in patients with > vs ≤ 3 prior therapy lines (p=0.29),
with or without prior treatment with bendamustine (p=0.42) or bone marrow infiltration
(p=0.95).
56 collections underwent further manufacturing, with production according to specification
in 38 (68%) collections. 18 collections resulted in production failure, 13 (72%) due
to insufficient proliferation of T-cells, 1 (6%) because of microbiological contamination
and 4 (22%) due to undeterminable test results for mycoplasms. The manufacturing failures
did not associate with age >60 (p=0.16), sex (p=0.99), bone marrow infiltration (p=0.47)
or >3 prior treatment lines (p=0.78). However, we did notice a trend regarding prior
treatment with bendamustine (p=0.07).
Importantly, the collections resulting in production failure had significantly lower
CD3+/CD27+/CD28+ counts of 16.1 (range 1.1-52.3) vs 33.2 (range 3.3-93.5) x108 (p<0.01),
as well as a trend for lower CD3+/CD4+ counts with 15.9 (range 3.0-43.9) vs 24.5 (range
2.4-102.7) x108 (p=0.05). However, there were no differences in CD3+/CD8+ counts (p=0.19),
CD4:CD8 ratio (p= 0.77) or CD3+/CD27-/CD28- cells (p=0.81; Figure 1).
Image:
Summary/Conclusion: In our real-life cohort of lymphoma patients intended to undergo
treatment with tisagenlecleucel, adequate numbers of CD3 cells could be collected
in most cases irrespective of the number of prior therapies. We could observe a negative
prognostic role of T-cell senescence on the manufacturing process. Further analyses
will be required to determine measures appropriate for further optimization and improvement
of collection outcomes.
S288: A 9 YEAR REVIEW OF BLOOD TRANSFUSION PRACTICE AND ADHERENCE TO NICE GUIDELINES
AT A DISTRICT GENERAL HOSPITAL, UK
N. Clayden1,*, E. O’Donovan1
1Haematology, Surrey and Sussex Healthcare Trust, Surrey, United Kingdom
Background: Blood transfusions are common medical practice in the UK, with approximately
2.1 million blood products being issued by UK blood services in 2020. The Serious
Hazard of Transfusion (SHOT) scheme reported that the risk of death and serious harm
related to transfusions was 1 in 53,193 and 1 in 15,142 in 2020, respectively. Multiple
complications can occur as a result of blood transfusions, with the most common cause
of death being Transfusion Associated Circulatory Overload (TACO). Although low risk,
there has been an increase in deaths related to blood transfusions from 17 deaths
in 2019, to 39 deaths in 2020, in the UK, with 81.6% of adverse reactions and events
being due to preventable errors. This stresses the importance of safe transfusion
practice.
Aims: To review the rates of blood transfusions from 2013 to 2021, and compare our
adherence to NICE guidance. We analysed rates of transfusion, number of red blood
cells (RBC) units per transfusion, haemoglobin levels and the incidence of iron deficiency
anaemia (IDA) prior to transfusion.
Methods: Data was collected retrospectively from patients’ records and the transfusion
laboratory database at Surrey and Sussex healthcare Trust. Adults (>16 years old)
who received inpatient or outpatient RBC transfusions from 2013 to 2021 were included
(n = 53,941). We looked in further detail at the RBC transfusions from the first 2
weeks of December from 2014 to 2021 (n=546).
Results: Figure 1 shows that there has been a significant decline in the number of
RBC transfused from 2014 to 2021; an average gradient of -36.25 units per year (R2=
0.72). There has also been a decline in the average number of units per blood transfusion,
with a downward trendline (gradient -0.15 RBC units/year, p= 0.001). There has been
a significant increase in the percentage of single unit transfusions from 14% in 2014
to 65% in 2021; the trendline has a gradient of +7% / year (p=0.0009). The average
haemoglobin at initiation of transfusion has remained relatively unchanged (69-78g/L),
which is in line with NICE guidance. There is no improvement in transfusion of patients
with IDA; defined as a low transferrin saturation (<20%). The percentage of these
patients being transfused varying from 43% to 79%, with no significant trend over
the years from 2014 to 2021 (p=0.71).
Image:
Summary/Conclusion: The total number of RBC transfused has significantly decreased
over 9 years. However, there is a slight rise from August 2020, which may have been
a result of expansion of the hospital bed base from 697 to 800 between years 2018-2021.
It also may have been impacted by the COVID-19 pandemic. There is a decrease in the
amount of RBC units transfused per patient. We are also encouraged to see a significant
increase in the number of single unit transfusions, in line with NICE Guidance. We
found there was no improvement in the number of patients with IDA being transfused
(average 64%). This indicates that, depending on the clinical scenario, some patients
may be receiving unnecessary blood transfusions. They may benefit from receiving iron
replacement as an alternative treatment, thereby minimising exposing patients unnecessarily
to the risks associated with blood transfusion. Appropriate management of IDA needs
further work in the trust, and we have initiated an IV iron service over the last
18 months to improve this. Limitations of this study include using two weeks of the
year to extrapolate for each year and data may have been skewed over the last two
years due to the COVID-19 pandemic.
S289: MOTIVATORS AND BARRIERS TO BLOOD DONATION AMONG POTENTIAL DONORS OF AFRICAN
AND CAUCASIAN ETHNICITY
H. Fogarty1 2,*, M. Sardana3, L. Sheridan4, P. Chieng3, S. Kelly3, N. Ngwenya2, C.
Sheehan2, K. Morris5, E. Tuohy2
1Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland; 2Department
of Haematology, St James’s Hospital; 3School of Medicine; 4School of Pharmacy, Royal
College of Surgeons in Ireland; 5Irish Blood Transfusion Service, Dublin, Ireland
Background: Blood donors of minority ethnicity, especially those of African origin,
are under-represented in many high-income Western countries, including in Ireland.
Conversely, the rising number of patients with Sickle Cell Disease (SCD) in many European
countries has resulted in increased demand for blood transfusion. Given the high risk
of alloimmunization among haemoglobinopathy patients, extended red blood cell matching
is now recommended. To increase the probability of a phenotypic match, donors and
recipients should share the same racial and/or ethnic background. Hence, the discrepancy
between the number of African blood donors and the number of SCD patients is creating
a strain on the blood supply chain and recruitment of African donors has become a
major priority for transfusion services worldwide.
Aims: A key step towards enhancing recruitment of blood donors is to first understand
perceived barriers and facilitators to blood donation, which may differ across different
demographic categories, including ethnicity. This study aims to explore 1) Barriers
and 2) Motivators to blood donation and 3) Awareness of SCD among potential donors
of diverse ethnic backgrounds in Ireland.
Methods: Following ethical approval, patients attending the National Sickle Cell Disease
and Thalassemia service at St James’s Hospital, Dublin, were invited to share an anonymous,
online, 33-item survey within their local communities to achieve snowball-sampling.
Inclusion criteria were adults aged ≥18 years who had not previously donated blood
in Ireland. Participants were asked to state their age, sex and ethnicity. Responses
for each survey item were recorded on a four-point Likert scale: Strongly Agree, Agree,
Disagree or Strongly Disagree. Survey data were collected from August-December 2021.
Statistical analysis was conducted using GraphPad Prism 9.1 (GraphPad Software Inc.,
USA).
Results: 387 respondents completed the survey, including 311 non-donors (median age
25 years, 67% female). Ethnic backgrounds included: African (59%), Caucasian (25%),
Asian (8%), Hispanic/Latino (3%), Middle Eastern (3%) and Multiracial (2%). The most
common barrier overall was lack of information on blood donation, identified by 60%.
African respondents were 3 times more likely to report lack of information (OR 3.1
CI 1.8-5.3, p<0.0001) and 5 times more likely to report malaria-related barriers than
Caucasians (OR 5.1 CI 2.1-12.3, p=0.0002). Conversely, Caucasians were twice as likely
to report fear of needles/pain (OR 2.1, CI 1.1-3.7, p=0.02). Motivators also varied
by ethnicity, with African respondents 9 times more likely to donate to help someone
within their own community (OR 9.3 CI 4.6-18.3, p<0.00001) and 5 times more likely
to donate for religious motivators (OR 5.5 CI 2.9-10.4 p<0.0001) than Caucasians.
Awareness of SCD was 5 times higher among African respondents (92% versus 66%, OR
5.3 CI 2.7-10.5, p<0.0001).
Summary/Conclusion: This study enhances our understanding of ethnic differences in
barriers and motivators to blood donation. While some barriers are shared across ethnic
groups, including lack of information, notable differences exist between Caucasian
and African respondents. Given that many European countries experience difficulties
in the provision of phenotypically matched blood for SCD patients, these data provide
valuable insights to inform future campaigns aimed at recruitment of African donors.
At a time when the importance of diversity and inclusivity are increasingly recognized
in society, enhancing ethnic diversity in blood donors may reflect a positive force
for healthcare and social equality.
S290: ATRA CAN CORRECT DEFECTIVE HIF-1Α/S1P AXIS-MEDIATED CYTOSKELETAL REORGANIZATION
IN PROPLATELET FORMATION OF ITP
Q.-S. Huang1 2,*, J. Xue1 2, F.-Q. Liu1 2, Q. Chen1 2, G.-C. Zhang1 2, X.-Y. Sun1
2, C.-C. Wang1 2, L.-P. Yang1 2, Y.-Y. Li3 4 5, Q.-F. Wang3 4 5, J. Peng6, M. Hou6,
X.-J. Huang1 2 7 8, X.-H. Zhang1 2 7 8
1Peking University People’s Hospital; 2National Clinical Research Center for Hematologic
Disease; 3Chinese Academy of Sciences (CAS) Key Laboratory of Genomic and Precision
Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute
of Genomics, CAS, Beijing, China; 4China National Center for Bioinformation; 5University
of Chinese Academy of Sciences, Beijing; 6Department of Hematology, Shangdong Key
Laboratory of Immunochematology, and Shandong Provincial Clinical Medicine Research
Center for Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University,
Jinan; 7Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; 8Collaborative
Innovation Center of Hematology, Beijing, China
Background: Bone marrow physiological hypoxia plays a crucial role in haematopoietic
stem cell homeostasis. Hypoxia inducible factor (HIF) is central to mediating the
cellular response to hypoxia. HIF-1α expression was decreased in the bone marrow of
patients with immune thrombocytopenia (ITP), and HIF-1α activation was shown to enhance
megakaryopoiesis in mice. Recent studies suggest that “inside-out” signalling by S1P
in megakaryocytes (MKs) plays a critical role in proplatelet formation (PPF) (Blood,
2013; J EXP MED, 2012). Our previous data indicated that impaired PPF contributed
to the development of thrombocytopenia in ITP. To further explore the underlying mechanism
of impaired PPF in ITP, we found that HIF-1α/S1P axis-mediated cytoskeletal reorganization
was defective in the PPF of ITP. All-trans retinoic acid (ATRA), which has been shown
to be a promising treatment option for ITP patients in our clinical studies (Blood,
2021; Lancet haematology, 2017; Lancet haematology, 2021), could restore cytoskeletal
reorganization and correct impaired PPF.
Aims: This study aimed to explore the role of hypoxia inducible factor-1α (HIF-1α)
in proplatelet formation (PPF) and the underlying mechanisms of ATRA treatment in
ITP patients.
Methods: Thirty consecutive patients with newly diagnosed ITP and 30 healthy donors
were included in our study. MKs were isolated from bone marrow samples. Targeted and
untargeted metabolomic profiling through metabolomic analysis was performed to explore
the relationship between the metabolome and ITP. Confocal microscopy and transmission
electron microscopy were used to observe the PPF and cytoskeleton structure of ITP
MKs. An ITP mouse model was established to observe the therapeutic effects of ATRA
in the PPF.
Results: In the present study, we observed that MKs displayed altered cytoskeletal
reorganization and impaired proplatelet formation (PPF) in ITP patients. Targeted
and untargeted metabolite profiling revealed a decreased sphingosine-1-phosphate (S1P)
level in ITP. Downregulated sphingosine kinase 2 (SPHK2) expression in MKs accounted
for the low level of S1P in ITP. S1P is essential for S1P receptor 1 (S1PR1) and Rac1
activation, Src family kinases (SFKs) activity, and subsequent cytoskeletal reorganization
and PPF regulation. Moreover, we demonstrated that HIF-1α mediated SPHK2 activation
and S1P production. Decreased HIF-1α levels were found in the MKs of patients with
ITP, contributing to impaired PPF.
We then investigated the effect of ATRA on PPF in ITP patients. ATRA upregulated HIF-1α
and SPHK2 expression, increased S1P production and corrected impaired PPF in vitro.
In an ITP mouse model, ATRA alleviated thrombocytopenia and restored cytoskeletal
reorganization. ATRA corrected impaired PPF by upregulating HIF-1α expression. The
exposure of ITP MKs to selective RARα (AM580) or RARγ (BMS961) agonists did not change
PPF. However, the treatment of ITP MKs with a selective RARβ agonist (CD2314) significantly
increased PPF. Furthermore, we found that the effect of ATRA on enhancing PPF in ITP
MKs was reversed in the presence of an RARβ antagonist (CD2665). These data suggest
that impaired PPF in ITP MKs is corrected by ATRA in a RARβ-dependent manner.
Summary/Conclusion: Together, our data show that the HIF-1α/S1P axis mediates altered
cytoskeletal reorganization and impaired PPF in ITP and suggest that ATRA correction
of impaired PPF is a potential mechanistic explanation for the clinical efficacy of
ATRA in ITP.
S291: PHASE I/II STUDY OF RILZABRUTINIB, AN ORAL BRUTON TYROSINE KINASE INHIBITOR,
IN PATIENTS WITH IMMUNE THROMBOCYTOPENIA: LONG-TERM FOLLOW-UP
D. J. Kuter1,*, M. Efraim2, Z. Kaplan3, J. Mayer4, P. Choi5, A. G. Jansen6, V. McDonald7,
R. Baker8, R. Bird9, M. Garg10, J. Gumulec11, M. Kostal12, T. Gernsheimer13, W. Ghanima14,
M. Yao15, A. Daak16, N. Cooper17
1Hematology Division, Massachusetts General Hospital, Harvard Medical School, Boston,
United States of America; 2Multiprofile Hospital for Active Treatment Sveta Marina
EAD, Varna, Bulgaria; 3Monash Medical Centre, Clayton, Australia; 4Department of Internal
Medicine, Hematology and Oncology, Masaryk University Hospital, Brno, Czechia; 5The
Canberra Hospital, Garran, Australia; 6Erasmus MC, University Medical Center, Rotterdam,
Netherlands; 7Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom;
8Perth Blood Institute, Murdoch University, Perth; 9Princess Alexandra Hospital, Woolloongabba,
Australia; 10Leicester Royal Infirmary, Leicester, United Kingdom; 11Department of
Hematooncology, University Hospital Ostrava and Faculty of Medicine University of
Ostrava, Ostrava; 12Fourth Department of Internal Medicine and Hematology, Faculty
of Medicine, University Hospital of Hradec Kralove, Hradec Kralove, Czechia; 13University
of Washington Medical Center, Seattle, United States of America; 14Østfold Hospital
Foundation, Gralum, Norway; 15Biostatistics, Sanofi US Services Inc., Bridgewater;
16Sanofi Genzyme, Cambridge, United States of America; 17Department of Medicine, Hammersmith
Hospital, London, United Kingdom
Background: Immune thrombocytopenia (ITP) is an autoimmune disease associated with
autoantibody-mediated platelet destruction and impaired platelet production, resulting
in thrombocytopenia and high bleeding risk. In a global phase I/II trial (NCT03395210)
of heavily pretreated patients with long-standing disease, the Bruton tyrosine kinase
inhibitor rilzabrutinib showed a rapid and durable platelet count increase and was
well tolerated. We present results for patients initiating rilzabrutinib 400 mg BID
who are continuing in the long-term extension (LTE).
Aims: Assess if the efficacy and safety of rilzabrutinib 400 mg BID continue to be
maintained in LTE patients on rilzabrutinib ± concomitant medication.
Methods: Patients with 2 baseline platelet counts <30×109/L were required to have
responded to ≥1 prior ITP therapy but at baseline were unable to maintain an adequate
response to prior/concomitant therapies. The main treatment period was 24 weeks; patients
who achieved platelet counts ≥50×109/L at ≥50% of the visits during the last 8 weeks
of the main period were permitted to continue 400 mg BID in the LTE. Primary endpoints
were safety and efficacy (≥2 consecutive platelet counts ≥50×109/L and increased ≥20×109/L
from baseline without requiring rescue medication). Stable doses of concomitant ITP
medication (thrombopoietin-receptor agonists [TPO-RA] and corticosteroids [CS]) were
allowed for patients with inadequate platelet response. Patients who received rescue
medication discontinued from the study. All patients provided informed consent.
Results: At baseline, the 45 patients initiating 400 mg BID in the main period had
a median age of 49 years, median duration of ITP of 6.1 years, median platelet count
of 15×109/L, and a median of 4 unique prior therapies (24% prior splenectomy). A total
of 15 patients (33%) received rilzabrutinib monotherapy, and 30 had concomitant therapy
(TPO-RA n=13 [29%], CS n=12 [27%], TPO-RA + CS n=5 [11%]). Primary platelet response
was achieved by 18 patients (40%): 6 on monotherapy and 12 on concomitant medication.
Platelet counts of ≥50×109/L, ≥30×109/L, and ≥20×109/L from baseline were maintained
for a median of 71%, 95%, and 87% of weeks, respectively. As of 21Jan2022, 16/60 patients
in the main study population had proceeded to LTE, of whom 11 were ongoing. Five LTE
patients (31%) continued on rilzabrutinib monotherapy and 11 used concomitant medication
(TPO-RA n=2 [13%], CS n=7 [44%], TPO-RA + CS n=2 [13%]; Figure); 1 patient used rescue
medication. Median platelet count at LTE entry was 87×109/L. In patients on rilzabrutinib
monotherapy, the median platelet count was 68×109/L, which was sustained at 6 months
of follow-up (Table). In all LTE patients, platelet counts ≥50×109/L, ≥30×109/L, and
≥20×109/L from baseline were maintained for a median of 88%, 100%, and 97% of weeks,
respectively. In patients on rilzabrutinib ± concomitant medication, results were
consistent at 3 and 6 months of follow-up (Table). Three patients (19%) experienced
related treatment-emergent adverse events (TEAEs), including 1 grade 2 upper respiratory
tract infection. Three patients (19%) discontinued rilzabrutinib due to a TEAE. Six
patients had ≥1 bleeding event, none were treatment-related; no related serious adverse
events or deaths. The average ITP-BAT bleeding scale score at 6 months showed no increase
in bleeding in the LTE.
Image:
Summary/Conclusion: With extended treatment duration, rilzabrutinib 400 mg BID showed
durable clinical efficacy and was well tolerated in patients on rilzabrutinib ± concomitant
medication.
S292: SUSTAINED RESPONSE OFF TREATMENT IN ELTROMBOPAG-TREATED PATIENTS WITH ITP WHO
ARE REFRACTORY OR RELAPSED AFTER FIRST-LINE STEROIDS: PRIMARY ANALYSIS OF THE PHASE
II TAPER TRIAL
N. Cooper1,*, W. Ghanima2, N. Vianelli3, D. Valcárcel4, I. Yavaşoğlu5, A. Melikyan6,
E. Yañez Ruiz7, J. Haenig8, J. Lee9, J. Maier8, N. Zolkin10, F. Zaja11
1Centre for Haematology, Department of Immunology and Inflammation, Imperial College
London, Hammersmith Hospital, London, United Kingdom; 2Department of Medicine, Østfold
Hospital Trust, Kalnes, Norway; 3Scientific Institute for Research, Hospitalization
and Healthcare (IRCCS), Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy;
4Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), University
Hospital Vall d’Hebron, Barcelona, Spain; 5Department of Hematology, Adnan Menderes
University, Aydin, Turkey; 6National Research Center for Hematology, Moscow, Russia;
7Hematology-Oncology Unit, Department of Internal Medicine, School of Medicine, Universidad
de la Frontera, Temuco, Chile; 8Novartis Pharma AG, Basel, Switzerland; 9Novartis
Pharmaceuticals Corporation, East Hanover, NJ, United States of America; 10IQVIA,
St. Petersburg, Russia; 11Department of Medical, Surgical and Health Sciences, University
of Trieste, Trieste, Italy
Background: Corticosteroids (CSs) are the standard 1st-line treatment for primary
immune thrombocytopenia (ITP); however, long-term CS use is associated with high relapse
rates and considerable toxicity. Treatments that achieve a sustained response and
reduce the need for long-term CS use are needed. Eltrombopag (EPAG) is a thrombopoietin
receptor agonist (TPO-RA) indicated in Europe for the treatment of patients (pts)
aged ≥1 year with primary ITP lasting ≥6 months who are refractory to other treatments
(eg, CSs). Evidence suggests that a proportion of pts treated with TPO-RAs achieve
sustained responses that are maintained after TPO-RA tapering and discontinuation;
however, much of the evidence is retrospective with only a few prospective studies
investigating sustained response off treatment (SRoT).
Aims: TAPER (NCT03524612), a Phase II, open-label, prospective, single-arm study,
aims to determine whether EPAG can induce SRoT in pts with ITP after 1st-line CS failure.
Methods: Adult (≥18 years) pts with ITP who did not respond to or had relapsed after
initial CS therapy, with platelet counts <30×109/L and assessed as needing treatment,
were included. Patients received a 50 mg/day starting dose of EPAG (25 mg/day for
East/Southeast Asian pts; 12.5 mg/day for Japanese pts in Japan), which could be increased
up to 75 mg/day (50 mg/day in Japan) if needed. The primary endpoint was the number
(%) of pts with SRoT by Month 12; SRoT was defined as achieving a complete response
(CR, ie, platelet count ≥100×109/L), then maintaining a stable platelet count (no
counts <70×109/L) for 2 months, followed by successful EPAG tapering and discontinuation
with platelet counts ≥30×109/L and no bleeding events or rescue therapy. Patients
with SRoT at Month 12 were followed for a further year. Secondary outcomes included
SRoT duration and platelet count changes from baseline. Data to Month 12 are presented.
Results: N=105 pts were enrolled. The median (interquartile range [IQR]) age was 46
(30-65) years; 61% were female. In the 1st 12 months, median (IQR) duration of exposure
to EPAG was 5.6 (2.3-11.9) months and median (IQR) EPAG dose was 57.1 (37.5-69.0)
mg/day. Overall, 89 pts (85%) achieved CR at least once and 65 pts (62%) maintained
a platelet count ≥70×109/L for 2 months after CR. EPAG tapering and discontinuation
was achieved in 44 pts (42%). The primary endpoint was met, with 32 pts (30.5% [95%
confidence interval, 21.9-40.2]; P<0.001 [H1: P>15%; alpha: 0.05]) achieving SRoT
until Month 12 (Fig. 1); SRoT was maintained from last dose to Month 12 for a median
(IQR) of 33.3 (25.7-45.3) weeks. The median (IQR) absolute increase in platelet counts
from baseline at Month 12 was 77.0×109/L (35.0-145.0). All-grade adverse events (AEs)
occurred in 92/105 (88%) pts (grade ≥3 AEs: 33/105 [31%]). Treatment-related AEs occurred
in 37 (35%) pts (8 [7.6%] grade ≥3). The most common all-grade AEs were headache (21%
of pts with ≥1 event [grade ≥3: 1% of all pts]), thrombocytopenia (17% [10.5%]), and
petechiae (11% [1%]). There were 4 deaths (none were considered treatment related):
3 were on-treatment (central nervous system hemorrhage [n=1], intracranial hemorrhage
[n=1], metastases to peritoneum [n=1]) and 1 death (malignant neoplasm) occurred 238
days after last dose.
Image:
Summary/Conclusion: Data from the TAPER study indicate that a significant proportion
of pts experience sustained response following tapering and discontinuation of EPAG,
even after a relatively short duration of exposure. Overall EPAG was well tolerated
with no unexpected AEs.
S293: RISK OF FRACTURES IN ADULT PATIENTS WITH PRIMARY AND SECONDARY IMMUNE THROMBOCYTOPENIA:
A DANISH NATIONWIDE COHORT STUDY
N. Mannering1 2,*, D. L. Hansen1 2, G. Moulis3 4, W. Ghanima5 6, A. Pottegård7, H.
Frederiksen1 2
1Department of Hematology, Odense University Hospital; 2Clinical Institute, University
of Southern Denmark, Odense, Denmark; 3Department of Internal Medicin; 4Clinical Investigation
Center 1436, Team PEPSS, Toulouse University Hospital, Toulouse, France; 5Department
of Hematology, Østfold Hospital; 6Institute for Clinical Medicine, University of Oslo,
Oslo, Norway; 7Department of Public Health, University of Southern Denmark, Odense,
Denmark
Background: The mainstay of first line treatment in immune thrombocytopenia (ITP)
has remained high-dose corticosteroids for decades. Although steroid treatment is
recommended to be tapered quickly in ITP, previous data shows that during the first
six weeks of primary ITP treatment the mean accumulated steroid dose was 2-3 g of
prednisolone. In addition, patients may be exposed to repeated courses of corticosteroids
during relapses due to its rapid effect on platelet count. Corticosteroids is a well-known
risk factor for bone demineralization and osteoporosis that may lead to fractures,
but it is unknown if patients with ITP suffer an increased risk of fractures.
Aims: In this study we investigated incidences of fractures in primary (pITP) and
secondary ITP (sITP) compared to the general population.
Methods: Incident patients with ITP ≥18 years were identified in the nationwide Danish
health registries during 1980-2016 by using the first registration of the designated
codes 287.10 (ICD-8) or D.69.3 (ICD-10). Prevalent ITP and other thrombocytopenic
conditions were excluded. Secondary ITP was classified when one or more associated
diagnoses were registered any time before or up to 30 days after ITP diagnosis.
Each patient with ITP was age-sex matched with up to 40 comparators from the general
population. Date of the first ITP registration marked start date of follow-up, and
comparators were assigned the same start date. Incident fractures were identified
using designated fracture registrations, and divided into five groups: hip and femoral,
humeral, antebrachial, axial, and any of the before mentioned. All individuals were
followed from start date until the first of the following: fracture, death, emigration,
or end of study. Using these data, we calculated rates, incidence-rate-ratios (IRR)
and cumulative incidences for fractures in patients vs. comparators after 1, 5 and
10 years, and end of study period.
Results: We identified 4,789 patients with pITP, 654 patients with sITP, and 217,370
comparators. Median age was 59.6 years for primary ITP and 57.3 for secondary ITP.
Women constituted 54% of primary ITPs, and 64% of secondary ITP patients.
IRR in pITP was 1.15 [95% CI 1.04; 1.27] for any fracture, and 1.34 [1.09; 1.62] for
axial fractures. For sITP, overall IRR was 1.37 [1.06; 1.74] for any fracture, and
1.65 [1.16; 2.29] for hip – and femoral fractures.
IRR was significantly elevated for any fracture during the first 5 years after diagnosis
of pITP, but equalized hereafter. The IRR of hip – and femoral fractures in the first
year after diagnosis was 2.04 [1.41; 2.86] in pITP, and 2.30 [1.32; 3.74] for axial
fractures, however these differences also equalized over time. For sITP, the risk
of any fracture was particularly elevated during the first year with an IRR of 2.65
[1.39; 4.63], as well as the risk of distal antebrachial fracture with an IRR of 3.16
[1.33; 6.46]. During year 2-5, IRR for hip – and femoral fracture was 1.92 [1.04;
3.26]. All remaining IRR-estimates were not statistically significant.
Cumulative incidence proportions showed similar trends (Figure) with largest differences
during first years after diagnosis, but equalizing over time.
Image:
Summary/Conclusion: The incidence of some fractures in ITP is increased during the
first years after diagnosis compared to the general population. Clinical attention
and action should be directed upon this matter.
Data on comorbidity, sub grouped cumulative incidences, and additional risk estimates
will be presented with the abstract.
S294: LONG-TERM SAFETY AND EFFICACY OF CAPLACIZUMAB FOR ACQUIRED THROMBOTIC THROMBOCYTOPENIC
PURPURA (ATTP): THE POST-HERCULES STUDY
M. Scully1,*, J. de la Rubia2 3, K. Pavenski4 5, A. Metjian6, P. Knöbl7, F. Peyvandi8
9 10 11, S. Cataland12, P. Coppo13, J. A. Kremer Hovinga14, R. De Passos Sousa15,
F. Callewaert16, S. Gunawardena17, J. Lin17
1Department of Haematology, University College London Hospitals, London, United Kingdom;
2Hematology Department, Internal Medicine, School of Medicine and Dentistry, Catholic
University of Valencia; 3Hospital Doctor Peset, Valencia, Spain; 4Departments of Medicine
and Laboratory Medicine, St. Michael’s Hospital; 5University of Toronto, Toronto,
ON, Canada; 6Division of Hematology, Department of Medicine, University of Colorado–Anschutz
Medical Center, Denver, CO, United States of America; 7Department of Medicine; 1,
Division of Hematology and Hemostasis, Medical University of Vienna, Vienna, Austria;
8Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico; 9Angelo Bianchi Bonomi
Hemophilia and Thrombosis Center; 10Fondazione Luigi Villa; 11Department of Pathophysiology
and Transplantation, Università degli Studi di Milano, Milan, Italy; 12Department
of Internal Medicine, Ohio State University, Columbus, OH, United States of America;
13Department of Hematology, Reference Center for Thrombotic Microangiopathies (CNR-MAT),
Saint-Antoine University Hospital, AP-HP, Paris, France; 14Department of Hematology
and Central Hematology Laboratory, Inselspital, Bern University Hospital, University
of Bern, Bern, Switzerland; 15Sanofi, Lisbon, Portugal; 16Sanofi, Diegem, Belgium;
17Sanofi, Cambridge, MA, United States of America
Background: The efficacy and safety of caplacizumab (CPLZ) for patients with acquired
thrombotic thrombocytopenic purpura (aTTP; also known as immune-mediated TTP) was
demonstrated in the Phase 3 HERCULES trial, with a 28-day follow-up period after end
of treatment.
Aims: To evaluate long-term safety and efficacy of CPLZ in patients with aTTP, and
safety and efficacy of repeated CPLZ use for aTTP recurrence.
Methods: In post-HERCULES (NCT02878603), patients who completed the HERCULES trial
were invited to attend twice-yearly visits over 3 years. Patients could receive open-label
CPLZ with therapeutic plasma exchange (TPE) and immunosuppressive therapy (IST) in
case of aTTP recurrence. Safety was assessed during the overall study period in the
intention-to-observe (ITO) population (all enrolled patients; n=104). TTP-related
events (TTP-related mortality, recurrence, or major thromboembolic events) were assessed
in patients without a recurrence during HERCULES or prior to the start of post-HERCULES
(efficacy ITO population, n=78). Safety and efficacy outcomes were also evaluated
during recurrences (recurrence population, n=19), including patients treated at least
twice with CPLZ (received CPLZ in HERCULES or twice in post-HERCULES; repeat-use population,
n=9). All patients provided informed consent.
Results: Of 104 patients who enrolled, 75 had been treated with CPLZ with TPE+IST
during HERCULES (CPLZ group) and 29 had been treated with TPE+IST only (placebo group).
Incidence of adverse events (AEs) and serious AEs during the overall study period
was similar between groups. Recurrence occurred in 11/75 patients (15%) in the CPLZ
group and 8/29 (28%) in the placebo group. In the efficacy ITO population, TTP-related
events occurred in 4/49 patients (8%) randomized to CPLZ versus 11/29 (38%) randomized
to placebo; in patients randomized to CPLZ, relapse rates were similar between patients
with (10%) versus without (7%) prior rituximab use. A total of 19 patients had ≥1
recurrence; 13 were treated with CPLZ (6/13 received concomitant rituximab during
the first recurrence period). The first recurrence episode was resolved (12/13) or
resolving (1/13) for all patients treated with CPLZ, including the 9 patients with
repeat CPLZ use. All second recurrences (6/6) were resolved. One patient who was not
treated with CPLZ in either HERCULES or post-HERCULES died as an outcome of recurrence.
For the first recurrence period, the median (min, max) time to initial platelet count
≥150x109/L with subsequent stop of TPE within 5 days was 7.0 (3, 24) days for patients
treated with CPLZ (n=13) and 10.0 (5, 15) days for patients not treated with CPLZ
(n=5); duration of TPE was 5.0 (2, 19) days (n=13) and 5.5 (4, 7) days (n=4), respectively;
length of hospital stay was 7.0 (4, 23) days (n=11) and 10.0 (9, 11) days (n=3), respectively.
The safety profile of CPLZ for recurrence was consistent with that observed in HERCULES
(Table). In the repeat-use population (n=9), bleeding events were reported in 5/9
patients (56%) for the first recurrence; 2 patients had a serious treatment-related
treatment-emergent AE: genitourinary bleeding (in 1 patient during the first recurrence)
and gastrointestinal bleeding (in 1 patient during the second recurrence).
Image:
Summary/Conclusion: The long-term safety profile of patients treated with CPLZ in
combination with TPE+IST was generally similar to those who received IST+TPE only,
with no observed increases in aTTP recurrence. Repeat use of CPLZ was efficacious,
with no new safety concerns.
S295: CLINICAL RELEVANCE OF SPLEEN VOLUME AND PLATELET COUNT WITH BLEEDING EVENTS
IN PATIENTS WITH ACID SPHINGOMYELINASE DEFICIENCY (ASMD)
M. Fournier1, J. Msihid1, A. Willemze2, F. Laredo3, R. Pulikottil-Jacob4,*
1Sanofi, Chilly-Mazarin, France; 2Sanofi, Amsterdam, Netherlands; 3Sanofi, São Paulo,
Brazil; 4Rare Disease, Health Economics and Value Assessment, Sanofi, Berkshire, United
Kingdom
Background: Acid Sphingomyelinase Deficiency (ASMD) is an ultra-rare, lysosomal storage
disease with a broad spectrum of clinical manifestations. Within that spectrum, ASMD
type B and type A/B are often referred to as chronic ASMD. In Type 1 Gaucher disease,
increased bleeding tendency is attributed to thrombocytopenia secondary to hypersplenism
and/or bone marrow infiltration noticed in patients with splenomegaly and thrombocytopenia.
However, ASMD patients have a bleeding tendency that is not proportionally attributed
to any association or explanation; hence further research is needed to study the cause
of bleeding among patients with ASMD.
Aims: To characterize the association between bleeding events (BE), spleen volume
(SV), and platelet count by retrospectively analyzing data from a previously published
prospective, multicenter, longitudinal study (MSC12840 [SPHINGO-001-00]) in patients
with chronic ASMD (N=59; age: 7–64 years; 30 were < 18 years; male: 53%; up to 3 visits
per patient with 6–11 years follow-up, 2001–2012).
Methods: This study retrospectively screened medical charts of 59 adults and pediatrics
for BE specified under the hematopoietic body system and their respective SV and platelet
count. SV (in multiple of normal, MN) and platelet count (in 103/UL) were measured
at baseline, year 1, and end of study [median years of observation (range), 10.2 years
(4.7–11.1)]. As reported in the medical history captured at each visit, BE were selected
with a series of keywords indicating hemorrhage, bleed, or transfusion post-medical
review. To characterize their association with BE, SV and platelet counts were considered
at the visit prior to the observation of the event. A general linear mixed model was
performed using SV as a continuous or a class variable (<6, 6–15, and >15 MN). The
associations between platelet counts (below 100 x 103/UL) and SV were also explored.
Results: Overall, 59 patients were enrolled, 50 patients completed year 1 visit, and
32 patients completed the final visit (9 died, 9 discontinued, and 9 were lost to
follow-up). Twenty-six patients reported BE across the two observed periods, i.e.,
baseline to 1 year and 1 year to final study visit. No BE was reported with SV <6
MN at any period; however, there was a trend for a higher event rate in patients with
higher SV, per visit (Table 1). A higher SV of +5 MN or +10 MN was associated with
a 1.8-times (odds ratio [95% confidence interval]: 1.85 [1.05, 3.29]) and 3.4-times
(3.45 [1.10, 10.82]) increased risk of BE, respectively. Severe splenomegaly (>15
MN) was associated with a higher and non-significant risk of BE vs. moderate splenomegaly
(6–15 MN; 2.54 [0.58, 11.08]). In addition, patients with low platelet counts (<100
x 103/UL) had higher SV, per visit (Table 1). The p-value for the chi-square test
for association between platelet count and SV across visits was 0.006; it was 0.058,
0.127, and 0.302 at baseline, year 1, and end of the study, respectively. There was
a moderate negative correlation (r = –0.47) between platelet count and SV across visits.
Image:
Summary/Conclusion: This analysis highlighted the association between SV and BE among
patients with chronic ASMD. Since a statistically significant association between
SV and thrombocytopenia was observed across visits, hypersplenism appears to be a
contributor for the pathophysiology of bleedings among these patients. These associations
help better understand the clinical relevance of SV measurements in ASMD. Prospective
validation is warranted to confirm the results of these analyses.
S296: IN VITRO ANALYSIS OF THE IMPACT OF INHIBITORS ON THE PROCOAGULANT EFFECTS OF
BYPASS AGENTS OR HEMOSTATIC FACTORS IN COMBINATION WITH EMICIZUMAB
A. Dos Santos Ortas1,*, E. García Arias-Salgado1, E. Monzón Manzano1, P. Acuña1, M.
T. Álvarez Roman1, M. Martín Salces1, M. I. Rivas Pollmar1, E. García Pérez1, M. Gutiérrez
Alvariño1, A. Gonzalez Ceberino1, S. García Barcenilla1, N. Butta1, V. Jiménez Yuste1
1HOSPITAL UNIVERSITARIO LA PAZ, MADRID, Spain
Background: Emicizumab is a non-replacement treatment used in hemophilia A patients
(PwHA). However, patients on prophylaxis with emicizumab exhibit mild-moderate bleeding
phenotypes, thus Factor (F) VIII or bypassing agents (BPAs) are required to control
breakthrough or perioperative bleeds. Increments of FIX levels, the main limiting
factor for the FIXa-emicizumab-FX complex formation, also produce an increase of the
emicizumab procoagulant effects similar to those obtained with FVIII or BPAs.
Aims: To elucidate the impact of FVIII inhibitors in the efficacy of this hemostatic
response, we compared the in vitro procoagulant effects of BPAs and FIX products in
samples from emicizumab-treated patients with or without inhibitors.
Methods: Blood from 21 patients on prophylaxis with emicizumab, 8 with inhibitor,
was collected in tubes with CTI (Corn Trypsin Inhibitor) to prevent activation of
the contact pathway.
Spiking with therapeutic doses of 100 IU/dl octocog-alfa (rFVIII), BPAs [1 µg/ml eptacog
alfa activated (rFVIIa); 5 U/dl activated prothrombin complex concentrate (aPCC)]
or 100 IU/dl of plasma-derived FIX (pFIX), two standard-half-life (SHL) rFIX (nonacog-alfa
and nonacog-gamma), and one extended-half-life (EHL) rFIX (albutrepenonacog-alfa)
were tested.
Clotting time (CT) was evaluated by Rotational Thromboelastometry (ROTEM) with whole
blood, and Thrombin Peak by thrombin generation test (TGT) in plasma using low tissue
factor concentration.
Results: Emizicumab-treated samples showed prolonged CT values (Fig.1A) and lower
thrombin peak levels (Fig.1B) than healthy controls and no significant differences
were observed between samples with and without inhibitors.
Addition of 100 IU/dl of all FIX products normalized the CT values similarly to the
addition of the same dose of rFVIII. Nonacog-alfa was more efficient producing similar
procoagulant effects than the addition of 1μg/ml rFVIIa or 5 U/dl aPCC. Reduction
of CT was similar in samples from PwHA without or with inhibitors in all assayed conditions
(Fig.1A).
Higher thrombin peak values were also obtained by the addition of all the factors
or BPAs tested by TGT but the procoagulant effect of all the recombinant FIX was significantly
higher in samples with inhibitors (Fig.1B).
Image:
Summary/Conclusion: Procoagulant effect of BPAs with emicizumab was similar with or
without inhibitors whereas thrombin generation increment produced by rFIX was enhanced
in the presence of inhibitors. These results provide a rationale for investigating
the concomitant use of rFIX and emicizumab in PwHA with inhibitor in breakthrough
bleeding and surgery.
Funding: ISCIII-FEDER (PI19/00631); CLS Behring; Catedra UAM-Roche; and Investigator-Initiated
Reserach grant (no. IISR-2019-104361) from Baxalta GmbH, a wholly owned subsidiary
of Takeda Pharmaceutical Comany Limited.
S297: ASSESSING THE COST-EFFECTIVENESS OF LONG-TERM PROPHYLAXIS STRATEGIES IN VON
WILLEBRAND DISEASE
M. Wilson1, G. Castaman2, G. Escolar3, W. Miesbach4, S. Santos5, S. Yan6,*
1RTI Health Solutions, Research Triangle Park, NC, United States of America; 2Center
for Bleeding Disorders, Careggi University Hospital, Florence, Italy; 3Department
of Hematopathology, Centre Diagnostic Biomedic, Hospital Clinic, Barcelona, Spain;
4Haemophilia Centre, Medical Clinic II, Institute of Transfusion Medicine, Goethe
University Hospital, Frankfurt am Main; 5CSL Behring, Hattersheim am Main, Germany;
6CSL Behring, King of Prussia, PA, United States of America
Background: Von Willebrand disease (vWD) is the most common inherited bleeding disorder.
Patients with vWD may experience excessive bleeding events resulting in morbidity,
reduced quality of life, and a substantial economic burden.
Management strategies include von Willebrand factor concentrates either as on-demand
treatment (ODT) of bleeds or long-term prophylaxis (LTP) to prevent bleeds. According
to recent treatment guidelines, long-term prophylaxis is recommended in individuals
with severe and frequent bleeds.
Aims: To assess the cost effectiveness of ODT and LTP treatment strategies in vWD
patients with low, medium and high annual bleed rate (ABR), using pdVWF/FVIII 2,4:1
products (2,4:1 product A in the United Kingdom (UK) and 2,4:1 product B in Sweden).
Methods: A Markov structure considering risk of joint surgery and bleed rate was used
to estimate the life years (LYs), quality-adjusted life years (QALYs), and costs of
vWD treatment over a lifetime horizon. Treatment options included ODT or LTP with
pdVWF/FVIII 2,4:1 or pdVWF/FVIII 1:1 in the UK and in Sweden. Product dosing for LTP
and for ODT were obtained from each product’s summary of product characteristics or
assumed equal to similar products where data are not available. Bleed risks (major
and minor bleed) were estimated for each product in ODT and LTP using published prophylaxis
studies and were used to estimate costs of product needed and to determine the probability
of requiring joint surgery over time based on the progression of Pettersson Score.
Resource use (inpatient and outpatient) costs for bleeds and joint surgery were obtained
from standard country-specific costing sources. All costs are shown in 2021 GBP or
Swedish Kr for the UK and Swedish analyses, respectively. Health-state utility weights
and disutilities of bleeds were obtained from published literature. Annual discount
rates for costs and outcomes were 3.5% for the UK and 3% for Sweden. One-way and probabilistic
sensitivity analyses were conducted to evaluate the impact of parameter uncertainty.
Each analysis considered a low, medium, and high ABR patient population.
Results: In the base case analyses (medium ABR) over a lifetime horizon, pdVWF/FVIII
2,4:1 LTP regimens were cost-effective compared with ODT. For pdVWF/FVIII 2,4:1 in
the UK, LTP was both cost-saving (-£831,206 incremental cost) and more effective (6.14
incremental QALY) than ODT. Costs of bleed events more than offset the costs of prophylaxis.
In Sweden with pdVWF/FVIII 2,4:1, LTP was also dominant (-8,841,901 kr; 6.52 QALY)
compared with ODT. In comparisons of LTP regimens, pdVWF/FVIII 2,4:1 was dominant
versus pdVWF/FVIII 1:1 in both the UK (-£2,307,370; 1.56 QALY) and in Sweden (-21,939,947
kr; 1.42 QALY). In comparison of ODT regimens, pdVWF/FVIII 2,4:1 was less expensive
than pdVWF/FVIII 1:1 in the UK (-£1,250,369; equal QALY) and in Sweden (-7,923,741
kr; equal QALY) due to differences in product costs. Results were similar for comparisons
in the high ABR populations. In probabilistic sensitivity analyses, pdVWF/FVIII 2,4:1
was dominant in over 95% of simulations in the UK and in over 85% of simulations in
Sweden, and was cost-effective in 95% or more of simulations in both countries.
Summary/Conclusion: These results suggest that LTP with pdVWF/FVIII 2,4:1 is a cost-effective
strategy compared with ODT, in both the UK and Sweden, for medium and high ABR vWD
patients. pdVWF/FVIII 2,4:1 was shown to be cost-effective compared with pdVWF/FVIII
1:1 in the LTP setting, as well as less costly for patients treated ODT for both the
UK and Swedish settings, respectively.
S298: CS585 IS A FIRST-IN-CLASS COMPOUND TARGETING THE IP RECEPTOR FOR PREVENTION
OF THROMBOSIS WITHOUT INCREASED RISK OF BLEEDING
S. Lambert1, R. Adili1, P. Yalavarthi1, N. Rhoads1, B. Dahlof2 3, A. White4, N. Bergh2
3, M. Holinstat1 5,*
1Pharmacology, University of Michigan Medical School, Ann Arbor, United States of
America; 2Cereno Scientific; 3Institute of Medicine, University of Gothenburg, Gothenburg,
Sweden; 4Medicinal Chemistry; 5Internal Medicine, University of Michigan Medical School,
Ann Arbor, United States of America
Background: Uncontrolled platelet activation leads to the formation of occlusive thrombi
resulting in myocardial infarction, stroke, VTE and critical limb ischemia. There
is an unmet need for more efficacious anti-thrombotics with less bleeding in this
area. Our group recently identify a novel oxidized lipid in the blood that potently
and selectively inhibits platelet activation through activation of the prostacyclin
receptor.
Aims: Develop a first-in-class antiplatelet drug, CS585, that potently and selectively
inhibits platelet reactivity and thrombosis without altering hemostasis and bleeding
risk.
Methods: We developed a mimetic of the oxidized lipid shown to selectively bind to
and activate the prostacyclin receptor. Using human platelets, we assessed the ability
of CS585 to inhibit platelet activation by assessing 1)aggregometry, 2)adhesion under
arterial flow, and 3) granule secretion and integrin activation using flow cytometry.
We additionally assessed thrombosis in vivo in 2 different mouse models as well as
bleeding. Finally, we assessed potential off-target effects using thromboelastography
(TEG).
Results: CS585 potently inhibited both collagen and thrombin-induced platelet aggregation.
This inhibition was confirmed by measuring inhibition of αIIbβ3 activation, α-granule
secretion, and dense-granule secretion by flow cytometry. Selectivity was confirmed
in human platelets by demonstrating a full reversal of inhibition when the IP receptor
was pharmacologically blocked or genetically eliminated in IP receptor knockout mice.
In vivo inhibition of injury-induced thrombosis in the small (laser-induced cremaster
thrombosis model) and large (FeCl3-induced carotid artery thrombosis model) vessels
by CS585 was demonstrated and no increased risk for bleeding was observed using the
tail vein bleeding model. Finally, TEG experiments in human blood spiked with CS585
demonstrated no delay or decrease in clot strength confirming the tail vein bleeding
assay experiments in the mouse.
Summary/Conclusion: We have shown for the first time that CS585, a first-in-class
analog of an oxidized lipid in the blood potently and selectively activates the prostacyclin
receptor resulting in inhibition of human and mouse platelet activation and thrombosis
without an increased risk of bleeding. This discovery represents a new class of inhibitors
for prevention of platelet activation and thrombosis and protection from myocardial
infarction, stroke, VTE, and critical limb ischemia.
S299: ROLE OF RED BLOOD CELL MEMBRANE-DERIVED PARTICLES ON ENDOTHELIAL DAMAGES DURING
ONSET STAGE OF DELAYED HEMOLYTIC TRANSFUSION REACTION
N.-P. Kim-Anh1 2,*, L. Kiger2, X. Decrouy3, L. Bencheikh1 2, A. Habibi4, F. Pirenne1
2 5, L. Roumenina6, P. Bartolucci2 4 5
1EFS recherche, Etablissement Français du sang; 2Team Pirenne; 3Imaging platform,
INSERM U955 IMRB; 4Sickle cell referral center, Hôpital Henri-Mondor; 5Université
Paris Est Créteil, Créteil; 6INSERM, UMR_S 1138, Centre de Recherche des Cordeliers,
Sorbonne Universités, Université de Paris, Paris, France
Background: Delayed hemolytic transfusion reaction (DHTR) is a life-threatening complication
of red blood cell (RBC) transfusion in sickle cell disease (SCD) patients, leading
to the increase of plasma hemoglobin (Hb) and heme which are responsible for endothelial
damage and organs failure. By the development of an in vitro model reproducing endothelial
damages at the early phase of DHTR (Nguyen et al, ASH 2018), our previous study suggested
that RBC membrane-derived particles released during the onset stage of DHTR could
be also involved in DHTR physiopathology.
Aims: In this study, we aim to determine the mechanism of endothelial damages induced
by these RBC particles.
Methods: Human Umbilical Vein Endothelial Cells (HUVECs) cultured in flow condition
for 24 hours were perfused by either serum only (decomplemented or not) or serum containing
either whole hemolysate (sonicated RBCs) or different hemolysate components or RBCs
membranes (ghosts) under shear stress 1 dyne/cm2. Annexin V and Eculizumab were used
for complement studies.
Endothelial activation and damages were assessed by membrane CD54 (PECAM-1, Platelet
endothelial cell adhesion molecule 1), CD106 (VCAM-1, Vascular Cell Adhesion Molecule-1)
and actin network staining. The whole blood adhesion assays and platelets aggregation
study on hemolysate-preconditioned HUVECs were performed in continuous flow.
Confocal microscopy was used to study interaction between HUVECs and hemoglobin (Hb)
and RBC membrane-derived particles.
Results: Our fluidic model reproduced a hemolysate-induced pro-inflammatory phenotype
and damages of HUVECs via NFkB signaling pathway in a TLR-4 independent manner. The
most deleterious effects were observed in whole hemolysate conditions in which, an
important quantity of RBC large-size particles (> 2 μm) mostly positive for C3 was
detected. These effects on HUVECs were significantly reduced after their elimination
by 14000 g centrifugation (Figure 1). Noteworthy, the suspension of ghosts induced
a similar activation on HUVECs compared to whole hemolysate suggesting the major role
of RBC membrane-derived particles in early hemolysis-induced endothelial damages.
Confocal microscopy for intracellular compartment of hemolysate-treated HUVECs demonstrated
that while the CD235a+ Hb+ particles were only found in whole hemolysate conditions,
the Hb was detected similarly in all conditions tested. Selective blocking of RBCs
particles and Hb endocytosis did not affect hemolysate-induced HUVECs activation.
These observations demonstrate that in the very early stage (≤4H) of intravascular
hemolysis, erythrocytes large-size particles can be internalized in HUVECs, but their
internalization is not necessary for endothelial activation effect.
Complement inactivation of serum or treatment of RBC particles by either Annexin V
or Eculizumab could reduced their effects on endothelial activation suggesting a complement-dependent
mechanism
Image:
Summary/Conclusion: This study suggested for the first time the major role of RBCs
large-size particles on endothelial damages at the onset of DHTR in a complement-dependent
manner justifying the complement blocking treatment in certain DHTR patients. Furthermore,
we have also demonstrated the endocytosis of free Hb by HUVECs, but its pathological
impact is still unclear. This endocytosis would facilitate the Hb clearance during
a massive hemolysis? These new understandings will allow a better and earlier management
to preserve endothelial functions.
S300: HEALTH-RELATED QUALITY OF LIFE IN TRANSPLANT-INELIGIBLE REAL-LIFE MULTIPLE MYELOMA
PATIENTS TREATED WITH BORTEZOMIB-MELPHALAN-PREDNISONE (VMP) VS. LENALIDOMIDE-DEXAMETHASONE
(RD)
M. D’Agostino1,*, S. Bringhen1, R. Ria1, F. Ciceri1, A. P. Falcone1, M. Michieli1,
M. Grasso1, F. Pane1, M. Quaresima1, F. Cattel2, M. Mirabile1, F. Fioritoni1, M. T.
Petrucci1, V. Cotugno2, A. Capra1, S. Pezzatti1, M. L. Mosca Siez1, M. Cantonetti1,
G. Margiotta Casaluci1, P. Bertazzoni1, R. Floris1, M. Offidani1, G. Pietrantuono1,
A. Evangelista3, M. Boccadoro1, A. Larocca1
1European Myeloma Network, (EMN), Italy; 2S.C. Farmacia Ospedaliera, A.O.U. Città
della Salute e della Scienza di Torino, Torino, Italy; 3Unit of Clinical Epidemiology,
A.O.U. Città della Salute e della Scienza di Torino and CPO Piemonte, Torino, Italy
Background: Multiple Myeloma (MM) is a chronic disease, and patients (pts) receive
long-lasting treatment. Thus, the impact on health-related quality of life (HRQoL)
may be burdensome, especially in elderly pts. Bortezomib-melphalan-prednisone (VMP)
and lenalidomide-dexamethasone (Rd) represented standard-of-care treatments for transplant-ineligible
(NTE) newly diagnosed (ND)MM pts before the introduction of daratumumab upfront. No
prospective data are available comparing HRQoL in pts receiving VMP vs Rd in a randomized
fashion.
Aims: We conducted an analysis of Patient Reported Outcomes (PROs) in the context
of a randomized multicenter phase IV trial (Real MM Trial, NCT03829371; funded by
the Italian Medicines Agency AIFA - Independent Research), to compare HRQoL differences
associated with VMP vs Rd treatment in an unselected real-life MM population.
Methods: NTE NDMM pts were randomized to receive 9 VMP cycles vs continuous Rd according
to standard practice. Pts gave written informed consent and were enrolled regardless
of performance status, comorbidities, renal function, or baseline laboratory values.
PROs were collected and analyzed using the validated EORTC QLQ-C30 scales and the
EQ-5D-5L visual analog scale (VAS) instruments. PROs were collected at baseline, every
3 months during the first year, and every 6 months thereafter. The PROs analyses included
pts from the interim analysis (median follow-up: 14.0 months) who had at least a baseline
and a follow-up questionnaire available. Data through month 12 are reported. Change
in HRQoL from baseline in VMP vs Rd pts was analyzed in a linear mixed model adjusted
for International Myeloma Working Group (IMWG) frailty score and cytogenetic risk.
Results are presented as least squares (LS) mean change from baseline.
Results: At the data cut-off (17-12-2021), 104 pts (56 in the VMP arm and 48 in the
Rd arm) had available PROs and were eligible for the analysis. Overall, 46% of pts
had >75 years and 40% were frail. No differences in terms of baseline characteristics
and response rates were found in the VMP vs Rd arms. Mean baseline values of PROs
reflected the deep impairment of pts’ HRQoL at MM diagnosis and were similar in the
two arms. After the start of treatment, the different QLQ-C30 scales were analyzed.
Global Health Status (GHS) was significantly worse with VMP vs Rd at 3 months (-3.3
vs +9.0; P=0.002), while from the 6-month time point onwards no differences can be
found due to an improvement in GHS in the VMP arm (Figure). The physical functioning
scale was worse in the VMP vs Rd arm at 3 (-7.8 vs +0.04; P=0.070) and 6 months (-8.2
vs +3.2; P=0.013), with no differences in later time points. The role functioning
scale behaved similarly (-5.9 vs +5.2 at 3 months with VMP vs Rd; P=0.038). VMP also
increased fatigue (+9.4 vs -2.7; P=0.015), nausea (+8.6 vs +2.8; P=0.04), and appetite
loss (+12.0 vs -3.0; P=0.007) at 3 months, with an improvement in the symptoms thereafter.
In both arms, pain decreased after treatment. No clear differences in the other QLQ-C30
scales were observed. The EQ-5D-5L VAS scale showed a significantly worse score in
the VMP vs Rd arm at 3 months (-7.9 vs +1.7; P=0.005) and at 12 months (-0.6 vs +10.3;
P=0.017).
Image:
Summary/Conclusion: A comparison of PROs in real-life NTE NDMM pts treated with VMP
vs Rd showed a worse HRQoL in the VMP arm early in the treatment course that improved
thereafter. At 3 months, after treatment start, VMP was associated with worse GHS,
physical functioning, role functioning, fatigue, nausea, appetite loss, and EQ-5D-5L
VAS scores.
S301: GERMAN AMLCG-SURVIVORSHIP STUDY: QUALITY OF LIFE AND LIFE SATISFACTION IN AML
LONG-TERM SURVIVORS
E. Telzerow1,*, D. Görlich2, C. Sauerland2, A. S. Moret1, M. Rothenberg-Thurley1,
F. H. A. Mumm1, S. Amler2 3, W. E. Berdel4, B. Wörmann5, U. Krug6, J. Braess7, P.
Heussner8, W. Hiddemann1, K. Spiekermann1, K. H. Metzeler9
1Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University
Hospital, LMU Munich, Munich; 2Institute of Biostatistics and Clinical Research, University
of Münster, Münster; 3Current adress: Friedrich Loeffler-Institut, Federal Research
Institute for Animal Health, Greifswald; 4Department of Medicine A, Hematology and
Oncology, University of Münster, Münster; 5Charité University Hospital Berlin, Berlin;
6Department of Medicine; 3, Hospital Leverkusen, Leverkusen; 7Department of Oncology
and Hematology, Hospital Barmherzige Brüder, Regensburg; 8Departement of Internal
Medicine, Hospital Garmisch-Partenkirchen, Garmisch-Partenkirchen; 9Department of
Medicine; 1, Hematology and Cell Therapy, University Hospital Leipzig, Leipzig, Germany
Background: An increasing proportion of patients with acute myeloid leukemia (AML)
become long-term survivors. Somatic and psycho-social outcomes in survivors are therefore
becoming increasingly important, but little is known about the long-term effects of
the disease and its treatment.
Aims: The primary aim of this study was to compare quality of life (QoL, measured
by the FACT-G questionnaire) and general and health-related life satisfaction (gLS/hLS,
measured by the FLZ-M questionnaire) of AML-LTS with normative data of German adults
who were not diagnosed with AML.
Methods: We designed a comprehensive analysis of AML survivorship outcomes including
psycho-social well-being and somatic health status and conducted a questionnaire-based
study collecting data from AML long term survivors (AML-LTS). This report focuses
on overall and health-related quality of life. Somatic morbidity in AML-LTS is reported
separately (Moret et al.).
Results: 427 former AML patients who had been enrolled in AMLCG trials (AMLCG-1999,
AMLCG-2004, AMLCG-2008) or the AMLCG patient registry, participated in this study
between 5 and 18.6 years (y) after their initial AML diagnosis (median, 11.3y). Median
age of AML-LTS was 61y (range 28y-93y), and 56% were female. Thirty-eight percent
of participants had been treated with chemotherapy alone, while 62% received at least
one allogeneic stem cell transplant (alloHSCT). A relapse occurred in 24% of the participants.
Unexpectedly, age- and sex-normalized quality of life and general life satisfaction
summary scores were significantly higher in AML-LTS (p<.001) compared to adults without
the diagnosis of AML. Raw score points of AML-LTS on the FACT-G summary scale also
were higher than in age and sex-matched normal adults by a median of 4.7 points (95%
CI: 2.82 – 7.2) – a differences that likely is clinically not relevant, considering
an established cutoff for clinical relevance of 7 raw score points. No difference
between AML-LTS and normal adults was found for health-related life satisfaction (hLS).
Using the cutoff for clinical importance (i.e., 7 points below age- and sex-matched
population norm), 26.1% of participants reported relevant impairment of overall QoL.
To identify factors potentially associated with poor overall QoL, we constructed a
logistic regression model including pre-specified cofactors (age, sex, time since
initial diagnosis, relapse and alloHSCT) and additional covariables that associated
with QoL in univariate analyses (Figure 1). We found that participants with no children,
lower educational level, shorter time since diagnosis and altered financial situation
reported significantly lower QoL. No influence was found for disease- and treatment
related factors including treatment (alloHSCT vs. no alloHSCT), previous relapse,
or de novo vs secondary or therapy-related AML.
Image:
Summary/Conclusion: Unlike previous studies of AML survivorship, our large cohort
included a diverse spectrum of patients regarding age, time since diagnosis, and treatment
modalities, which allows for new insight into long-term QoL. Our study establishes
that overall QoL in AML long-term survivors is comparable to the general population,
with further improvement from five years post diagnosis onwards. Importantly, disease-
and treatment-related factors, such as prior relapse or alloHSCT, are not associated
with overall QoL. However, we were able to identify risk factors for worse QoL, delineating
a subgroup of patients that may still have a need for targeted psycho-social interventions
five or more years after an AML diagnosis.
S302: COST-EFFECTIVENESS OF KTE-X19 IN PATIENTS WITH RELAPSED/REFRACTORY MANTLE CELL
LYMPHOMA
M. Marchetti1,*, C. Visco2 3
1Hematology & TMO Unit, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria;
2University of Verona; 3Section of Hematology, Department of Medicine, Azienda Ospedaliera
Universitaria Integrata, Verona, Italy
Background: Relapsed/refractory (R/R) mantle-cell lymphoma (MCL) post BTK-i has a
poor prognosis when treated with currently available treatments with median overall
survival between 6 to 15 months.
In the ZUMA-2 study, KTE-X19, a chimeric antigen receptor (CAR) T-cell therapy, displayed
very high response rate (93% overall objective response rate) with 83% of patients
in the study alive at 12 months (Wang et al 2020). An indirect comparison of overall
survival from ZUMA-2 with patients receiving standard of care (SOC) treatment reported
propensity-adjusted HR 0.32-0.49 (Hess et al 2021). Based on such data, international
scientific societies (NCCN, ESMO) recommend the use of CAR-T for R/R MCL patients
and marketing authorization of KTE-X19 was granted by FDA and EMA.
Aims: The present study aimed to estimate the long-term clinical benefits and overall
healthcare costs of KTE-X19 in R/R MCL patients post BTK-i from the perspective of
the Italian HealthCare System, compared to current standard therapy for these patients
in Italy: rituximab-bendamustine-cytarabine (R-BAC).
Methods: A partitioned-survival model was used to extrapolate overall survival, progression-free
survival, quality-adjusted survival and healthcare costs of R/R MCL patients over
a lifetime horizon, assuming that patients who had not relapsed within 5 years experienced
long-term remission. R-BAC (6 cycles) was used as baseline SOC, however a mix of treatments
was tested as sensitivity analysis. The source of safety and survival data for KTE-X19
was the ZUMA-2 trial (median potential follow up 28.8 mo), while data from retrospective
real-world studies, including SCHOLAR-2, were used for SOC.
The healthcare costs for patients assigned to the CAR-T strategy included bridging
chemotherapy (36.8% of the patients), hospitalization (median length 21.2 days, of
which 23% in intensive care), and the price of KTE-X19. The healthcare costs of patients
assigned to SOC included in addition to R-BAC treatment costs, the cost for allogeneic
hematopoietic stem cell transplant (€179,418) for 31% of patients (McCullogh 2020).
Life years (LYs), quality-adjusted life years (QALYs) and costs were estimated by
discounting future outcomes (3% per year). Uncertainty analysis included one-way and
probabilistic sensitivity
Results: Over a lifetime horizon, mean estimated survival was 8.85 years for KTE-X19
and 1.56 years for R-BAC, while discounted QALYs were 6.40 for KTE-X19 versus 1.20
for R-BAC. Discounted lifetime costs were €411,403 for KTE-X19 versus €74,415 for
R-BAC, which corresponds to a cost of €64,798 per QALY gained and €46,264 per LY gained.
Detailed results are reported in the table below. The ICERs for KTE-X19 versus SCHOLAR-2
based and meta-analysis based comparisons were €57,915, and €56,010, respectively.
The most influential model parameters were patients age and quality of life values,
KTE-X19 acquisition cost, and the proportion of patients receiving allo-SCT in the
comparator arm, and model time horizon.
Probabilistic sensitivity analysis showed that at a willingness-to-pay of €70,000
per QALY, the probability of KTE-X19 to be cost-effective was 65%.
Image:
Summary/Conclusion: This analysis shows that KTE-X19 may be a cost-effective alternative
to R-BAC for patients with RR-MCL pre-exposed to BTK-i and chemoimmunotherapy, from
an Italian third-party payer perspective
S303: EFFECT OF PEGCETACOPLAN ON QUALITY OF LIFE IN COMPLEMENT-INHIBITOR NAÏVE PATIENTS
WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA: RESULTS FROM THE PHASE 3 PRINCE STUDY
D. Gomez-Almaguer1,*, R. Wong2, T. Dumagay3, M. Al-Adhami4, J. Savage4, Z. Hakimi5,
A. Bogdanovic6
1Hematology Service, Hospital Universitario “Dr José Eleuterio González,” Universidad
Autónoma de Nuevo León, Monterrey, Mexico; 2Sir Y.K. Pao Centre for Cancer & Department
of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of
Hong Kong, Sha Tin, Hong Kong; 3Department of Cellular Therapeutics, Makati Medical
Centre, Makati, Philippines; 4Apellis Pharmaceuticals, Inc., Waltham, United States
of America; 5Swedish Orphan Biovitrum AB, Stockholm, Sweden; 6Clinic of Hematology,
Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Belgrade,
Serbia
Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare, life-threatening
disease characterized by complement-mediated hemolysis and thrombosis. Historically,
PNH was treated with C5 inhibitors (eculizumab [ECU)/ravulizumab). Despite their proven
efficacy in the control of intravascular hemolysis (IVH), up to 72% of patients experience
persistent hemolysis leading to suboptimal hemoglobin (Hb) levels and 36% require
transfusions despite ECU treatment which results in a significant impact on the quality
of life (QoL), including persistent fatigue. Pegcetacoplan (PEG) is an FDA/EMA-approved
C3 complement-inhibitor that provides broad hemolysis control (including IVH and extravascular
hemolysis) in patients with PNH.
Aims: This analysis evaluates QoL measures in the Phase 3 PRINCE study (NCT04085601),
a multicenter, randomized, open-label, controlled study evaluating the efficacy and
safety of PEG compared to control treatment (CTRL; excluding complement-inhibitors)
in complement-inhibitor naïve patients with PNH.
Methods: PRINCE has been described previously and was superior for the coprimary endpoints
of hemoglobin (Hb) stabilization (avoidance of >1 g/dL decrease in Hb from baseline),
change from baseline in lactate dehydrogenase at 26 weeks and PEG-treated patients
saw clinically meaningful increases in the FACIT-Fatigue scores (Wong SR, et al. Blood
2021; 138 [Supplement 1]: 606). Secondary endpoints specific to QoL measures included
the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Core
30 Scale (EORTC QLQ-C30), Linear Analog Scale Assessment (LASA) and The Functional
Assessment of Chronic Illness Therapy-Fatigue (FACIT-Fatigue). The EORTC QLQ-C30 contains
30 questions comprising 5 functional, 9 individual symptoms, and one global health
status/QoL item(s), where a ≥10-point change in score is considered clinically meaningful.
The LASA consists of 3 sections asking to rate the perceived level of functioning
(scale 0-100, where a higher score corresponds to a better QoL) and contains specific
domains for activity level, ability to carry out daily activities, and overall QoL.
The FACIT-Fatigue is a 13-item Likert scaled instrument (scale 0-51), where a ≥3-point
increase is considered clinically meaningful.
Results: Relevant baseline characteristics for PRINCE are reported in Table A. Mean
baseline scores of the EORTC QLQ-C30 functional and symptom scales were generally
similar between groups (except for role and cognitive functioning and insomnia; Table
B). After 26 weeks of PEG-treatment, patients displayed clinically meaningful improvements
in most of the functional scales and the global health status/QoL at Week 26 as indicated
by a ≥10-point score increase from baseline (Table B). Improvements in the symptom
scales, including fatigue and dyspnea scales, were also observed in the PEG group
at Week 26 as indicated by a reduction in the symptom score close to the general population
norms (general population norm: fatigue: 24.1, dyspnea: 10.9). Mean total LASA scores
increased in the patients receiving PEG treatment for 26 weeks (Table B). FACIT-Fatigue
scores increased for PEG-treated patients to near the general population norm [43.6],
supporting the gains shown in the EORTC QLQ-C30-Fatigue scale.
Image:
Summary/Conclusion: Substantial and clinically relevant improvements in QoL were consistently
observed with PEG across both EORTC QLQ-C30, total LASA and FACIT-Fatigue scores and
clinically significant improvements in fatigue and dyspnea are highly relevant to
the symptomology of PNH.
S304: SUTIMLIMAB, A COMPLEMENT C1S INHIBITOR, PROVIDES SUSTAINED IMPROVEMENTS IN PATIENT-REPORTED
OUTCOMES IN PATIENTS WITH COLD AGGLUTININ DISEASE (CAD): 2 YEAR FOLLOW-UP FROM THE
CARDINAL STUDY
A. Röth1,*, C. M. Broome2, W. Barcellini3, T. Henrik Anderson Tvedt4, Y. Miyakawa5,
S. D’Sa6, D. Cella7, F. Joly8, J. Wang9, T. Sourdille9, F. Shafer10, M. Wardęcki11,
I. C. Weitz12
1Department of Hematology and Stem Cell Transplantation, West German Cancer Center,
University Hospital Essen, University of Duisburg-Essen, Essen, Germany; 2Division
of Hematology, MedStar Georgetown University Hospital, Washington, DC, United States
of America; 3Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy;
4Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen,
Norway; 5Thrombosis and Hemostasis Center, Saitama Medical University Hospital, Saitama,
Japan; 6UCLH Centre for Waldenström’s Macroglobulinemia and Related Conditions, University
College London Hospitals NHS Foundation Trust, London, United Kingdom; 7Department
of Medical Social Sciences, Center for Patient-Centered Outcomes, Institute for Public
Health and Medicine, Feinberg School of Medicine, Northwestern University, Chicago,
IL, United States of America; 8Sanofi, Chilly-Mazarin, France; 9Sanofi, Cambridge,
MA; 10Sanofi, Bridgewater, NJ, United States of America; 11Sanofi, Warsaw, Poland;
12Keck School of Medicine of USC, Los Angeles, CA, United States of America
Background: CAD is a rare chronic autoimmune hemolytic anemia characterized by classical
complement pathway (CP)-mediated hemolysis, anemia, fatigue, and poor quality of life
(QOL). Sutimlimab is a first-in-class humanized monoclonal antibody that selectively
inhibits C1s of the C1 complex, preventing CP activation, while leaving the alternative
and lectin pathways intact. One-year interim follow-up from the CARDINAL study (NCT03347396)
have previously demonstrated continuous classical CP inhibition with sutimlimab resulted
in rapid, sustained improvements in all patient-reported outcomes (PROs) measures
evaluated.
Aims: To report sutimlimab effect on PROs at 2 years, from the CARDINAL Part B extension.
Methods: CARDINAL was a Phase 3, open-label, single-arm study with a 26-week treatment
period (Part A) and a 2-year extension (Part B) after the last patient finishes Part
A. Sutimlimab was administered through intravenous infusions on Days 0 and 7, followed
by biweekly dosing. PRO data through Week 135, the last data recording within the
2-year Part B time period, are reported here. Efficacy endpoints included hemolytic
markers and the Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-Fatigue)
as a measure of QOL. Exploratory QOL endpoints included mean change from baseline
in EuroQol 5-dimension 5-level (EQ-5D-5L) scores, 12-Item Short Form Health Survey
(SF-12), Patient Global Impression of Change (PGIC) and Patient Global Impression
of Fatigue Severity (PGIS). PRO measures were evaluated every 3 months and conducted
in the following order: FACIT-Fatigue, PGIS, PGIC, SF-12 and EQ-5D-5L. Descriptive
statistics, frequency, or percentage were used to analyze outcomes.
Results: Overall, 24 patients enrolled and 22 finished Part A and entered Part B,
with 19 (86.4%) patients completing Part B. The mean (SD) FACIT-Fatigue score at baseline
was 32 (11) and improved by 7 (8) within 1 week of sutimlimab treatment; the mean
change score from baseline remained ≥5 from Week 1 to Week 135 (minimum score of 38
(9) at Week 123, maximum score of 44 (5) at Week 7), consistent with a clinically
meaningful improvement. Efficacy for the EQ-5D-5L visual analogue scale (VAS) was
sustained over 2 years; the mean (SD) change from baseline in EQ-5D-5L visual analogue
scale (VAS) score (n=15/22) at Week 135 was 17.1 (21.6) (Figure A). The majority of
evaluable patients (n=13/15) indicated an improved disease state compared to baseline
on the PGIC at Week 135. No or mild fatigue was reported in patients (80%, n=12/15)
on completing PGIS at Week 135, compared with one-third at baseline (n=2/6). The mean
increase in SF-12 physical and mental component scores (n=6/22) from baseline to Week
123 were 4.7 (6.9) and 3.8 (14.1) (Figure B), consistent with the clinically important
changes of 3.9 and 2.8 respectively.
Image:
Summary/Conclusion: From this CARDINAL follow-up extension study, sutimlimab has shown
to produce rapid and sustained improvements in FACIT-Fatigue and other PRO measures
evaluated up to 2 years, demonstrating the continued meaningful impact of sutimlimab
on patient QOL.
Posters
P305: COMPREHENSIVE TRANSCRIPTIONAL AND CYTOGENETIC PROFILING IMPROVES CLASSIFICATION
AND DETECTION OF RISK-STRATIFYING MARKERS IN THE B-OTHER PEDIATRIC ACUTE LYMPHOBLASTIC
LEUKEMIA
Ž. Antić1,*, P. Chouvarine1, J. Lentes1, C. Schröder1, J. Alten2, A. Möricke2, M.
Brüggemann3, E. Carrillo-de Santa Pau4, T. Illig1, T. Laguna4, D. M. Schewe2,5, M.
Stanulla6, M. Tang1, M. Zimmermann6, M. Schrappe2, B. Schlegelberger1, G. Cario2,
A. K. Bergmann1
1Institute of Human Genetics, Hannover Medical School, Hannover; 2Berlin-Frankfurt-Münster
ALL Study Group Germany (BFM-G), Department of Pediatrics; 3Department of Hematology,
University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany; 4Computational
Biology Group, Precision Nutrition and Cancer Research Program, IMDEA Food Institute,
Madrid, Spain; 5University Children’s Hospital, Medical Faculty, Otto-von-Guericke-University-Magdeburg,
Magdeburg; 6Pediatric Hematology and Oncology, Hannover Medical School, Hannover,
Germany
Background: B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is the most common
childhood malignancy. Improvements in the genetic-based risk stratification and treatment
adaptations have resulted in an increased overall survival, reaching 90% in the contemporary
treatment protocols. However, in 25-30% of patients, known as B-other, no recurrent
genetic aberrations relevant for the risk stratification and treatment personalization
can be detected using conventional cytogenetic methods. Therefore, identification
and implementation of new risk-stratifying markers in the current treatment protocols
may aid further improvement in the management of children with BCP-ALL.
Aims: Our aim was to investigate the applicability of the integrated use of the whole
transcriptome RNA sequencing and conventional cytogenetic methods in the routine diagnostic
of the B-other BCP-ALL cases.
Methods: We performed RNA sequencing and analyzed gene fusions, expression profiles,
and mutations in diagnostic samples of 174 children with B-other ALL. In order to
further refine genetic classification and validate findings obtained with RNA sequencing,
we integrated our results with the findings obtained using immunophenotyping, FISH,
karyotyping, arrayCGH and Sanger sequencing.
Results: Our analysis unraveled the presence of risk-stratifying fusion transcripts,
in the cases in which these alterations were not detectable using conventional cytogenetic
methods. These included six cases with cryptic KMT2A rearrangements and one of each
with TCF3-PBX1 and TCF3-HLF fusion genes. In addition, we were able to detect 10 cases
with recently described fusions involving ZNF384 gene (ZNF384r), four cases with NUTM1
rearrangements and three cases with fusions involving MEF2D gene. Gene expression-based
clustering unraveled a subset of B-other cases which cluster together with the known
subtypes, indicating the presence of previously described ZNF384r-like, ETV6-RUNX1-like
and BCR-ABL1-like subtypes. Furthermore, we identified 27 previously unassigned B-other
cases co-clustering together with 13 DUX4-positive ALL cases. This finding suggests
that gene expression-based clustering can identify the cases with fusions involving
DUX4 gene, known to be cryptic to most of cytogenetic and NGS approaches.
We further assessed the ability of the analysis pipeline to detect fusion transcripts
in the samples with <50% of tumor blasts. In five tested cases, with BCR-ABL1 or ETV6-RUNX1
fusions, our analysis pipeline was able to detect the presence of fusion transcripts,
with high reliability, even in the samples with down to 7% of tumor blasts. Finally,
we assessed the applicability of using commonly available EDTA tubes and RNA stabilizing
PAXgene tubes for bone marrow sampling, RNA isolation and whole transcriptome sequencing.
We assessed the quality of isolated RNA, library complexity and ability of our analysis
pipeline to detect relevant fusion transcripts. PCA analysis of matched samples stored
in either EDTA or PAXgene tubes showed high similarity in gene expression, while the
correlation between TPM expression values and decay constants indicates that genes
over-abundant in EDTA samples are not dominated by slow-degrading transcripts. These
findings suggest that for short-term storage EDTA tubes are a viable alternative to
the RNA stabilizing PAXgene tubes.
Summary/Conclusion: Taken together, our findings demonstrate the applicability of
whole transcriptome sequencing for personalized diagnostics in pediatric ALL, including
the tentative classification of the B-other cases that are difficult to diagnose using
conventional methods.
P306: PAX5 DEFICIENCY AND GERMLINE SUSCEPTIBILITY TO PEDIATRIC LEUKEMIA
F. Auer1,*, A. Escudero2, M. Sipola3, A. Viitasalo3, M. Morcos1, U. A. Friedrich4,
A. Pandyra5, A. Borkhardt5, M. Takagi6, J. Hauer1
1Department of Pediatrics, Technical University Munich, Munich, Germany; 2Institute
of Medical and Molecular Genetics (INGEMM) La Paz University Hospital, Madrid, Spain;
3Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio,
Finland; 4Pediatric Hematology and Oncology, Department of Pediatrics, University
Hospital “Carl Gustav Carus”, TU Dresden, Dresden; 5Department of Pediatric Oncology,
Hematology and Clinical Immunology, Heinrich-Heine University Duesseldorf, Medical
Faculty, Duesseldorf, Germany; 6Department of Pediatrics and Developmental Biology,
Tokyo Medical and Dental University (TMDU), Tokyo, Japan
Background: Somatic mutations affecting the PAX5 gene are common in pediatric B-cell
acute lymphoblastic leukemia (B-ALL). However, germline PAX5 mutations are extremely
rare. While it is known that PAX5 acts as an indispensable master regulator of proliferation
and differentiation in B-cells, the pre-leukemic setting mediated by reduced PAX5
levels is still poorly understood.
Aims: To investigate the genetic and immunological effect of inherited PAX5 germline
mutations in humans and to characterize the effect of Pax5 heterozygosity on B-cell
development utilizing single-cell RNA Sequencing in mice.
Methods: Aiming to understand the etiology of childhood BCP-ALL in the setting of
reduced Pax5 levels, we used 9-color FACS staining and scRNA/bulk RNA-Sequencing,
as well as whole exome Sequencing technology.
Results: Here, we describe two new families with inherited PAX5 germline variants
affecting amino acid 183 and B-ALL development, including one family with a novel
amino acid substitution p.Gly183Arg. Exome analysis of both families together with
one family harboring PAX5 p.Gly183Ser previously reported by Auer et al., (Leukemia,
2014) yielded no additional common germline variants that could explain the incomplete
penetrance in the observed leukemia development. A study of the respective B-cell
differentiation comparing PBMCs from unrelated non-carriers, healthy carriers, and
patients revealed a significant decrease of intermittent B-cells and memory B-cells
in carriers and patients compared to non-carriers (Student’s T-test; p<0.05 and p<0.01,
respectively). Finally, immunological studies performed using bone marrow samples
from a patient and an unrelated control showed a reduction in the number of mature
B cells, suggesting a block of the B-cell differentiation at the pre-B stage in the
patient.
In line, a comprehensive flow cytometry analysis of the B-cell development in the
bone marrow of healthy Pax5 heterozygous (Pax5
+/-) mice, showed a robust and age independent enrichment of the pre-BII population
(B220+CD19+IgM-CD25+) compared to their Wildtype (WT) littermates (Student’s t-test;
p=0.003). Bulk RNA-Sequencing of sorted preB-II cells (3 WT vs. 3 Pax5
+/- mice) revealed deregulations in major B-cell receptor (BCR) signaling components
including downregulation of Cd79a/b, Lyn, MTor and various ITIMs. Subsequent scRNA-Sequencing
(4 WT vs. 4 Pax5
+/- mice, pool of 20,000 cells hashtag labeled, with 50,000 reads/cell and VDJ-rearrangement
analysis) confirmed a delayed BCR assembly and showed a skewing of light chain rearrangements
towards the lambda isotype in Pax5
+/- mice.
Summary/Conclusion: Overall, our data provide evidence for B-cell dysregulations mediated
by reduced Pax5 levels in the germline of mice and men. Ultimately these characterizations
will help to advance the understanding of molecular mechanisms and susceptible populations
associated with B-ALL. In turn, these findings are important to address the far more
frequent somatic PAX5 mutations with the goal to prevent or treat a significant proportion
of childhood leukemias.
P307: DEL(17)(Q11) IS TYPICAL MARKER OF IMMATURE T-ALL OF ADULTS, WITH NF1, UTP6,
AND SUZ12 HAPLOINSUFFICIENCY, GENOME INSTABILITY, AND GENE DOWNREGULATION
V. Bardelli1,*, V. Pierini1, S. Arniani1, E. Mvridou1, C. Matteucci1, A. G. Lema Fernandez1,
M. Moretti1, L. Elia2, F. Giglio3, F. Forghieri4, M. Cerrano5, N. Fracchiolla6, M.
Delia7, S. Sica8, C. Mecucci1, R. La Starza1
1Hematology and Bone Marrow Transplantation Unit, Department of Medicine and Surgery,
University of Perugia, Perugia; 2Hematology, Department of Translational and Precision
Medicine, Sapienza University, Rome; 3Hematology and Bone Marrow Transplantation Unit,
IRCCS Ospedale San Raffaele, Milan; 4Section of Hematology, Department of Medical
and Surgical Sciences, University of Modena and Reggio Emilia, Modena; 5Department
of Molecular Biotechnology and Health Sciences, Division of Hematology, University
of Turin, Turin; 6Hematology, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico,
Milan; 7Hematology and Stem Cell Transplantation Unit, AOUC Policlinico of Bari, Bari;
8Section of Hematology, Department of Radiological and Hematological Sciences, Catholic
University of the Sacred Heart, Rome, Italy
Background: T-ALLs derive from the accumulation of multiple genetic and epigenetic
events. Some deregulate T-cell transcription factors and define specific genetic subtypes,
while the large majority are involved in pivotal cellular processes, such as signaling,
cell cycle, apoptosis, proliferation, ribosome biogenesis, and epigenetic modulation.
Genomic imbalances, and in particular deletions, are the most frequent cytogenetic
rearrangements in T-ALL. Whether cryptic or large, they often share loss regions -
the common deletion region (CDR) - where they map putative suppressor genes that undergo
haploinsufficiency or inactivation, thus driving the leukemogenic process.
Aims: Our study focused on T-ALL cases harboring interstitial del (17q) with the aim
of assessing the incidence and distribution of this cytogenetic marker, and to detect
associated clinical and molecular features.
Methods: The study was carried out on 378 adult (=223) and pediatric (=155) T-ALL,
including 279 males and 99 females. The genetic background was investigated by integrated
molecular-cytogenetics (FISH and SNPa),1 RNA microarrays (Affymetrix), targeted sequencing
(custom card by Sophia Genetics, Arrow Diagnostics), and RTq-PCR. Data analysis was
carried out referring to available databases (UCSC, NCBI, Data of Genomic Variants,
Biocarta, KEGG, Reactome).
Results: Cases were classified according to genetic rearrangements as HOXA (=104),
TAL/LMO (=74), TLX1/NKX2.1 (=50), TLX3 (=42), BCL11B-a (=8), SPI.1 (=2); 98 were undetermined.
Interstitial monoallelic del(17q) was detected 31/378 cases and was mainly found in
cases with HOXA related abnormalities (8/104) or cases undetermined (17/98). There
were 27 adults and 4 children (p<0.0001). In 19/24 with available flow cytometry the
diagnosis was consistent with ETP/near ETP ALL (p<0.001).
All cases shared a common deleted region (CDR) of about 650kb, that invariably involved
SUZ12, UTP6, and NF1 which were expressed at a significantly lower level than in T-ALL
without del(17q). RNA microarray showed that 148 differentially expressed genes (DEG),
67 up- and 81 down- regulated, distinguished del(17q) T-ALL cases from T-ALL without.
Among up-regulated there were MEF2C, MN1, and IGFBP7, which have been all associated
with an immature phenotype, and MAF and RUNX2 transcription factors. Instead, among
down-regulated, there were putative suppressors involved in genome stability, i.e.
BRCA1, FANCI, BRIP1, XRCC2, and CHEK1. Accordingly, cell cycle, homologous recombination,
DNA replication, and Fanconi anemia pathways, were significantly deregulated in T-ALL
with del(17q), and a significant association with deletions of RB1 (37%)(p<0.001)
and TP53 (20%)(p=0.017) emerged. In addition, del(17q) cases had a higher number (≥5)
of copy number abnormalities (p<0.001). Other recurrent alterations affected JAK/STAT
members and/or modulators (56% of cases).
Summary/Conclusion: Del(17q) was a recurrent marker in immature T-ALL of adults and
characterized 19% of cases undetermined at molecular-cytogenetic level. It caused
the monoallelic loss/haploinsufficiency of three genes, i.e. NF1, UTP6, and SUZ12,
that restricted the CDR. Our study provided evidence that del(17q) identified a major
leukemogenic mechanism, through the cooperation of: 17q genes loss of function, JAK/STAT
constitutive activation, and altered cell cycle. Interestingly, RUNX2 and IGFBP7,
high-risk markers in T-ALL,2,3 were aberrantly expressed in this subset of T-ALL.
1La Starza R, et al. J Mol Diagn 2020
2Matthijssens F, et al. JCI 2021
3Bartram I, et. al., BMC Cancer 2015
P308: NOVEL COPY NUMBER ABERRATION-BASED CLASSIFICATION METHODS REFINE RISK ASSESSMENT
IN PEDIATRIC B-CELL PRECURSOR ACUTE LYMPHOBLASTIC LEUKEMIA
G. Bedics1,*, B. Egyed2, L. Kotmayer1, A. Benard-Slagter3, K. de Groot3, A. Bekő1,
L. L. Hegyi1, S. Krizsán1, G. Kriván4, D. J. Erdélyi2, G. Kovács2, B. Kajtár5, L.
Pajor5, Á. Vojcek6, G. Ottóffy6, A. Ujfalusi7, C. Kiss8, I. Szegedi8, K. Bartyik9,
G. Péter10, E. Sebestyén1, Z. Jakab11, A. Matolcsy1, S. Savola3, C. Bödör1, D. Alpár1
1HCEMM-SU Molecular Oncohematology Research Group, Department of Pathology and Experimental
Cancer Research; 22nd Department of Pediatrics, Semmelweis University, Budapest, Hungary;
3Department of Oncogenetics, MRC Holland, Amsterdam, Netherlands; 4Central Hospital
of Southern Pest - National Institute of Hematology and Infectious Diseases, Budapest;
5Department of Pathology; 6Department of Pediatrics, University of Pécs Clinical Centre,
Pécs; 7Department of Laboratory Medicine; 8Department of Pediatric Hematology-Oncology,
Institute of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen; 9Department
of Paediatrics and Paediatric Health Care Center, Faculty of Medicine, University
of Szeged, Szeged; 10Hemato-Oncology Unit, Heim Pál Children’s Hospital; 11Hungarian
Childhood Cancer Registry, Hungarian Pediatric Oncology Network, Budapest, Hungary
Background: The genomic landscape of pediatric acute lymphoblastic leukemia (ALL)
is heterogeneous with distinct copy number aberrations (CNAs) being detectable in
vast majority of the patients. A subset of these alterations provides prognostic and/or
predictive information; therefore, various CNA-based patient classifiers were introduced
in the past.
Aims: We applied a next-generation sequencing (NGS) based method to comprehensively
screen for recurrent, disease-relevant CNAs in a cohort of Hungarian patients, allowing
us to establish novel patient risk stratification approaches.
Methods: Diagnostic bone marrow samples from 261 children with B-ALL and 22 matching
samples drawn from 21 patients at first or second relapse were investigated by digital
multiplex ligation-dependent probe amplification (digitalMLPA) using the ALL-specific
D007 probemix. DigitalMLPA libraries were sequenced on an NGS platform. Whole chromosome
gains and losses, as well as subchromosomal CNAs were simultaneously profiled. Survival
rates were estimated using the Kaplan-Meier method and compared by log-rank tests
in R version 4.1.2.
Results: In total, 1,400 CNAs including numerical chromosomal aberrations and subchromosomal
CNAs were detected in 93.5% of the diagnostic samples. On average, 5.36 CNAs were
observed per patient with a mean of 2.45 subchromosomal alterations. Subtype-defining
aberrations were identified in 36.0% of the patients with hyperdiploidy and iAMP21
detected in 32.2% and 3.8% of the cases, respectively. Numerous CNAs in disease-relevant
genes responsible for cell cycle control, lymphoid development, signaling, or tumor
suppression were identified. Considering all affected exons and an upstream region,
10 distinct patterns of IKZF1 deletion were observed. Unbiased co-segregation analysis
revealed 15 positive and 3 negative correlations between the various CNAs, e.g. common
co-occurrence of iAMP21 and CDKN2A/B deletion, and negative correlation between ETV6
and BTG1 deletions. Comparative analysis of diagnostic and matching relapse samples
revealed characteristic temporal changes of copy number profiles with additional aberrations
(65%), as well as both emerging and disappearing CNAs (30%) detected at relapse as
compared with diagnosis. One patient did not show CNAs in the analyzed samples (5%).
Prognostic subgroups determined based on cytogenetic findings were combined (i) with
IKZF1del/IKZF1plus status and (ii) with CNA subgroups defined by a comprehensive digitalMLPA
profiling. The combined genetic classification methods identified 3 and 4 patient
subgroups, respectively, with significantly different 5-year progression-free survival
rates.
Summary/Conclusion: Comprehensive and highly optimized CNA profiling with digitalMLPA
revealed subtype-defining gross-chromosomal changes and additional disease-relevant
subchromosomal CNAs in a large cohort of Hungarian patients treated with ALL IC BFM
protocols. Comparative scrutiny of matching diagnostic and relapse samples from 21
patients unveiled two different patterns of temporal evolution. Two novel risk stratification
approaches have been established by combining cytogenetic data with digitalMLPA-based
copy number profiling, with one of those laying more emphasis on CNA data than seen
in previously introduced classifications, and the other one combining cytogenetic
data with IKAROS status for the first time. DigitalMLPA offers a fast, reliable and
standardizable DNA copy number analysis which is easily implementable in the diagnostic
workflow of pediatric ALL.
P309: IMAGE-BASED HIGH-CONTENT DRUG SENSITIVITY SCREENING IDENTIFIES CONVENTIONAL,
EMERGING, AND POTENTIAL NOVEL THERAPEUTIC TARGETS IN HIGH-RISK ADULT ACUTE LYMPHOBLASTIC
LEUKEMIA
H. Bell1,*, M. Singh1, H. Blair1, A. Poll1, E. Law1, O. Heidenreich2, F. van Delft1,
A. Moorman1, J. Lunec3, J. Irving1
1Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne,
United Kingdom; 2Prinses Maxima Centrum, Utrecht, Netherlands; 3Paul O’Gorman Building,
Newcastle University, Newcastle upon Tyne, United Kingdom
Background: Despite concerted efforts to optimize and intensify treatment approaches
in adult acute lymphoblastic leukemia (ALL) more than half of patients still succumb
to their disease within 5 years of diagnosis, emphasizing the limits of current therapy
and the critical requirement for more effective and safer therapeutics. Patient-derived
xenograft (PDX) ALL models represent reliably predictive preclinical models and allow
in vivo amplification of patient material in quantities sufficient for high-throughput
screening applications.
Aims: To identify novel and effective therapies for high-risk ALL using an in vitro
co-culture model of PDX ALL blasts and mesenchymal stem cells (MSCs) by high-content
screening of >1200 pharmacologically-active small-molecule drug compounds in various
stages of clinical development.
Methods: PDX leukemic cells derived from seven patients with high-risk ALL, including
relapsed Philadelphia-positive B-ALL, low hypodiploid B-ALL, and ABL-class T-ALL,
were used for the drug screen. ALL cells were co-cultured with hTERT-immortalized
MSCs to support survival of the ALL blasts during a 96 hour short-term culture with
the drug library compounds (final concentration, 1mM) in 384-well format. Drug sensitivity
was assessed using an automated, high-throughput, fluorescence image-based microscopy
pipeline which identifies and discriminates live ALL cells and MSCs using random forest
machine learning algorithms based on cellular nuclear staining.
Results: Drug sensitivity profiles revealed a wide diversity of responses to the library
compounds reflected by different “hit” rates (4.5-7.4%), where “hit” is defined as
compounds reducing cell survival by more than 50%. Despite this heterogeneity in response,
59 unique compounds were identified with broad anti-leukemic activity; that is, “hits”
in 4/7 PDX samples. Concurrent image analysis of MSCs in the co-culture system revealed
most hit compounds did not affect MSC numbers relative to vehicle-treated controls
(n=50/59), indicating a possible therapeutic window for targeting leukemic cells specifically.
Several hit compounds (n=14) were drugs or from drug classes which form part of conventional
ALL treatment regimens — including anthracyclines, glucocorticoids, and nucleoside
analogues — validating the screening approach for successful identification of anti-leukemic
compounds. Drug classes with recently recognized and emerging preclinical anti-leukemic
activity were also identified including cyclin-dependent kinase (CDK) inhibitors (n=5),
cell cycle-related kinase inhibitors (n=6), bromodomain and extra-terminal domain
(BET) inhibitors (n=4), histone deacetylase family inhibitors (n=3), and nicotinamide
phosphoribosyltransferase (NAMPT) inhibitors (n=3). Nine hit compounds elicited significant
reductions in cell survival consistently across the panel of PDX samples (7/7), providing
some evidence towards anti-leukemic activity independent of cytogenetic background.
These included pan-CDK family inhibitors alvocidib, CGP60474, and roscovitine-derived
CR8. Additionally, the selective CDK (-1, -2, -9) inhibitor AZD 5438 exhibited anti-leukemic
activity in all samples (n=6/7), bar the Philadelphia-positive sample.
Summary/Conclusion: We provide evidence that high-content drug screening in a co-culture
system is an effective strategy to identify clinically-relevant novel and emerging
therapeutic targets in high-risk ALL. Our analyses promote a strong rationale for
exploring the targeting of CDKs as a potential pan-effective approach in high-risk
adult ALL.
P310: BH3 PROFILING IDENTIFIES SELECTIVE BCL-2 DEPENDENCE OF ADULT EARLY T-CCELL PROGENITOR
AND TYPICAL ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS
K. Bhatia1, A. Mahesh1, E. Olesinski2, J. Garcia2, N. Jain3, W. Y. Jen4, M. Ooi Gaik
Ming5, A. Letai2, M. Konopleva3, S. Bhatt1,*
1Department of Pharmacy, National University of Singapore, Singapore, Singapore; 2Department
of Medical Oncology, Dana-Farber Cancer Institute, MA; 3Department of Leukemia, University
of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America; 4Department
of Haematology-Oncology, National University of Singapore; 5Department of Medicine,
Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
Background: T-cell acute lymphoblastic leukemia (ALL) results from malignant transformation
of immature T-cells, accounting for 10-15% of childhood and 20-25% of adult ALL cases.
Although long-term survival rates for standard-risk childhood T-cell ALL (T-ALL) have
shown striking improvements to 90%, response outcomes in adults remain much lower.
Among T-ALL, a high-risk subtype originating from clonal expansion of recently immigrated
thymocytes, defined as early T-cell precursor (ETP) leukemias, has a significantly
worse outcome in adults.
Aims: BH3 mimetics, small molecule antagonists of anti-apoptotic BCL-2 family proteins,
have shown remarkable clinical success in the treatment of hematological malignancies.
We previously showed that pediatric ETP-ALL, is dependent on BCL-2, while typical
T-ALL is dependent on BCL-XL. These findings importantly led to the clinical trial
of venetoclax in combination with hyperCVAD in relapsed/refractory (R/R) T-ALL patients.
However, whether adult patients with T-ALL also show the selective pattern of anti-apoptotic
dependence related to the differentiation stage of T-cell has not been studied.
Methods: BH3 profiling, and cell cytotoxicity assay.
Results: We investigated the survival dependencies of adult ETP-ALL and T-ALL on BCL-2
family proteins using 42 primary samples collected from 3 centers (Dana-Farber, MD
Anderson, and National Cancer Institute Singapore). All 15 ETP-ALL samples showed
robust mitochondrial depolarization response to the BAD peptide as compared to the
HRK peptide and DMSO control using BH3 profiling assay, suggesting primary dependence
on pro-survival BCL-2 protein. We next performed cell death assays by exposing ETP-ALL
primary tumors to short-term treatment with venetoclax and navitoclax and found that
consistent with BCL-2 dependence, adult ETP-ALL can be efficiently targeted for apoptosis
via venetoclax. We then asked if there is a difference in dependencies between adult
ETP-ALL and T-ALL. In ETP-ALL, cytochrome c release was caused by the BAD peptide
alone, while in T-ALL, both BAD and HRK peptides induced cytochrome c release. This
suggests that ETP-ALL primary tumors are primarily BCL-2 dependent, while T-ALL primary
tumors are co-dependent on BCL-2 and BCL-XL. Further, this dependence endorses mechanistic
efficacy of venetoclax and navitoclax, where venetoclax serves to antagonize BCL-2,
and navitoclax serves to antagonize BCL-XL and to a lesser extent BCL-2, through BIM
displacement.
Image:
Summary/Conclusion: We hereby validated that BH3 profiling predicted remarkable on-target
cytotoxicity of venetoclax in adult ETP-ALL and T-ALL. In addition, some T-ALL with
enriched HRK signaling and BCL-XL dependence revealed that these patients can benefit
from navitoclax. These findings provide pre-clinical evidence for venetoclax and navitoclax
as a potentially efficacious combination therapy of BH3 mimetics for adults with T-ALL.
P311: IMBALANCES IN HOMOCYSTEINE METABOLISM AND LEUKEMIA
N. Bouayed Abdelmoula1,*
1Genomics of Signalopathies at the service of Medicine, Medical University of Sfax,
Sfax, Tunisia
Background: The correlation between imbalances in homocysteine metabolism and leukaemia
is a matter of debate. While the effects of polymorphisms of key homocysteine metabolism
enzymes on homocysteinemia are confirmed, their direct causality in the genesis of
leukaemias or their role as disease markers (diagnostic or prognostic) remains controversial.
It has been reported in several studies that the C677T and A1298C polymorphisms of
the MTHFR may play a role in the pathophysiology of acute leukaemias and various other
types of signalopathies such as cancers. Homocysteine could indeed modulate the expression
of certain tumour suppressor genes and induce epigenetic alterations.
Aims: In this work, the aim was to identify this correlation using a molecular study
of MTHFR polymorphisms in a Tunisian cohort with acute lymphoblastic leukemia.
Methods: The molecular study involved a cohort of 41 Tunisian children with acute
lymphoblastic leukemia. Genomic DNA was extracted from blood samples. Amplification
of the regions of interest of the MTHFR gene at exons 4 and 7 was performed by PCR
using primers designed in silico. Genotyping of the 677C>T and 1298 A>C polymorphisms
was performed by PCR-RFLP of the amplicons using two restriction enzymes HinfI and
MboII.
Results: MTHFR polymorphisms were detected in 39% of the children. The frequency of
polymorphic alleles corresponded to 15.85% for the A1298C polymorphism and 4.87% for
the C677T polymorphism. In fact, The C677T and A1298C MTHFR polymorphisms were detected
in respectively 4 and 11 patients as heterozygous. Only one patient harbored homozygous
A1298C MTHFR polymorphism. Only two cases were heterozygotes for both polymorphisms.
Summary/Conclusion: Imbalances in MTHFR homocysteine metabolism associated with the
polymorphisms do not appear to be a risk factor for ALL in the study cohort.
P312: EMPLOYING WHOLE GENOME OPTICAL MAPPING TO CHARACTERIZE THE LANDSCAPE OF STRUCTURAL
VARIANTS IN B-CELL PRECURSOR ACUTE LYMPHOBLASTIC LEUKEMIA
D. Brandes1,2,*, T. Brozou1, S. Soura1, A. Bergmann3, U. Fischer1,4, A. Borkhardt1,4,
R. Wagener1,4
1Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University
Düsseldorf, Germany and University Hospital Duesseldorf; 2Duesseldorf School of Oncology
(DSO), Duesseldorf; 3Institute of Human Genetics, Hannover Medical School (MHH), Hannover;
4German Cancer Consortium (DKTK), partner site Essen/Duesseldorf, Duesseldorf, Germany
Background: B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is the most frequent
pediatric cancer. The most common subtypes are ETV6::RUNX1+ BCP-ALL, which harbor
a reciprocal translocation between chromosome 12 and 21 leading to a juxtaposition
of ETV6 to RUNX1, and high hyperdiploid (HHD) BCP-ALL harboring 51-67 chromosomes.
In large-scale genomic studies, the mutational landscape of somatic single nucleotide
variants and indels as well as copy number gains and losses based on array CGH have
been well described in both subtypes. However, a comprehensive analysis of somatic
structural variants (SVs) is still lacking which may reveal significant new insights
on tumor development, progression, and treatment options in BCP-ALL.
Aims: We aimed to primarily employ the novel technology whole genome optical mapping
(WGOM) to unmask the landscape of structural variants in BCP-ALL. Firstly, we determined
the concordance of chromosomal abnormalities identified by WGOM in comparison to standard
techniques. Superiorly, we explored the landscape of somatic aberrations in ETV6::RUNX1+
and HHD tumors to identify recurrent alterations and novel SVs affecting genes not
yet related to BCP-ALL pathogenesis.
Methods: We extracted ultra-high molecular weight (UHMW) DNA of 13 ETV6::RUNX1+ and
18 HHD BCP-ALL with a tumor cell content of 25-96% at diagnosis. Additionally, we
isolated DNA of fibroblasts or remission material of each pediatric patient as matching
non-tumor control. Informed consent was obtained from all participants. DNA was fluorescently
labeled by a sequence specific enzyme and subsequently linearized and imaged using
a Saphyr instrument (Bionano Genomics). Using this approach, we generated comprehensive
datasets of 31 matched tumor/non-tumor pairs, allowing the identification of somatically
acquired SVs >500bp. In addition, corresponding karyotype, fluorescence in situ hybridization
and SNP-array data of the leukemia samples were integrated and compared to WGOM data.
Results: We validated 95% of 233 events observed by standard techniques with WGOM.
All hallmark alterations including ETV6::RUNX1 fusion and high hyperdiploidy were
identified by WGOM. In addition, we detected a median of 11 (1-36) somatically acquired
events in leukemic cells with ETV6::RUNX1 fusion, compared to 3 (1-10) events in HHD
cases. WGOM identified recurrent somatic structural aberrations occurring in at least
three BCP-ALL cases, including focal deletions affecting ETV6 (9/31), PAX5 (5/31),
ARPP21 (3/31), CD200/BTLA (3/31) and RAG2 (3/31). We detected subtype specific recurrent
focal deletions disrupting ATF7IP (7/13) and BTG1 (5/13) in ETV6::RUNX1+ subclones
whereas deletion of the CDKN2A/B locus (3/18) was only observed in HHD BCP-ALL. Interestingly,
we discovered novel recurrently deleted regions on 12q24.11 (GPN3, 4/31) and Xq25
(STAG2, 3/31) in all BCP-ALL cases as well as focal loss of 19q13.11 (UBA2, 3/13)
specific for ETV6::RUNX1+ subclones using WGOM. Moreover, complex three-way translocations
in ETV6::RUNX1+ tumors were discovered by WGOM, including t(6;8;12)(p12.3;q24.23;q24.21)
that leads to the disruption of MED13L and HMGCLL1.
Summary/Conclusion: Our study showed that WGOM detects SVs as translocations and aneuploidies
in BCP-ALL as good as conventional methods with a 95% concordance. However, WGOM allows
a higher resolution and more precise localization of chromosomal breakpoints compared
to standard techniques. Hence, we identified novel recurrent somatically acquired
SVs including a deletion on Xq25 affecting STAG2, which might play an important role
in pediatric BCP-ALL leukemogenesis.
P313: RECURRENT PATHOGENIC GERMLINE CHEK2 VARIANTS IN PEDIATRIC PATIENTS WITH HEMATOLOGICAL
MALIGNANCIES
T. Brozou1,*, R. Wagener1, U. Fischer1, A. Borkhardt1
1Department of Pediatric Hematology, Oncology and Clinical Immunology, Uniklinik Dusseldorf,
DUSSELDORF, Germany
Background:
CHEK2 is a tumor-suppressor gene encoding CHEK2 kinase that plays a crucial role in
DNA repair and cell cycle arrest as part of the ATM-CHEK2-p53 pathway. Predisposing
germline variants in CHEK2 are associated with breast cancer and other adult-type
malignancies such as prostate, gastric and thyroid cancer. Only few studies have examined
the occurrence of germline CHEK2 variants in adults with leukemia, but the association
between inherited CHEK2 variants and hematological malignancies in childhood has not
been studied.
Aims: Our aim is to understand the frequency and the impact of germline variants in
the cancer predisposing CHEK2 gene in pediatric hematological malignancies.
Methods: We analyzed parent-child WES data of 150 children (≤18yrs) with hematological
malignancies (n=107 leukemia, n=43 lymphoma) and mined the data for CHEK2 variants.
We performed functional analysis of the identified pathogenic variants to analyze
the effect of the alteration on protein function.
Results: We detected in 15/150 children with hematological malignancies a CHEK2 germline
variant (Figure 1A). The majority of those patients had a leukemia (n=11 BCP-ALL,
n=1 T-ALL, n=1 CML, n=2 Hodgkin Lymphoma). Overall, eight different CHEK2 variants
including three truncating variants were detected. Three variants were recurrent (p.Ile157Thr
n=5, p.Ile160Met n=3, p.Thr367Metfs*15 n=2). According to the classification of the
American College of Medical Genetics and Genomics 7/8 variants were predicted to be
pathogenic or likely pathogenic. For two of these variants an impaired function had
been already described in the literature. To test the functional consequences of the
five remaining variants, we overexpressed these, in comparison to two control CHEK2
variants, in U2OS cells. Four out of seven variants (n=1 truncating, n=1 in-frame
deletion, n=2 missense) led to loss of protein expression (Figure 1B) and loss of
CHEK2-Thr68 phosphorylation upon irradiation induced DNA damage. Five out of fifteen
patients carried a CHEK2 variant impairing its function. Interestingly, 4/5 patients
with a pathogenic variant had a BCP-ALL and 1/5 had a CML that rapidly progressed
to CD10+, CD19+ lymphoid blast crisis while under imatinib therapy (400 mg/m2).
Figure 1: (A) Schematic representation of the CHEK2 protein indicating the localization
of the identified variants. The length of the bars correlates to the number of patients
with the respective CHEK2 variant and the color of the bars to the variant type (green:
missense, red: frameshift indels, orange: splicing). (B) Western blot analysis of
HA-tagged CHEK2 protein variants ectopically expressed in U2OS cells. GAPDH was used
as a loading control. CHEK2 variants Arg145Trp and Glu161 del were included as controls
in our experiments. (C) Bar blot depicting the quantification of the CHEK2 variant
protein expression analyzed by Western blotting of three independent experiments.
Each experiment is represent by a dot, the bar indicates the mean of all three experiments.
Image:
Summary/Conclusion: We identified heterozygous CHEK2 variants in the germline of 10%
of children with hematological malignancies by WES-based screening of 150 cases. Expression
studies revealed that two of the variants identified in our cohort were associated
with reduction/loss of CHEK2 expression. We conclude that reduced CHEK2 expression/phosphorylation
upon DNA damage may be an important germline-encoded host factor predisposing to the
development of BCP-ALL in children.
P314: THERAPEUTICALLY TARGETING THE UNIQUE BARCODE OF MLL/AF4
R. Cameron1,*, L. Swart1, M. Rasouli1, O. Heidenreich1
1Prinses Maxima Centrum, Utrecht, Netherlands
Background: Patients harbouring MLL (KMT2A)-rearrangements with AF4 remain a high-risk
subgroup in acute lymphoblastic leukaemia cases, with currently <40% event free survival
in infants (<1 year). Occurring in >70% of infant cases, improved therapies are essential.
The presence of the MLL/AF4 is vital for transformation and maintenance of the leukaemia,
making it an ideal therapeutic target. Small interfering RNAs (siRNA) can silence
disease driving genes. Fusion genes such as MLL/AF4 present with a unique sequence
across the breakpoint that is only present in the cancer cells, potentially allowing
for an on-target. We therefore designed an siRNA targeting the fusion site within
MLL/AF4. Since siRNA alone has been shown to have poor pharmacokinetic (PK) and pharmacodynamic
(PD) properties, we aimed to improve efficacy chemically modifying siRNA and encapsulating
them in lipid nanoparticles.
Aims: The aim of this study is to develop a lipid nanoparticle delivery system to
encapsulate a therapeutic siRNA targeting the MLL/AF4 breakpoint and explore the impact
of this in t(4;11) cell lines.
Methods: Cells expressing the MLL/AF4 fusion were treated with an MLL/AF4 (siMA6)
or control (siMM) siRNA through electroporation or encapsulation by various LNP formulations.
Cellular uptake and on target efficiency were investigated in tissue culture using
the t(4;11)-positive cell line SEM.
Results:: Electroporations were used to optimise chemically modified siRNA for improving
PK and PD of siMA6, this data suggested that >200 nM was required to achieve 60% knockdown
(KD) within 24 hours. Following this we tested to reduced concentrations of siRNA
to investigate if LNPs improve delivery and efficacy. Cells were given 2ug/ml of siMA6
encapsulated in either cholesterol and PEG rich LNP (LNP-chol) or sphingomyelin rich
LNP (LNP-SM) and compared. Uptake of LNP-SM is slower compared to LNP-chol within
the first 3 hours, 60% and 90% retrospectively. However, by 6 hours, only a 5% difference
with LNP-SM achieving 90% compared to 95% in LNP-chol. Surprisingly, functional studies
suggest that consequential effects are enhanced in LNP-SM. Sequentially dosed cells
with LNP-chol shows a knockdown ranging from 25%-65% in both MLL/AF4 and downstream
target genes when compared to the control over 9 days of sequential dosing. A 10-15%
reduction in proliferation and viability was seen at day 9. When treated with LNP-SM
these consequences were enhanced, KD of MLL/AF4 is reduced 40-60% and downstream genes
such as PROM1 and ANGPT1 to >70% by day 9. Furthermore, proliferation of the cells
is impaired, reflected in substantial decrease in viable cells by 1.5-fold and notable
increase was observed in the G1 and subg1 populations of the cells cycle. Surprisingly,
both LNP-chol and LNP SM exhibited a strong impact on self-renewal with a reduction
of ≥50% and ≥60% retrospectively by day 9.
Summary/Conclusion: This study has demonstrated the advantage of targeting fusion
genes with a therapeutic siRNA. We further prove the impact of using an LNP delivery
system to improve therapeutic potential of siRNA. This allowed use to more clinically
relevant concentrations to achieve a 60% of MLL/AF4 and downstream target genes. Notably,
using an LNP-SM formulation enhanced activity of siMA6, showing a substantial impact
on cell proliferation, viability, and capacity to self-renew. The next important step
of this project is to test PK properties in-vivo and validate the promising findings
observed in-vitro. These encouraging results could lead to improved treatments for
these ALL patients with a clinically poor prognosis
P316: THE ROLE OF TET2 IN (PRE)-LEUKEMIC T-ALL: TET2 TO THE RESCUE?
S. De Coninck1,2,3,*, P. Van Vlierberghe1,2,3, S. Goossens2,4, B. Lintermans1,2,3,
J. Roels1,2,3
1Biomolecular Medicine; 2Cancer Research Institute Ghent (CRIG); 3Center for Medical
Genetics; 4Diagnostic Sciences, Ghent University, Ghent, Belgium
Background: T-cell acute lymphoblastic leukemia (T-ALL) accounts for 15% of pediatric
and 25% of adult ALL patients. Therapy has been intensified over the last decades,
leading to gradual improvements in survival. However, 10-15% of pediatric and 50%
of adult T-ALL cases relapse and ultimately die because of refractory disease.
During malignant transformation of T-cells, a clonal expansion of immature thymic
cells is selected through the gradual accumulation of advantageous epigenetic changes
and genetic mutations. Long-lived preleukemic stem cells (pre-LSCs) were uncovered
as an initiating event in various blood-born cancers, including in mouse models of
T-ALL.
Advanced next-generation sequencing technology allowed for the identification of these
predisposing mutations in preleukemic myelodysplastic syndrome patients before they
progress towards acute myeloid leukemia. Substantial enrichment for mutations in regulators
of DNA methylation were identified, including loss of TET2. TET2 is an enzyme that
catalyzes DNA demethylation and Tet2 knockouts are prone to develop lymphoid malignancies,
including T-cell lymphoblastic leukemia/lymphoma. In T-ALL, the role of TET2 remains
to be investigated. We hypothesize that TET2 acts as a tumor suppressor in T-ALL,
involved in a premalignant clonally expanded pre-LSC population.
Aims: We aimed to evaluate the tumor suppressor role of TET2 in the development of
preleukemic stem cells that ultimately give rise to T-ALL, and to gain a better understanding
of the molecular mechanisms involved in this process of malignant T cell transformation.
Methods: We evaluated the expression of TET2 in RNAseq data of primary T-ALL samples
and compared this with their normal T cell counterparts. Next, we used a spontaneous
T-ALL mouse model, CD2-LMO2tg mice. Already at young age, preleukemic CD2-Lmo2tg thymocytes
gain aberrant self-renewal capacity, which allows clonal expansion of an immature
thymocyte precursor subpopulation. This T-ALL mouse model was crossed with a R26-Tet2
knock-in mouse model that allows conditional (CD2-icre) Tet2 expression in all lymphocytes,
at an early differentiation stage, before T-cell commitment.
Results: In T-ALL, TET2 mutations are sporadically identified. However, gene expression
profiling of primary T-ALL samples identified a large subset of T-ALLs characterized
by a profound loss of TET2 expression compared to healthy T cell precursors (Fig.
1A). These low TET2 levels occur in patients from different T-ALL subtypes and were
confirmed in a subset of T-ALL cell lines.
To further explore the relevance of TET2 silencing and its potential implications
in clonal expansion, preleukemic CD2-Lmo2tg mice were sacrificed at different time
points (8 and 24 weeks). Gene expression levels in premalignant T-cells were compared
with age matched wild-type control mice. A gradual decrease of Tet2 expression, with
a co-occurrent increase of Tet2 promotor methylation, was detected in preleukemic
CD2-Lmo2tg mice (Fig. 1B-C), which confirms the important role of Tet2 in pre-LSC.
To support the tumor suppressor role of Tet2 in T-ALL, an aging cohort of CD2-Lmo2tg
with and without Tet2 overexpression was built. Survival analysis showed that CD2-LMO2tg
mice with Tet2 overexpression have an increased latency of T-ALL (Fig. 1D, p=0,03).
Image:
Summary/Conclusion: Tet2 plays an important role in pre-LSC and can act as a tumor
suppressor in a spontaneous T-ALL mouse model, which supports the possible tumor suppressor
role of TET2 in a subgroup of T-ALL patients. Loss of TET2 in these patients might
be a good predictor for response to DNA methylation inhibitors.
P317: TARGETING HYPERACTIVE PDGFR-B IN T-ALL AND T-LBL
S. De Coninck1,2,3,*, P. Van Vlierberghe1,2,3, S. Goossens2,4, R. De Smedt1,2,3, B.
Lintermans1,2,3
1Biomolecular Medicine; 2Cancer Research Institute Ghent (CRIG); 3Center for Medical
Genetics; 4Diagnostic Sciences, Ghent University, Ghent, Belgium
Background: T cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic
malignancy that accounts for 10-15% of pediatric and 25% of adult ALL cases. General
prognosis of T-ALL has been improving over time. However, the outcome of T-ALL patients
with primary resistant or relapsed leukemia remains dismal.
To this end, we have screened a T-ALL/T-LBL patient cohort for copy number variations
and identified a novel MYH9-PDGFR-β fusion in a T-LBL case, driving aberrant activation
of platelet derived growth factor β (PDGFR-β). Moreover, RNA sequencing data of an
independent T-ALL patient cohort showed overexpression of PDGFR-β in the TLX subgroup
and in immature T-ALL patients.
PDGFR-β is a transmembrane glycoprotein dimer molecule that functions as a receptor
tyrosine kinase. Ligand binding results in receptor dimerization and subsequent activation
via autophosphorylation, causing downstream activation of various signaling pathways
such as PI3K/AKT, MAPK/ERK, JAK/STAT and Notch.
As constitutively active PDGFR-β is observed in a variety of malignancies, multi-target
kinase inhibitors are already used in the clinic. However, they are not specific and
often result in adverse side-effects.
CP-673451 is a novel and selective PDGFR inhibitor. Mechanistically, CP-673451 inhibits
autophosphorylation of dimeric PDGFR-β and in this way prevents phosphorylation of
downstream PI3K/AKT and GSK-3α/β. Previous studies showed growth inhibition of multiple
tumor xenografts (lung and colon carcinomas) mainly due to a direct antitumor effect.
Aims: We aim to assess whether specific PDGFR-β inhibitors can be used as a novel
and targeted therapy in T-ALL/T-LBL treatment or if they can be of added value to
the current chemotherapy regimen for T-ALL/T-LBL patients. In the future, we would
like to investigate the transforming capacity of the MYH9-PDFR-β fusion in an in vitro
pro-T cell culture system.
Methods: T-ALL cell lines were screened for PDGFR-β levels and cell lines were selected
with both high (CTV-1, SEM) and low (MOLT-16, Jurkat, Loucy) PDGFR-β levels. CP-673451
was tested on the selected cell lines. Cell viability was measured using an ATP assay
(CellTiter Glo) and downstream signaling pathways were quantified with flow cytometry
and western blot. Additionally, spleen cells from PDGFR-βhigh and PDGFR-βlow T-ALL
PDX models were treated ex vivo using CP-673451.
Results: Both PDGFR-βhigh cell lines and PDGFR-βhigh PDX models showed sensitivity
to CP-673451 treatment in nanomolar range, whereas PDGFR-βlow models did not (Fig.
1A-B). Further characterization of CP-673451 treatment showed inhibition of PDGFR-β
autophosphorylation both on western blot and flow cytometry. Downstream, phosphorylation
levels of STAT3, STAT5 and GSK-3β were decreased upon PDGFR-β inhibition (Fig. 1C).
Dephosphorylation of GSK-3β results in activation of this kinase, leading to phosphorylation
of MCL1, promoting degradation of this antiapoptotic protein. Interestingly, high
MCL1 levels are associated with lowered sensitivity to glucocorticoids, core components
of T-ALL/T-LBL treatment. To this end, combination therapy of PDGFR-β inhibition with
glucocorticoids can result in synergistic effects. Next, we will conduct in vivo studies
to verify if specific PDGFR-β inhibitors could be advantageous in the treatment of
PDGFR-βhigh T-ALL/T-LBL cases.
Image:
Summary/Conclusion: In this study, we have identified MYH9-PDGFRB as a novel rare
fusion activating PDGFR-β in a T-LBL patient sample. Moreover, in vitro evaluation
in T-ALL cell lines and ex vivo treatment of T-ALL PDX models showed therapeutic benefits
from PDGFR-β inhibition by CP-673451.
P318: A DUAL ROLE FOR PSIP1 IN T-ALL
L. Demoen1,2,*, F. Matthijssens1,2, Z. Debyser3, S. Goossens2, P. Van Vlierberghe1,2
1Center for Medical Genetics; 2Cancer Research Institute Ghent, Ghent University,
Gent; 3Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit
(KU) Leuven, Leuven, Belgium
Background: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological
cancer characterized by the diffuse infiltration of malignant T-cell progenitors in
the bone marrow. Currently, the cure rates have augmented to 80% for pediatric and
60% for adult T-ALL cases due to modern intensified chemotherapy treatments. These
cure rates sadly come at the cost of severe side effects and current therapy still
provides a poor outcome for primary therapy-resistant or relapse cases. Therefore,
there is an urgent need for the identification of key drivers and pathways in T-ALL
to develop targeted and less toxic therapies. PSIP1, a histone mark reader, was primarily
found as a dependency factor in MLL-rearranged AML, which was dispensable for normal
hematopoietic development, making it an interesting therapeutic target. Nevertheless,
the identification of inactivating mutations and deletions in T-ALL patients and lower
expression in all T-ALL subtypes, with exception of the TLX1-subtype, compared to
normal T-cell subsets, is rather an indication that PSIP1 can also serve as a tumor
suppressor in certain contexts.
Aims: In pursuance of determining the role of PSIP1 in tumor initiation and maintenance
in T-ALL, we studied the effect of loss of PSIP1 by utilizing several different T-ALL
mice models and human cell lines.
Methods: To identify the role of PSIP1 during tumor initiation, we crossed a conditional
Psip
fl/fl knock-out mouse model with two different spontaneous T-ALL mouse models. On
the other hand, to unravel the consequences of losing PSIP1 expression during tumor
maintenance a proliferation competition assay was performed and RNA-seq profiling
was used to identify downstream targets of PSIP1. Furthermore, tamoxifen-inducible
Psip
fl/fl mouse cell lines were created from Lmo2-driven T-ALL mouse tumors to test the
loss Psip1 during tumor maintenance both in vitro and in vivo.
Results: A conditional Psip
fl/fl knock-out mouse model was crossed with two different spontaneous T-ALL mouse
models, namely the Notch1 independent Lck-Cretg/+ Ptenfl/fl model and the Notch1 dependent
CD2-Lmo2tg/+ transgenic, which can also give rise to ETP-like murine T-ALL tumors.
The loss of Psip1 was shown to significantly accelerate T-ALL development in both
models, with a p-value of 0.0156 and 0.0002 (see figure) respectively.
Interestingly, in contrast with the role of PSIP1 loss in tumor initiation, the loss
of PSIP1 expression impaired cell proliferation in five different human T-ALL cell
lines. Further, tamoxifen-inducible Psip1 knock-out mouse cell lines were established
and are being used to further confirm the dependency role of Psip1 during tumor maintenance.
Lastly, COX20, an assembly factor of the cytochrome c oxidase in the mitochondria,
was found to be a downstream target of PSIP1 upon knock-down and could explain the
impairment of proliferation upon the loss of PSIP1 in these cell lines.
Image:
Summary/Conclusion: Altogether, these data indicate that PSIP1 can exert a dual role
in the context of T-ALL, either as a tumor suppressor gene during tumor initiation
or as a dependency factor in tumor maintenance. Its dispensability for normal hematopoiesis
and its dependency role during tumor maintenance, illustrate the potential of PSIP1
as a therapeutic target in T-ALL.
P319: THE TESTICULAR NICHE OF ACUTE LYMPHOBLASTIC LEUKEMIA – MOLECULAR AND CELLULAR
FACTORS FOR THE PREFERENTIAL MIGRATION AND SURVIVAL
T. Skroblyn1, J. J. Joedicke1, M. Pfau2, K. Krüger1, J.-P. Bourquin3, S. Izraeli4,
C. Eckert2,*, U. E. Höpken1
1Max-Delbrück-Center for Molecular Medicine; 2Paediatric Oncology/Haematology, Charité
- Universitätsmedizin Berlin, Berlin, Germany; 3University Children’s Hospital, Zurich,
Switzerland; 4Schneider Children’s Medical Center of Israel, Petach Tiqva, Israel
Background: The testis is the second most frequent (30%) non-hematological extramedullary
site of relapse in pediatric acute lymphoblastic leukemia (ALL) treated according
to European protocols. Testicular relapses usually occur late (> 6 months after completion
of frontline treatment), and boys with a ETV6::RUNX1 fusion gene positive ALL have
a significantly higher risk of a testicular relapse. In adult ALL, only about 1% of
patients have a testicular relapse. Event-free survival is between 40%-80% depending
on timing of relapse, involvement of the contralateral testis and of the bone marrow.
The surgical removal of the clinically-involved testis or high-dose irradiation are
the only current treatment options for testicular relapses to achieve a long-term
event-free survival, but it impacts long-term quality of life. The molecular mechanisms
regulating leukemic cell migration, growth, and survival in the testis have not been
sufficiently assessed.
Aims: We aimed at investigating our hypothesis that the testis is a frequent source
of extramedullary relapse in pediatric ALL because of the physiologically strong CXCL12
gradient. This allows circulating leukemia cells to migrate into the testis during
pre-puberty and to survive for potentially longer periods because of the specific
immune suppressive microenvironment at this unusual anatomical site.
Methods: To dissect the pre-pubertal cellular requirements and molecular pathways
contributing to testicular leukemic cell dissemination and survival, we combined analysis
of primary human leukemias with a patient derived xenograft (PDX) mouse model. We
analyzed chemokine receptor expression profiles of pediatric B-ALL samples from patients
with different relapse sites, studied the crosstalk of leukemia cell-stroma in co-cultures,
and established a pediatric B-ALL PDX mouse model with testicular involvement.
Results: The CXCL12-CXCR4 was identified as the driving force for B-ALL cell migration
and survival in the testicular leukemic niche. Analysis of primary pediatric patient
samples revealed that CXCR4 was the only chemokine receptor being robustly expressed
on B-ALL cells both at the time of diagnosis and relapse. One prerequisite for leukemic
cell infiltration in the testis mice was high surface expression of CXCR4 on PDX-ALL
cells, and CXCL12 secretion from testicular stroma. In affected patient testes, leukemic
cells localized within the interstitial space in close proximity to testicular macrophages.
Another requirement for migration and survival of leukemia cells in the testis was
pre-pubertal age of the recipient mice. Leukemic cell interactions with specific testis
macrophage subpopulations, isolated from affected testes, altered their phenotype
towards a pro-tumorigenic M2-like phenotype. The blockage of CXCR4-mediated functions
by anti-CXCR4 antibody treatment reduced testicular infiltration of PDX-ALL cells.
Summary/Conclusion: Collectively, a pre-pubertal condition together with high CXCR4
expression are factors affecting the leukemia permissive testicular microenvironment.
CXCR4 could be proposed as a promising target for therapeutic prevention of testicular
relapses in childhood B-ALL.
P320: TCF3-PBX1 LEUKEMIA INVADES IPSC-DERIVED CEREBRAL ORGANOIDS
P. Gebing1,*, S. Hänsch2, L. Lenk3, D. Schewe3,4, A. Borkhardt1,5, U. Fischer1,5,
S. Bhatia1,5
1Department of Pediatric Oncology, Hematology and Clinical Immonology, Medical Faculty,
Heinrich Heine University Düsseldorf; 2Center for Advanced Imaging - Cai, Heinrich
Heine Universität Düsseldorf, Düsseldorf; 3Department of Pediatrics I, ALL-BFM Group,
Christian-Albrechts University Kiel and University Medical Center, Kiel; 4Department
of Pediatrics, Otto-von-Guericke-University, Megdeburg; 5German Cancer Consortium
(DKTK), Düsseldorf, Germany
Background: Infiltration to the central nervous system (CNS) remains a major clinical
challenge to this day in treating childhood B-cell precursor acute lymphoblastic leukemia
(BCP-ALL). The relevant biological mechanisms underlying the invasion of the CNS,
are not yet understood and are mainly investigated using 2D cell culture and in vivo
mouse models1. Differences between species exist, particularly in cell types and architecture
between human and murine CNS, as well as the lack of specificity of infiltration between
leukemia subtypes. Human brain organoids potentially address some of these drawbacks
and may therefore present a complemental approach to study CNS invasion.
Aims: We aimed to establish human iPSC-derived cerebral organoids2 as a 3D model for
CNS invasion. Applying these organoids in coculture with primary patient-derived xenograft
(PDX) BCP-ALL cells (TCF3-PBX1), we aimed to investigate CNS-specific engraftment
while using cord-blood-derived CD34-positive hematopoietic stem cells as a healthy
control.
Methods: Brain organoids were derived from induced pluripotent cell lines (iPSCs)
of different donors and differentiated for 30 and 60 days. Intermediate progenitor,
as well as matured neuronal cell types, were controlled by immunofluorescent staining
of their respective markers (PAX6/Nestin, TUJ1/MAP2) which assemble in the complex
neuroepithelium of cerebral organoids. We cocultured CFDA-SE stained TCF3-PBX1-positive
patient-derived leukemia cells and cell lines with cerebral organoids. The initial
co-cultivation was designed to investigate when/if infiltration could occur. Furthermore,
a limiting dilution was introduced to determine the lowest cell concentration required
for invasion. 3D imaging of cleared organoids was performed to analyze invasion.
Results: Patient-derived TCF3-PBX-leukemic xenografted cells and cell lines invade
cerebral organoids after 14 days of coculture. We found that this is independent of
the iPSCs donors (testing in different batches an iPSC donors). The invasiveness was
determined by quantifying single and larger colonies of cells engrafted into the organoids.
TCF3-PBX1 revealed high invasiveness (P-value: 0.0006) compared to normal cord-blood-isolated
CD34+ hematopoietic stem cells (HSCs). In addition, as few as 50 cells (TCF3-PBX1)
per organoid were sufficient to invade the organoid tissue. Interestingly, the shRNA-downregulation
of CD79a/Igα in TCF3-PBX1 (697) cells, impeded CNS infiltration to cerebral organoids
(P-value: 0.0006), annealing their results in murine models1.
Image:
Summary/Conclusion: Human iPSC derived brain organoids display some key specificity
with regards to of BCP-ALL CNS infiltration. Although key features (vasculature, leptomeninges
etc.) which could strengthen the model, have not yet been successfully developed for
brain organoids, they may present a promising 3D culture system to adopt in CNS infiltration
studies.
References:
1. Lenk, L. et al. Commun. Biol.
4, 73 (2021).
2. Lancaster, M. A. et al. Nature
501, 373–379 (2013).
P321: PERTURBATIONS IN STIMULUS-INDUCED CYTOKINE RESPONSES IN CHILDREN WITH BCP-ALL
N. Ruechel*1,, M. Oldenburg*1, S. Janssen*2, A. Pandyra1, L. Wei3, D. Hein1, V. Jepsen1,
U. Fischer1, J. Hauer4, F. Auer4, A. Beer5, O. Adams6, C. Mackenzie7, M. Jaeger8,
M. Netea9, A. Borkhardt*1, K. Goessling*1
1Pediatric Oncology, Hematology and Clinical Immunology, University Hospital Duesseldorf,
Duesseldorf; 2Algorithmic Bioinformatics, Department of Biology and Chemistry, Justus
Liebig University, Gießen; 3Department of Molecular Medicine II, Medical Faculty,
Heinrich-Heine University, Duesseldorf; 4Department of Pediatrics and Children’s Cancer
Research Center (CCRC), Technical University (TU), Muenchen; 5Pediatric Hematology
and Oncology, Department of Pediatrics, University Hospital Carl Gustav Carus, Dresden;
6Institute for Virology, Medical Faculty, University Hospital; 7Institute of Medical
Microbiology and Hospital Hygiene, University Hospital Duesseldorf, Duesseldorf, Germany;
8Radboudumc, Nijmegen, Department of experimental internal medicine, Radboucumc; 9Radboudumc,
Nijmegen, Department of experimental internal medicine, Radboudumc, Nijmegen, Netherlands
Background: B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is the most common
subtype of childhood leukemia with a peak during early childhood when children are
exposed to viral infections. About 5% of all newborns carry a preleukemic clone, but
only about 0,2% of them develop BCP-ALL later in life. It has been assumed that a
dysregulated, altered immune response underlies the outgrowth of a pre-leukemic clone
in infection-triggered B-cell precursor acute lymphoblastic leukemia (BCP-ALL).
Aims: We investigated whether children with BCP-ALL differ in their cytokine response
from healthy, age-matched, non-leukemic children, after immune cells were challenged
with viral, fungal or bacterial stimuli.
Methods: We set up a functional experimental platform with 73 stimulus-cytokine pairs
to comprehensively characterize the immune profile of pediatric patients with BCP-ALL
with the two most common genetic subtypes, either carrying the ETV6/RUNX1 (E/R) gene
fusion or the high-hyperdiploid (HD) karyotype in comparison with an age-and gender-matched
healthy control cohort without BCP-ALL and their healthy parents (in total n = 101
individuals). Children were in stable first remission, at least two years after the
end of therapy and had a fully reconstituted immune system. Blood was taken of patients
and controls in parallel and peripheral blood mononuclear cells (PBMCs) were isolated
using a density gradient centrifugation. Cells had been stimulated with various fungal,
bacterial and viral pathogens, such as Candida albicans, Staphylococcus aureus, Influenza
virus, Respiratory Syncytial virus and toll-like receptor ligands (TLR), such as lipopolysaccharide
(TLR4), R848 (TLR7/8) or CpG (TLR9); subsequently cytokines (IFNa, IL-1b, IL-1Ra,
IL-10, IL-12p70, IL-17, IFNg) had been measured in the cell culture supernatant.
Results: Cytokine base line level did not differ between the groups, while the challenge
with various infectious antigens resulted in an overall elevated cytokine production
of the BCP-ALL patients with E/R fusion (n=11) (p < 0.01, Mann-Whitney-Wilcoxon test
two-sided with Benjamini-Hochberg correction). By deciphering the characteristics
of the immune response, a remarkable difference for the anti-viral immune response
(p < 0.01) was identified, while the anti-bacterial and anti-fungal response did not
differ: The IFNa-response induced by Influenza virus and R848 and the IL-17-response
induced by R848 differed most prominently, although not significantly after correction
for multiple testing. Further analysis of the cytokine profile of the E/R parents
compared with an age- and gender-matched healthy control group revealed an overall
significantly elevated cytokine response (p < 0.05) after stimulation, but the different
sub-features, such as the fungal, bacterial and viral immune responses were not different.
Most strikingly, we could identify this pattern specifically for the E/R cohort. Comparing
the cytokine responses of the HD patients (n=8) with the healthy controls (n=22) did
not result in any significant differences.
Image:
Summary/Conclusion: Our study suggests that an altered pro-inflammatory anti-viral
cytokine response pattern in children with E/R-BCP-ALL is specific to those children,
who later developed the BCP-ALL. It is still present years after chemotherapy and
possibly contributes to the outgrowth of a pre-leukemic clone, consistently generated
in utero during fetal hematopoiesis.
P322: THERAPEUTIC EFFECT OF TARGETING CASEIN KINASE II / WDR5 AXIS IN T-CELL ACUTE
LYMPHOBLASTIC LEUKEMIA
Q. Han1, C. Song2,3, Z. Ge1,*
1Department of Hematology, Zhongda Hospital,School of Medicine, Southeast University,
Institute of Hematology Southeast University, Nanjing, China; 2Hershey Medical Center,
Pennsylvania State University, Hershey; 3Division of Hematology, The Ohio State University
Wexner Medical Center, the James Cancer Hospital, Columbus, United States of America
Background: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological
malignance with limit treatment choices and extremely poor prognosis. WD repeat domain
5 (WDR5) is essential for the methyltransferase activity of MLL1 complex and plays
an important role in various biological functions. We reported that targeting Casein
Kinase II (CK2)/Ikaros axis by CK2 inhibitor CX-4945 has therapeutic efficacy in B-ALL.
However, the effect of WDR5 inhibitor and CX-4945 in T-ALL remains unknown.
Aims: This study is to explore the synergistic effect of WDR5 inhibitor OICR-9429
and CK2 inhibitor CX-4945 and the potential underlying mechanisms of this novel drug
combination in T-ALL.
Methods: The expression profile and clinical significance of WDR5 and ATAD2 in T-ALL
were explored by analyzing the transcriptome data of T-ALL patients from the GEO database.
CCK-8 assay was performed to analyze the synergistic effect of OICR-9429 combined
CX-4945 in CEM and MOLT4 cell lines. Cell cycle effect of inhibitors were detected
by flow cytometry after 72h cultured. RNA-seq was performed after CEM cells were treated
with OICR-9429, CX-4945, and vehicle control for 72 hours. Differential expression
genes (DEGs) and pathways were analyzed by R-software. WDR5 and ATAD2 were knockdown
by shRNA in CEM and MOLT4 cells. qPCR and Western Bolt were performed to test the
DEGs detected by RNA-seq.
Results:
WDR5 is significantly over-expressed in T-ALL patients compared with healthy controls
in two independent cohorts and is associated with shorter overall survival (P<0.01).
Bioinformatic analysis revealed that the cell cycle was the most significantly altered
pathway between WDR5 high and low expression patients (P<0.001) (Figure 1A). In vitro
assay showed that OICR-9429 and CX-4945 alone exhibited an anti-proliferation effect
on T-ALL cells after being treated for 72h. In addition, we found that OICR-9429 and
CX-4945 had a significant synergistic effect on cell proliferation arrest in CEM (Figure
1B) and MOLT4 cells compared with a single drug (P<0.05). Cell cycle assay revealed
that OICR-9429 (Figure 1C) and CX-4945 induced a G1-phase arrest in CEM and MOLT4
cells and this effect was significantly increased upon OICR-9429 and CX-4945 combination
(P<0.01) (Figure 1D). The anti-proliferation and cell cycle arrest effects were also
exhibited in WDR5 knockdown CEM and MOLT4 cells (P<0.05). Pathway analysis of the
DEGs showed that the cell cycle pathway was obviously altered after OICR-9429 and
CX-4945 treated (P<0.001) (Figure 1E). In addition, CX-4945 strongly down-regulated
WDR5 (Figure 1F) expression in both transcriptome level and protein level, and knockdown
of WDR5 increased the anti-leukemic effect of CX-4945 (P<0.01) (Figure 1G), suggesting
that WDR5 was the downstream target of CK2 and CX-4945 in T-ALL. Moreover, RNA-seq
data showed that ATAD2, an important oncogene was suppressed upon treatment of OICR-9429,
CX-4945 (Figure 1F), and WDR5 knockdown (P<0.001). Moreover, the combination of OICR-9429
and CX-4945 strongly induced ATAD2 downregulation compared with either single drug
(P<0.001) (Figure 1H). Likewise, we found that ATAD2 is up-regulated in T-ALL patients
and associated with poor prognosis and cell cycle pathway abnormal activation (P<0.001).
ATAD2 knockdown significantly inhibited cell proliferation and induced a G1-phase
cell cycle arrest in T-ALL cells (P<0.01) (Fig 1I).
Image:
Summary/Conclusion: Our data showed that OICR-9429 and CX-4945 have synergistic efficacy
in T-ALL. Dual targeting WDR5 and CK2 may be a potential therapeutic approach for
T-ALL through the CK2-WDR5-ATAD2 axis.
P323: STREAMLINING PRECLINICAL IN VIVO TREATMENT TRIALS BY MULTIPLEXING GENETICALLY
LABELLED PDX MODELS OF SEVERAL PATIENTS IN A SINGLE MOUSE
K. Hunt1,*, D. Amend1, R. Ludwig1,2, B. Vick1,2, A. K. Wirth1, T. Herold1,3, I. Jeremias1,2,4
1Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German
Research Center for Environmental Health (HMGU), Munich; 2German Cancer Consortium
(DKTK), partner site Munich; 3Laboratory for Leukemia Diagnostics, Department of Medicine
III, University Hospital, LMU Munich; 4Department of Pediatrics, University Hospital,
Ludwig Maximilian University (LMU), Munich, Germany
Background: Better treatment options are intensively needed for acute lymphoblastic
leukemia (ALL) and acute myeloid leukemia (AML). Before application in clinical trials,
novel therapies require preclinical testing which is resource intensive. In previous
work, we had shown that multiplexing several PDX cell populations within a single
mouse in competitive in vivo experiments gives identical results to studying each
population in separate mice (Liu et al., Biomarker Research 2020).
Aims: Here, we aimed at transferring multiplexing to preclinical in vivo treatment
trials. We aimed to increase the efficiency of preclinical drug testing in vivo by
simultaneously testing several samples in parallel in a single mouse, using a competitive
in vivo approach.
Methods: From primary patient material, we had established patient derived xenograft
(PDX) mouse models of ALL and AML which allow serial transplantation in immunodeficient
NSG mice. We designed and cloned 5 different lentiviral constructs to label 5 individual
samples with both a unique fluorochrome as well as a connected unique genetic barcode,
for later detection by flow cytometry or next generation sequencing, respectively.
In addition, luciferase was recombinantly expressed in all PDX models to allow repetitive
monitoring of leukemia growth and treatment response using bioluminescence in vivo
imaging (BLI). Up to 5 PDX samples, either ALL or AML, were multiplexed and aliquots
injected into groups of mice (n=4-6). For post mortem analysis, PDX cells were reisolated
from murine bone marrow and spleen and absolute tumor burden of PDX cells was quantified
by flow cytometry of exactly 1/10 of the bone marrow and spleen. Determining fluorochrome
composition allowed quantifying the proportion that each of the 5 samples contributed
to the entire tumor load. In case of treatment, a response rate for each individual
samples was determined by comparing cell numbers in treated versus control mice.
Results:
From our pool of established PDX samples, we selected 5 ALL or 5 AML samples with
different molecular alterations, but similar in vivo growth behaviour. Each PDX model
was labelled by its individual fluorochrome and genetic barcode, while all models
expressed luciferase. Tumor growth as well as the ratio between the samples was repetitively
analysed over the entire course of leukemia outgrowth. The composition of the injection
mixture was optimized such that each sample accounted for roughly 20% of the total
leukemic burden at the time point of start of treatment.
Mice containing multiplexed PDX cells from 5 different patients with ALL were treated
for 2 weeks with either verum treatment or with solvent as control. In a study using
the BCL-2 inhibitor Venetoclax in PDX ALL models, we were able to distinguish between
sensitive and resistant PDX samples. While two samples showed a drastic decrease in
tumor burden, three samples showed no or only a mild response. This effect could be
observed both in cells isolated from bone marrow as well as spleen. Variances between
mice were rather small, with few exceptions for some treated mice.
With this approach, we were able to define the effect of a single drug on up to five
distinct samples in parallel, reducing resources by factor 5.
Image:
Summary/Conclusion:
Taken together, we established a multiplex protocol for in vivo therapy trials that
allows simultaneous testing of up to 5 PDX samples in competitive in vivo trials.
The approach reduced the required number of experimental mice by a factor 5, in line
with the 3R concept. In the future, our approach might rationalize in vivo drug trials.
P324: PHARMACOLOGIC INHIBITION OF DYRK1A RENDERS HIGH-RISK KMT2A-R ALL SENSITIVE TO
VENETOCLAX
C. Hurtz1,*, G. Wertheim2, J. Chukinas3, R. Bhansali4, S. Swaminathan5, J. Crispino6,
J. Shi7, S. Tasian3, M. Carroll8
1Fels Institute for Personalized Medicine, Temple University; 2Department of Pathology
and Laboratory Medicine, University of Pennsylvania; 3Oncology, CHOP, Philadelphia;
4Hematology Oncology, Upenn, Pehiladelphia; 5Systems Biology, City of Hope, Monrovia;
6Experimental Hematology, St. Jude Children’s Research Hospital, Memphis; 7Cancer
Biology; 8Medicine, Upenn, Philadelphia, United States of America
Background:
KMT2A-rearranged (R) ALL is a high-risk disease with a frequency of 70% in infants
and 10% in children and adults with ALL and is associated with chemoresistance, relapse,
and poor survival. Current intensive multiagent chemotherapy regimens induce significant
side effects, yet fail to cure many patients, demonstrating continued need for novel
therapeutic approaches.
Aims: Determine if pharmacologic DYRK1A inhibition may be a novel treatment strategy
for patients with KMT2A-R ALL.
Methods: To identify novel targets in KMT2A-R leukemia, we performed a domain-specific
kinome-wide CRISPR screen and identified multiple kinases required for cell growth.
We focused on DYRK1A as it met the following three criteria: 1) Growth inhibition
upon kinase targeting was greater in KMT2A-R leukemic cells than in non-KMT2A-R cells,
2) DYRK1A was not found to be common essential gene assessed through the Cancer Dependency
Map, 3) Small molecule inhibitors are available.
Results: We analyzed multiple ChIP-Seq experiments and identified that KMT2A-fusions
directly bind to the DYRK1A promoter. Our RT-PCR and Western blot analyses demonstrate
that KMT2A-R ALL cells treated with a menin inhibitor to disrupt the transcriptional
activity of the KMT2A-R complex, downregulate DYRK1A, indicating direct regulation
of DYRK1A by the KMT2A-fusion. We further observed that pharmacologic inhibition of
DYRK1A with EHT1610 induced leukemic cell growth inhibition in vitro and in vivo,
demonstrating that DYRK1A could be a new therapeutic target in KMT2A-R ALL cells.
To further elucidate the mechanism of DYRK1A function, we treated several KMT2A-R
ALL cell lines in vitro with EHT1610, which surprisingly resulted in the upregulation
of MYC and hyperphosphorylation of the RAS/MAPK target ERK. Given that ERK hyperactivation
stops B cell proliferation during early B cell development to allow them to rearrange
their B cell receptor, we hypothesized that cell cycle inhibition upon ERK hyperactivation
remains as a conserved mechanism of cell cycle regulation in KMT2A-R ALL. Strikingly,
combining DYRK1A inhibition with the MEK inhibitor trametinib antagonistically rescued
KMT2A-R ALL cell proliferation, indicating that ERK hyperactivation is the main driver
of DYRK1A inhibitor mediated cell cycle arrest. Given that DYRK1A inhibitor does not
induce apoptosis and cells restart cell proliferation after EHT1610 withdrawal we
concluded that a DYRK1A monotherapy may not be an ideal new treatment option. However,
it has been reported that increased MYC activity induces the accumulation of BIM in
Burkitt’s Lymphoma. Given the increased expression of MYC following DYRK1A inhibition
we performed a new Western blot analysis and validated increased expression of BIM
in our KMT2A-R ALL cell lines after EHT1610 treatment. To test if targeting the interaction
of BIM with BCL2 will induce an apoptotic effect when combined with EHT1610, we treated
four KMT2A-R ALL cell lines with increasing concentrations of EHT1610 and the BCL2
inhibitor venetoclax. Strikingly, the combination of DYRK1A inhibition with BCL2 inhibition
synergistically killed KMT2A-R ALL cells and significantly reduced the leukemia burden
in vivo.
Image:
Summary/Conclusion: Our results validate DYRK1A as an important molecule to regulate
cell proliferation via inhibition of MYC and ERK. Targeting DYRK1A results in the
accumulation of BIM, which renders the cells sensitive to BCL2 inhibition via venetoclax.
While further in vivo validation is needed, we predict that combining DYRK1A inhibition
with venetoclax may be a novel precision medicine strategy for the treatment of KMT2A-R
ALL.
P325: A KINASE-DEAD VARIANT OF CDK6 IS ASSOCIATED WITH REDUCED TUMORIGENIC POTENTIAL
AND A DISTINCT TRANSCRIPTIONAL REWIRING IN AN ALL MODEL
T. Klampfl1,*, S. Nebenführ1, M. Zojer1, F. Bellutti1, V. Sexl1, K. Kollmann1
1Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna,
Vienna, Austria
Background: The cell cycle kinase Cdk6 is a major regulator of cell cycle progression
from G1 to S phase. Cdk4/6 kinase inhibitors are approved for the treatment of breast
cancer and show promising results in pre-clinical studies of other cancers, including
hematological malignancies. Cdk6 was also shown to exhibit kinase-independent functions
including transcriptional regulation of gene expression.
Aims: A systematic understanding of the kinase independent functions of Cdk6 should
allow for a better comprehension of the mechanism of action of Cdk4/6 inhibitors.
Methods: To model kinase-independent functions of Cdk6 in Acute Lymphoblastic Leukemia
(ALL), mouse pre/pro B-cell lines were generated. Whole bone marrow from mice carrying
a kinase-dead mutant of Cdk6 (Cdk6-K43M), Cdk6 knock-out (Cdk6-KO) or wildtype Cdk6
(Cdk6-WT) were retrovirally transduced with the BCR-ABL1p185 fusion gene. The resulting
cell lines were analyzed in-vivo in transplant settings as well as on the molecular
level where RNA-seq and ChIP-seq were performed.
Results: Tail vain injection of Cdk6-K43M, Cdk6-KO and Cdk6-WT pre/pro B-cell lines
into NSG mice led to the development of lymphoid malignancies albeit with different
latencies depending on genotype. Mice injected with Cdk6-KO lines showed a significantly
longer survival than mice injected with Cdk6-WT lines. The injection of Cdk6-K43M
cells led to an intermediate phenotype with significantly shorter survival than Cdk6-KO,
but significantly longer survival than Cdk6-WT. Subcutaneous injection of the cell
lines also allowed for the formation of tumors, where tumor weight was largest in
mice injected with Cdk6-WT cells, intermediate in the case of Cdk6-K43M cells and
lowest in the case of Cdk6-KO cells. Therefore, while kinase-dead Cdk6 had reduced
tumorigenic potential compared to Cdk6-WT, loss of kinase function alone exhibited
stronger tumorigenic potential than the complete Cdk6 KO.
Differential gene expression analysis between cell lines of the three genotypes revealed
common alterations associated with Cdk6-K43M and Cdk6-KO compared to Cdk6-WT. Some
of these changes could directly be attributed to the loss of kinase function, including
for example the downregulation of E2F target genes. Additionally, changes specific
for either Cdk6-K43M or Cdk6-KO were observed. As Cdk6-K43M retains kinase-independent
functionality, which includes transcriptional regulation, we investigated the DNA
binding profile of transcriptional complexes containing Cdk6-K43M. ChIP-seq analysis
revealed that such complexes bind the DNA at largely similar sites as complexes containing
Cdk6-WT. The majority of genes differentially expressed between Cdk6-WT and Cdk6-K43M
were associated with transcriptional complexes harboring Cdk6.
Summary/Conclusion: Pre/pro B-cell lines carrying kinase-dead Cdk6 (Cdk6-K43M) showed
intermediate tumorigenic potential between lines with Cdk6-WT and Cdk6-KO. While Cdk6-K43M
is associated with the DNA at similar sites as Cdk6-WT, there are distinct transcriptional
changes associated with this mutation. The further elucidation of these alterations
is expected to provide a better understanding of both, kinase independent functions
of Cdk6 as well as the effects of its pharmacological inhibition.
P326: PROGNOSTIC UTILITY OF NON-HOMOLOGOUS END JOINING DNA REPAIR PATHWAY GENES IN
T-CELL ACUTE LYMPHOBLASTIC LEUKEMIA
S. KUMARI1,*, J. SINGH1, M. Arora2, S. Bakhshi3, J. K. Palanichamy2, A. Sharma3, P.
Tanwar1, A. R. Singh1, I. Qamar4, A. Chopra1
1Lab Oncology Unit; 2Department of biochemistry; 3Department of medical oncology,
All India Institute of Medical Sciences, New Delhi; 4School of Biotechnology, Gautam
Buddha University, Gerater Noida, India
Background: T-ALL is a molecularly heterogeneous malignancy characterized by transformation
and differentiation blockage at different stages of T cell development. Efficient
DNA repair has shown to enable leukemic cells to survive the damaging effects of therapeutics,
therefore, understanding of DNA repair alterations in leukemia may provide novel druggable
targets and prognostic biomarkers. Notably, it is important to determine that whether
the variable ability of T cell to repair DNA damage also translates into the corresponding
molecular profiles of T-ALL. Several components of different DNA repair pathways have
been recently identified to play critical role in ALL pathophysiology and drug resistance.
However, the clinical significance of major DNA double strand break repair pathway
Non-Homologous End Joining (NHEJ) in T cell acute lymphoblastic leukemia remains to
be studied in detail.
Aims:
This study aims to assess the importance of DNA damage response pathways in T-cell
acute
lymphoblastic leukemia cancer (ALL).
Methods: We assessed the gene expression pattern of 21 genes involved in NHEJ pathwayincluding
six core genes, accessory genes and other regulators such as MRE complex genes in
207 T-ALL patients. In order to determine the changes in expression pattern of NHEJ
genes during the course of T-ALL, gene expression was assessed at different time points,
including clinical sample at diagnosis, minimal residual diseaseand at relapse. Real
time PCR was used to assess the expression of these genes. We further determined the
association of NHEJ pathway gene expression with clinical and molecular features.
Results: We observed higher expression of TOX, WRN, NHEJ1, APLF, TDP2 and reduced
expression of XRCC4, POLL, PRKDC, APTX, PNKP, XRCC5, DCLRC1C, MRE11, XRCC6, NBN in
diagnostic T-ALL samples compared to normal bone marrow. Further, PRKDC, RAD50, and
XRCC6 displayed lesser expression in prednisolone resistant group compared to sensitive
group. At minimal residual disease, XRCC4, POLL, WRN, POLM, APTX, PNKP, XRCC5, DCLRC1C,
XRCC6, NBN, TDP1 exhibited higher expression and TOX, LIG4, MRE11 exhibited reduced
expression compared to diagnostic samples. XRCC4, PRKDC, NHEJ1, PNKP, XRCC5, DCLRC1C,
MRE11, XRCC6, NBN and TDP1 exhibited higher expression in relapse samples compared
to diagnostic samples.
The clinical significance of expression of NHEJ genes was explored in detail. Low
expression of XRCC4, PRKDC, RAD50, XRCC5, XRCC6 and TDP1 gene was associated with
immature immunophenotype (IPT) as compared to cortical and mature IPT.Patients with
ETP-ALL IPT more frequently had lesser expression of XRCC4, PRKDC, RAD50, LIG4, APTX,
MRE11, XRCC6andgene compared to other IPT groups. In diagnostic samples, higher XRCC4
expression was associated with better overall survival (OS) and even free survival
(EFS). Additionally, low WRN expression was associated with poor EFS and relapse free
survival (RFS). Interestingly, high DCLRC1C expression was associated with favorable
OS, EFS and RFS.
Summary/Conclusion: We observed a distinct expression profile of NHEJ genes among
molecularly heterogeneous groups of T-ALL patient’spossibly explaining different extent
of DNA repair among patient with different immunological groups and in response to
therapy. Interestingly, this depicted a clear change in the DNA repair responses in
the leukemia patients before and post chemotherapy. Gene expression emerged as the
predictor of OS, EFS and RFS in T-ALL patientsthat might be helpful in the improvement
of the treatment outcome in T-ALL therapy
P327: QUANTITATIVE EXPRESSION PROFILING OF SURFACE ANTIGENS ON PERIPHERAL BLOOD LEUKOCYTE
SUBSETS AND CHILDHOOD T-ALL CELLS USING A STANDARDIZED FLOW CYTOMETRY WORKFLOW: A
HCDM CDMAPS INITIATIVE
D. Kužílková1,*, J. Puñet-Ortiz2, P. M. Aui3, J. Fernandez2, F. Karel1, P. Engel2,
M. C. van Zelm3,4, T. Kalina1
1CLIP (Childhood Leukemia Investigation Prague), Department of Pediatric Haematology
and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
and University Hospital Motol, Prague, Czechia; 2Department of Biomedical Sciences,
University of Barcelona, Barcelona, Spain; 3Department of Immunology and Pathology,
Central Clinical School, Monash University; 4Department of Allergy, Immunology and
Respiratory Medicine, Central Clinical School, Monash University and Alfred Hospital,
Melbourne, Australia
Background: Human Leukocyte Differentiation Antigen (HLDA) workshops are organized
by the Human Cell Differentiation Molecules (HCDM) consortium to test and validate
the reactivity of particular antibody clones to specific targets. Thereby the consortium
provides the scientific community validated antibody clones reactive to particular
cluster of differentiation (CD) markers. Although this approach has been used since
the 1980s, quantitative profiling of CD markers at “single-cell“ level and benchmarking
of reagents are currently lacking.
Aims: We aimed to develop a flow cytometric procedure allowing CD marker expression
profiling in a standardized way in time and place
Methods: First, we developed and titrated two antibody panels with a free position
in the phycoerythrin (PE) channel. The panels enable identification of 27 innate and
adaptive leukocyte cell populations present in peripheral blood. The panels were custom
dried in 96-well plates, and the Quantibrite™ PE Beads were used for quantification
of the PE signal. Subsequently, we developed a high content framework to evaluate
the titration of PE conjugated monoclonal antibodies using fluorescently barcoded
cell lines and peripheral blood cells. The selected titer and critical antibody information
(such as clone, catalogue number, vendor, gene and CDname etc.) were centrally stored
in an inventory table, which was expanded into an experiment master table (EMT) following
inclusion of experimental details (e.g. the position of individual mAbs in 96-well
plate, experiment name, operator etc.). The EMT was used to generate an experimental
protocol with automated calculation of reagent amounts and volumes. Post acquisition,
the fcs files were annotated using all relevant experimental information from the
EMT table.
Results: To validate our approach, we quantified protein expression of four selected
CD markers (CD11b, CD31, CD38 and CD40) with well-known expression pattern on peripheral
blood leukocytes that showed high reproducibility across centers. We also performed
benchmarking of four anti-CD3 clones, of which the titration curves revealed variable
performance: from high-performance TB3 clone, through intermediate-performance UCHT1
and SK7 clones to low-performance MEM-57 clone. Three out of four anti-CD3 clones
showed similar pattern of staining, whereas the MEM-57 clone showed decreased intensity
on CD4 and CD8 T cells while retaining comparable intensity to the other clones for
TCRγδ+ T cells. Our pilot results on childhood T-ALL patient samples (n=7) revealed
potential targets for minimal residual disease monitoring.
Summary/Conclusion: In summary, we optimized a procedure for quantitative expression
profiling of surface antigens on subsets of blood leukocyte and proved its feasibility
with inter-laboratory comparison in three different laboratories. The presented workflow
enables (i) to map the expression patterns of HLDA-approved antibody clones to CD
markers, (ii) to benchmark new antibody clones to established CD markers, (iii) to
define new clusters of differentiation in future HLDA workshops and (iv) mapping of
childhood T-ALL cells.
Acknowledgement: The reagents were kindly provided by Exbio and BioLegend. The work
was financially supported by project NU20-05-00282 of the Czech Republic Ministry
of Health
P328: DYNAMIC EVOLUTION OF TCF3-PBX1 LEUKEMIAS AT THE SINGLE-CELL LEVEL UNDER CHEMOTHERAPY
PRESSURE
M. Kusterer1, M. Lahnalampi2,*, M. Voutilainen2, G. Gentile1, S. Pennisi1, J. Norona1,
G. Greve1, M. Lübbert1, R. Sankowski3, M. Prinz3, S. Killmer4, M. Salvat Lago4, B.
Bengsch4, M. Cleary5, V. Zachariadis6, M. Enge6, O. Lohi7, M. Heinäniemi2, J. Duque-Afonso1
1Department of Hematology/Oncology/Stem Cell Transplantation, University of Freiburg
Medical Center, Freiburg, Germany; 2Institute of Biomedicine, University of Eastern
Finland, Kuopio, Finland; 3Department of Neuropathology; 4Department of Gastroenterology,
University of Freiburg Medical Center, Freiburg, Germany; 5Department of Pathology,
Stanford University, Standford, United States of America; 6Department of Oncology-Pathology,
Karolinska Institute, Stockholm, Sweden; 7Tampere Center for Child, Adolescent, and
Maternal Health Research, Tampere University, Tampere, Finland
Background: Acute lymphoblastic leukemia (ALL) is the most common childhood cancer.
While research has been focused on high-risk patients, the biology of low-to-intermediate-risk
patients has so far been inadequately investigated and can lead to overtreatment with
severe side effects and under treatment with risk of relapse. The translocation t(1;19)
codes for chimeric fusion protein TCF3-PBX1, which is associated with intermediate
risk ALL. Using our previous generated TCF3-PBX1 conditional knock-in mice, we established
a model to study in vivo chemotherapy resistance.
Aims: We hypothesize that chemotherapy and microenvironment play a crucial role in
development of resistance in TCF3-PBX1 leukemias by influencing transcriptional regulation,
activation of signaling pathways and the hierarchical structure of leukemic cells.
In this project we aim to characterize dynamic changes of TCF3-PBX1 leukemia cells
in different tissues under chemotherapy pressure.
Methods: Recipient C57/BL6 healthy mice were sub-lethally irradiated, transplanted
with mouse TCF3-PBX1 leukemia cells and treated with vehicle (n=15), prednisolone
(n=13) and daunorubicin (n=15) for 20 days. Mice were monitored for signs of disease
regularly and circulating GFP+ TCF3-PBX1 leukemia cells were assessed by flow cytometry.
Sick mice were sacrificed, leukemia cells were isolated from five different organs
(bone marrow, spleen, lymph nodes, spinal cord and brain) and characterized by immunophenotyping,
sanger sequencing, bulk RNA sequencing (bulk RNAseq) of GFP+ sorted cells, single
cell RNA sequencing and mass cytometry (CYTOF).
Results: All mice transplanted with TCF3-PBX1 leukemia cells and treated with vehicle
succumbed to disease with a median survival of about 70 days. We optimized chemotherapy
drug concentration and transplanted cell dose, so 60% of mice treated with prednisolone
or daunorubicin survived at least 150 days. Leukemic infiltration was showed by histological
stainings in analyzed tissues including central nervous system (CNS) (spinal cord,
brain) and GFP+ leukemia cells were quantified by flow cytometry. No major differences
were observed in immunophenotype of TCF3-PBX1 leukemia and variant allele frequency
(VAF) of the known PTPN11 mutation in analyzed tissues or depending on in vivo treatments.
Bulk RNAseq of FACS-sorted GFP+ TCF3-PBX1 leukemia cells were clustered and CNS cells
separated from other tissues. scRNAseq revealed additional heterogeneity within each
tissue based on signaling pathway and cell cycle activity. Interleukin signaling,
regulation of apoptosis pathways, and signaling by the B cell receptor (BCR) were
regulated based on pathway analysis in CNS compared to other tissues. Hence, we elucidated
the hierarchical structure of hematopoietic cells, interaction of leukemic cells with
the microenvironment, and changes in signaling response to chemotherapy in vivo treatment
by mass spectrometry (CyTOF) to validate the identified altered signaling pathway
activities at protein level.
Summary/Conclusion: We have developed a mouse model in order to characterize in vivo
chemotherapy resistance depending on niche. Global transcriptomics and phospho-proteomics
at the single-cell level might elucidate novel mechanisms of chemotherapy resistance
suitable for pharmacological therapies.
P329: UNRAVELING THE ROLE OF GATA3 IN EARLY HUMAN T CELL DEVELOPMENT AND EARLY T CELL
PRECURSOR ALL (ETP-ALL)
N. Lambrechts1,2,*, K. L. Liang1,2, T. Putteman1, J. Roels1,3, J. Van Hulle1, T. Taghon1,2
1Diagnostic Sciences, Ghent University; 2Cancer Research Institute Ghent; 3Biomolecular
Medicine, Ghent University, Ghent, Belgium
Background: ETP-ALL is a subtype of T cell acute lymphoblastic leukemia (T-ALL) and
can be identified by a characteristic immunophenotype and gene expression profile.
Treatment of relapsed ETP-ALL remains challenging using conventional chemotherapy
and little targeted therapies are available. A better understanding in the molecular
mechanisms involved in the leukemogenesis and maintenance of ETP-ALL could lead to
improved and more targeted treatments. An estimated 9% of ETP-ALL patients carry mutations
in GATA3, a transcription factor known to regulate T cell commitment by inducing T
lineage genes and inhibiting genes important for other lineages such as NK and B cells.
Although these mutations have been described to be loss-of-function mutations, the
exact mechanisms through which they contribute to ETP-ALL remain to be elucidated.
Aims: We aim to gain insights into the molecular mechanisms through which GATA3 regulates
normal human early T cell development and how disruption of GATA3 function, as a result
of mutation, can contribute to the emergence of ETP-ALL.
Methods: We have studied the effect of the most prevalent GATA3 mutation (c.827G>A,
p.R276Q) in ETP-ALL on normal human T cell development using in vitro differentiation
cultures. We examined the impact of GATA3 p.R276Q on gene expression, DNA binding
and chromatin accessibility through RNA-, CUT&Tag- and ATAC-sequencing respectively,
starting from primary CD34+CD1a- early human T cell progenitors isolated from thymus
or the ETP-like cell line PER117.
Results: While overexpression of wild-type GATA3 in thymic progenitor T cells induces
an acceleration of development towards the CD4+CD8ab+ double positive stage, overexpression
of the mutant form results in a delay and an accumulation of CD4 immature single positive
cells in OP9-DL1 co-cultures. Remarkably, the inhibition of NK cell development, which
is an important function of wild-type GATA3, is maintained by GATA3 R276Q. This indicates
that GATA3 R276Q is not merely a loss-of-function mutation. RNA-, CUT&Tag- and ATAC
sequencing after overexpression in human CD34+CD1a- immature thymocytes or the ETP-ALL-like
cell line PER117 revealed that most key regulatory effects are maintained by GATA3
R276Q compared to its wild-type counterpart, including induction of T cell genes and
inhibition of stem cell and NK cell genes, binding to its target regions by recognizing
GATA motifs and exerting a pioneering function by opening and closing chromatin.
Summary/Conclusion: Continuous GATA3-R276Q has a detrimental effect on T cell development
in vitro. Since the inhibition on NK cell development is maintained, these findings
suggest that this decision is uncoupled from the induction of T cell lineage commitment.
Many important effects on RNA expression, DNA binding and chromatin accessibility
are maintained by the R276Q mutant compared to wild-type GATA3, pointing out that
this mutation not purely acts as a loss-of-function. The exact mechanisms through
which GATA3 R276Q contributes to ETP-ALL still remain to be elucidated, but our results
show that inhibition of T cell development most likely is one piece of the puzzle
that leads to malignant transformation.
P330: TARGETING BTN2A1 BY A UNIQUE ACTIVATING MAB IMPROVES ANTI-TUMOR FUNCTIONS OF
VΓ9VΔ2 T CELLS
A.-C. Le Floch1,*, C. Imbert1, A. de Gassart2, A. Le Roy1, L. Gorvel1, N. Vey3,4,
A. Anastasio1, A. Briantais1, N. Boucherit1, C. E. Cano2, D. Olive3,4
1Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068; 2ImCheck Therapeutics;
3Institut Paoli-Calmettes; 4Aix-Marseille Université UM105, CNRS UMR 7258, Marseille,
France
Background: Vγ9Vδ2 T cells are new promising cytotoxic effectors in cancer immunotherapy.
In acute myeloid leukemia and in non-Hodgkin lymphomas, Vγ9Vδ2 T cells-based immunotherapy
has shown encouraging results both in preclinical models and in early phase clinical
trials. But very few data are currently available on susceptibility of acute lymphoblastic
leukemia (ALL) cells to Vγ9Vδ2 T cell cytotoxicity. Vγ9Vδ2 T cells are activated by
phosphoantigens bound to BTN3A1 on target cells. BTN3A targeting agonist antibody
ICT01 is being developed in a multicentric Phase 1 and 2 study called EVICTION by
Imcheck Therapeutics. Recently the biology of Vγ9Vδ2 T cells has recently undergone
a new paradigm with the identification of BTN2A1 as the direct ligand for Vγ9 chain
of γδ TCR. BTN2A1 seems to be mandatory for Vγ9Vδ2 T cell activation but its precise
role in modulating functions of Vγ9Vδ2 T cells remains unknown.
Aims: Here, we show we show that Vγ9Vδ2 T cells exert cytolytic functions against
ALL cell lines and primary ALL blasts and that Vγ9Vδ2 T cell cytotoxic activity is
enhanced after treatment with a unique agonist mAb targeting BTN2A1 called 107G3B5.
Mechanistically, anti-BTN2A1 enhances interactions between Vγ9Vδ2 T cells and target
cells and improves the binding of Vγ9Vδ2 TCR on target cells.
Methods: 15 hematological cancer cell lines and PBMC from 17 adults ALL patients at
diagnosis (7B-ALL, 7T-ALL and 3Ph+ ALL) were tested in functional assays.
We quantified the relative surface expression of BTN2A (using 107G3B5 and 7.48 mAbs)
and BTN3A (using 20.1 and 108.5 mAbs) on cell lines and primary ALL blasts. In parallel,
allogenic Vγ9Vδ2 T cells functions against cell lines and primary ALL blasts were
evaluated. ALL samples were also tested for their expansion capacities after 14 days
of PBMC culture in presence of Zoledronate. Interactions of Vγ9Vδ2 T cells with target
cells was investigated using a 3D imaging in real-time by holo-tomographic microscopy.
Binding assays were realized using a recombinant tetramerized Vγ9Vδ2 TCR.
Results: We showed that Vγ9Vδ2 T cells exert spontaneous cytotoxicity against hematological
cell lines and primary ALL blasts with a heterogeneous susceptibility depending on
the target. We demonstrated that anti-BTN2A1 agonist mAb (107G3B5) significantly enhanced
Vγ9Vδ2 T cells mediated apoptosis, in comparison to control condition. This effect
was increased over time even for the less spontaneously susceptible cells. We confirmed
these observations with autologous Vγ9Vδ2 T cells expanded from 4 ALL patients at
diagnosis for which effector functions were increased after treatment with 107G3B5.
In live microscopy, 107G3B5 enhanced interactions between target cells cocultured
during 24h with Vγ9Vδ2 T cells. Finally, we observed that 107G3B5 strongly increased
binding of a recombinant Vγ9Vδ2 TCR to target cells.
Summary/Conclusion: Here, we demonstrated that targeting BTN2A1 led to improve Vγ9Vδ2
T cell antitumor response at molecular, cellular and functional levels. Our results
highlighted that Vγ9Vδ2 T cells exert cytolytic functions against ALL cells, both
in allogenic and autologous setting and demonstrated that BTN2A1 targeting with our
unique agonist mAb could potentiate effector activities of Vγ9Vδ2 T cells against
ALL blasts. These results indicate that the sensitization of leukemic cells can be
induced by activation BTN3A as reported previously as well as with BTN2A1 mAbs.
These findings could be of great interest for the design of innovative Vγ9Vδ2 T cells-based
immunotherapy strategies for treating ALL that could be extended to other cancer types.
P331: T-CELL RECEPTOR REPERTOIRE DIVERSITY AND L-ASPARAGINASE HYPERSENSITIVITY IN
CHILDHOOD ACUTE LYMPHOBLASTIC LEUKEMIA
S. Lee1,*, Z. Li1, E. Lim2, E. Chiew2, A. M. Tan3, H. Ariffin4, J. J. Yang1, A. Yeoh2
1St. Jude Children’s Research Hospital, MEMPHIS, United States of America; 2National
University of Singapore; 3KK Women and Children’s Hospital, Singapore, Singapore;
4University of Malaya Medical Centre, Kuala Lumpur, Malaysia
Background: Asparaginase is an indispensable component of therapy of acute lymphoblastic
leukemia (ALL). Treatment with asparaginase is associated with a plethora of adverse
effects, amongst which the occurrence of hypersensitivity is one of the most common.
Hypersensitivity affects the ability to maintain dose intensity which in turn results
in poorer outcomes, especially if there is no suitable replacement.
The basis of L-asparaginase hypersensitivity is undoubtedly immune-mediated. T-cell
recognition of antigens is arguably one of the most important facets for establishing
an immune response, of which a crucial component is the repertoire and diversity of
T-cell receptors (TCR). The exact aspects of T-cell immunity underpinning association
with asparaginase allergy has yet to be characterized.
Aims: To evaluate the associations of TCR repertoire and its diversity with asparaginase
hypersensitivity in children with ALL.
Methods: We longitudinally profiled the TCR repertoires in 67 children with ALL treated
on the frontline Ma-Spore ALL 2010 trial, all of whom receive native L-asparaginase
in induction, consolidation, and reinduction. Only patients who developed grade 2
or higher reactions (CTCAE 3.0) were included in this study. These children had bone
marrow cells sampled at 3 time-points – diagnosis, week 5 (post-induction), and week
12 (post-consolidation). We performed DNA-based TCR-sequencing on N=180 samples, and
evaluated TCR characteristics to determine their associations with asparaginase hypersensitivity.
Results: 12 out of 67 children (17.9%) had allergy in our cohort, representing 16
episodes of varying severity (Figure 1A). First, we evaluated the association of TCR-repertoire
diversity early on in treatment with the occurrence of subsequent allergy. We found
that a higher TCR diversity at all time-points was significantly associated with late
allergy (P=0.03 for Shannon’s entropy, P=0.01 for inverse Simpson’s index, Figure
1B). This TCR diversity was characterized by a higher proportion of infrequent clones
occurring <0.5% (P=0.008, Figure 1C). Patients with allergy had significantly lower
proportion of shared clonotypes (P=0.003, Figure 1D). Examining the dynamic changes
of the TCR repertoire between diagnosis and week 5 for each patient, we found that
patients with allergy had a much less similar clonotypic set between timepoints compared
to non-allergic patients (P=0.003, Figure 1E). In fact, patients who demonstrated
a higher variability in their clonotypes between timepoints (i.e. similarity coefficient
of <0.05) had an 8.1-fold risk of allergic event (95% CI 1.7 – 39.1, P=0.001, Figure
1F). Evaluating the TCR-repertoire before and after an allergy, we found that there
was convergence of TCR towards a common antigen (Figure 1G), where clonotypes became
more closely related after an allergy (Figure 1H). Finally, we found that allergic
patients had a lower proportion of public clonotypes (i.e. public clonotypic sequences
found on VDJDB database) compared to non-allergic patients (P=0.02, Figure 1I), supporting
the “hygiene hypothesis” as a predisposing factor in development of allergy.
Image:
Summary/Conclusion: Higher TCR-repertoire diversity is associated with the risk of
L-asparaginase hypersensitivity. This more diverse immunological mileau is characterized
by infrequent, dissimilar, and less shared clonotypes, which increases the chance
of a reactive clonotype matching to the allergen. Understanding the immunological
basis of T-cell response in allergy may help in developing strategies to mediate this
toxicity, to further improve outcomes in children with ALL.
P332: MIR-625-5P IS NOVEL CANDIDATE ONCOMIR IN T-CELL ACUTE LYMPHOBLASTIC LEUKEMIA
IMPLICATED IN REGULATION OF APOPTOSIS VIA REPRESSION OF HARAKIRI
N. Maćkowska-Maślak1,*, M. Drobna-Śledzińska1, R. Jaksik2, M. Kosmalska1, M. Witt1,
M. Dawidowska1
1Institute of Human Genetics Polish Academy of Sciences, Poznan; 2Institute of Automatic
Control Silesian University of Technology, Gliwice, Poland
Background: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive subtype of
ALL, arising from T-cell precursors. miRNAs are non-coding RNAs, contributing to leukemogenesis
by involvement in key cellular functions such as proliferation, apoptosis, and signaling.
We previously found in miRNA transcriptome analysis that miR-625-5p is overexpressed
in T-ALL patients, thus is a potential oncomiR in T-ALL. Here, we confirm the implication
of miR-625-5p in the regulation of apoptosis in T-ALL cells in vitro. We show that
it acts via repression of HARAKIRI (HRK). HRK was originally identified as a proapoptotic
gene induced by reduced levels of cytokine in hematopoietic cells and repressed by
the expression of death-repressor proteins. Although its proapoptotic function is
well described, little is known about the post-transcriptional mechanisms that may
participate in HRK inactivation.
Aims: The aim of this study was to investigate the mechanism of miR-625-5p oncogenic
action via unraveling its targetome and functional effects in T-ALL in vitro.
Methods: T-ALL JURKAT cells (with high endogenous miR-625-5p level) were transduced
for stable inhibition of this miRNA. To evaluate the influence of miR inhibition on
growth of T-ALL cells, we used flow cytometry GFP competition assay and CCK8 proliferation
assay. To identify the genes mediating the effect of miR-625-5p, we subjected JURKAT
cells to Ago2-RIP-seq. We used magnetic beads coated with anti-AGO2 antibody to immunoprecipitate
RNA bound to RISC complexes. Ago2-IP and total RNA fractions were sequenced (polyA
RNA-seq, 150 bp reads, 60M PE reads/sample, Illumina NovaSeq 6000). Transcripts depleted
in Ago2-IP fraction upon miR inhibition were screened for the presence of miR-625-5p
binding sites. Luciferase assay was performed to confirm direct interaction between
hsa-miR-625-5p and 3’UTR of HRK gene. The changes of HRK protein level upon miR-625-5p
inhibition were evaluated via Western Blot. To estimate the apoptosis rate in T-ALL
cells we used two flow cytometry assays based on Annexin V and Caspase 3/7 staining.
Results: Inhibition of miR-625-5p decreased the growth of JURKAT cells. In RIP-seq
analysis we identified 384 transcripts depleted in RISC by at least 20% upon miR-625-5p
inhibition and having at least one putative 3’UTR binding site for this miRNA. Overrepresentation
analysis performed for these genes indicated their involvement in the apoptotic process.
Among transcripts depleted in RIP-seq with predicted binding sites for miR-625-5p,
we found HRK gene, previously reported to have proapoptotic activity. We postulate
that repression of this gene by overexpressed miR-625-5p contributes to decreased
apoptosis and to growth advantage of T-ALL cells.
To further confirm that miR-625-5p affects growth of T-ALL cells via negative regulation
of apoptosis, we performed apoptotic assays in JURKAT cells and showed that inhibition
of this miRNA increased apoptosis rate, in line with our hypothesis on the oncogenic
potential of miR-625-5p.
We confirmed the direct interaction between miR-625-5p and HRK 3’UTR by luciferase
assay. Additionally, in Western blot a subtle (approaching statistical significance)
increase of HRK protein upon miR-625-5p inhibition was observed, supporting our notion
that this miRNA is indeed a negative regulator of HRK.
Image:
Summary/Conclusion: miR-625-5p is a novel candidate oncogenic miRNA in T-ALL, implicated
in the negative regulation of apoptosis. Post-transcriptional repression of the proapoptotic
HRK gene is a putative mechanism contributing to oncogenic properties of this miRNA
when overexpressed in T-ALL cells.
P333: MULTI-COHORT GENE EXPRESSION RESOURCE FOR AUTOMATIC SUBTYPING OF B-CELL ACUTE
LYMPHOBLASTIC LEUKEMIA
V.-P. Mäkinen1,*, J. Rehn1, J. Breen2, D. Yeung1, D. White1
1South Australian Health and Medical Research Institute; 2University of Adelaide,
Adelaide, Australia
Background: Acute lymphoblastic leukemia (ALL) is the most common childhood cancer
and comprises multiple distinguishable genomic subtypes. RNA sequencing provides a
functional snapshot that we propose will provide rapid diagnostic information for
most ALL subtypes and deep biological insight to guide diagnostic efforts of atypical
cases.
Aims: The aims of this study were to elucidate the predictive associations between
mRNA-seq profiles and independently verified genomic lesions, and to develop easy-to-interpret
transcriptome-wide biomarkers for ALL subtyping in the clinical setting.
Methods: A total of 1,279 ALL patients were included in the discovery dataset from
six publicly available North American cohorts and 767 Australian ALL patients were
included as an external validation cohort. Additional publicly available mRNA-seq
data (n = 1,160) were used for quality control modelling. A novel batch correction
method was introduced and applied to adjust for cohort differences. Three machine
learning models (random forest, projections to latent structures and nearest Euclidean
neighbor) were trained with the discovery set and validated in the Australian dataset.
Results: Out of 18,503 genes with usable expression, 11,830 (64%) were confounded
by cohort effects and excluded. There were no substantial differences between the
machine learning methods in the validation set; we chose the nearest neighbor technique
as the simplest alternative. Six ALL subtypes (ETV6-RUNX1, KMT2A, DUX4, PAX5 P80R,
TCF3-PBX1, ZNF384) that covered 32% of patients were robustly detected by mRNA-seq
(PPV ≥ 87%). Five other frequent subtypes (CRLF2, hypodiploid, hyperdiploid, PAX5
alterations and Ph-positive) were distinguishable in 40% of patients, although overlapping
transcriptional profiles led to lower accuracy (52% ≤ PPV ≤ 73%). Based on these findings,
we developed the Allspice R package that predicts ALL subtypes and driver genes from
unadjusted mRNA-seq read counts as encountered in real-world settings.
Image:
Summary/Conclusion: We demonstrated high diagnostic accuracy for a fully automated
mRNA-seq method for a third of ALL patients and additional contribution to diagnostic
efforts for patients with inconclusive or mixed genomic characteristics.
P334: CHARACTERIZATION OF A DUX4-R INHIBITOR AS A POSSIBLE TREATMENT FOR ACUTE LYMPHOBLASTIC
LEUKEMIA
S. Mara1,*, V. Runfola1, M. Pannese1, C. Caronni1, R. Giambruno1, D. Campolungo1,
C. Ghirardi1, D. Gabellini1
1Division of Genetics and Cell Biology, IRCCS San Raffaele Hospital, Milan, Italy
Background: Acute lymphoblastic leukemia (B-ALL) is the most common pediatric cancer
and the major cause of cancer-related death before the age of 20. In up to 10% of
cases, the disease is caused by translocation of the Double Homeobox 4 (DUX4) transcription
factor to the immunoglobulin heavy chain (IGH) locus, giving rise to rearranged DUX4
(DUX4-r), maintaining the DNA binding domain (dbd) but with a rearranged C-terminus.
Contrary to wild type DUX4, which induces apoptosis, DUX4-r acquires transforming
ability in cellular and animal models. Through proteomics, we identified a potential
DUX4-r inhibitor (iD) for its ability to bind directly to DUX4-r dbd.
Aims: The goal of my project is to test the antileukemic potential of iD in cellular
and animal models of DUX4-r leukemia.
Methods: Human and murine cell lines and primary murine bone barrow progenitor cells
are lentivirally-transduced to induce the expression of DUX4-r alone or in combination
with iD, and the effects on proliferation, transformation, and B lymphoid differentiation
potential are being evaluated.
To establish a B-ALL model, DUX4-r PDX cells have been expanded in NSG mice. These
cells have been transduced with GFP or iD fused to GFP (GFP-iD) and injected into
new NSG recipients for expansion. Serial xenograft transplantation of the purified
transduced cells have been performed to test iD-dependent effect on PDX cells expansion
and leukemia latency in mice. Human markers and GFP expression were monitored over
time in peripheral blood (PB) to assess disease progression.
Results: We confirmed that DUX4-r can transform NIH-3T3 murine fibroblasts and inhibits
B-cell differentiation of primary murine bone marrow progenitor cells in vitro. We
found that iD decreases DUX4-r dependent transactivation. Moreover, lentiviral expression
of iD in B-ALL NALM6 cells (carrying an endogenous DUX4-r translocation) reduces the
levels of DUX4-r targets and impairs cell proliferation compared to transduced control
cells. Preliminary results show that PDX cells expressing GFP-iD expand less and have
a delayed PB engraftment compared to GFP control cells. Moreover, we noticed that
in iD cohort only contaminant GFP-ve PDX cells take over in vivo despite purification
by FACS sorting of transduced cells.
Summary/Conclusion: So far, iD expression is associated with reduced proliferation
of human DUX4-r leukemia cells and reduced DUX4-r targets expression. Pre-clinical
validation of iD could identify effective therapeutic approaches for the treatment
of DUX4-r B-ALL patients.
P335: GENOMIC DETERMINANTS OF THERAPY RESPONSE IN ETV6-RUNX1 LEUKEMIA
L. Oksa1,*, S. Moisio2, K. Maqbool3,4, H. Foroughi3,4, R. Kramer2, A. Nikkilä1, V.
Zachariadis5, M. Enge5, K. Vepsäläinen6, J. Duque-Afonso7, J. Hauer8, V. Wirta3,4,
O. Lohi1,9, M. Heinäniemi2
1Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine
and Health Technology, Tampere University, Tampere; 2The Institute of Biomedicine,
University of Eastern Finland, Kuopio, Finland; 3Clinical Genomics facility, Science
for Life Laboratory; 4Department of Microbiology, Tumor and Cell biology; 5Department
of Oncology-Pathology, Karolinska Institute, Stockholm; 6Department of Pediatrics,
Kuopio University Hospital, Kuopio, Sweden; 7Department of Hematology/Oncology/Stem
Cell Transplantation, Faculty of Medicine, University of Freiburg Medical Center,
Freiburg; 8School of Medicine; Department of Pediatrics, Technical University of Munich,
Munich, Germany; 9Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
Background: Acute lymphoblastic leukemia (ALL) is the most common cancer in children,
with about 3500 children diagnosed yearly in Europe. A quarter of cases harbor the
ETV6-RUNX1 (E/R) fusion gene. E/R leukemias are usually classified into low risk group.
However, a fraction of patients still encounters disease recurrence. The increased
relapse risk has been linked with a non-optimal initial therapy response as measured
by minimal residual disease (MRD).
Aims: The aim of this study is to identify genetic differences between E/R cases stratified
by MRD and to identify potential mediators of poor therapy response.
Methods: We analyzed the genetic landscape of E/R leukemia in a discovery cohort of
35 patients treated with the NOPHO2008 protocol using whole genome sequencing (WGS).
Targeted sequencing was included in validation, including additional cohorts. Patients
were classified into three categories (slow, intermediate or fast responder) based
on MRD at the end of induction therapy (day 29). Variant callers available in the
Balsamic workflow were used to detect DNA alterations and subclonal events analyzed
using Battenberg and FastClone tools. Results were compared against published recurrent
ALL mutations, bone marrow single cell RNA-sequencing and genome-wide CRISPR-screens
in the E/R+ cell line REH, with vincristine, cytarabine, methotrexate, L-asparaginase,
maphosamide, daunorubicin and 6-mercaptopurine sensitivity. Nalm-6 screen data was
used for evaluating glucocorticoid response.
Results: We found comparable number of single nucleotide variants (SNVs), indels and
structural variants (SVs) between the MRD-categories. However, based on mutation signature
analysis, a significant negative correlation of MRD-level at day 15 was found with
Signature 2 (APOBEC), while day 29 MRD correlated positively with Signature 3 (failure
of DNA double-strand repair). Interestingly, the signature related to AID/APOBEC family
has been reported with oncogenic role in T-ALLs, where APOBEC3 family genes display
high expression during thymocyte development. We used single cell RNA-seq profiles
from healthy bone marrow to study the relevance of APOBEC3 expression during precursor
B-cell development, and showed that APOBEC3B is highly expressed in the dividing hematopoietic
precursor, pro-B and especially pre-B cell stages.
Well-known ALL-related driver genes did not differ between MRD categories, with the
exception of KRAS. Instead of activating mutations, we found prevalent deletions in
the fast responder category.
To focus specifically on drug response modulating genes, we focused on genes that
significantly increased or decreased sensitivity in genome-wide CRISPR screens (top
5% screen hits). Overall, each case harbored multiple DNA alterations in these genes.
Moreover, fast responder cases had increased number of hits in sensitizing genes.
Summary/Conclusion: Combining MRD analysis with WGS studies carries the potential
to reveal crucial information about sensitizing and resistance mutations and underlying
cellular mechanisms. The found DNA variants that associate with MRD-status can be
utilized in the design and implementation of personalized therapy in E/R+ ALL.
P336: A CHEMOTRANSCRIPTOMIC SCREENING IDENTIFIES THE REVERSAL OF GLUCOCORTICOID RESISTANCE
IN NOTCH1 MUTATED T-ALL
L. Pagliaro1,*, L. Moron Dalla Tor1, V. Federica2, P. Andrei3, L. Monica3, S. Kleissle4,
M. Neuenschwander5, A. Gherli1, E. Cerretani2, A. D’Antuono1, E. Simoncini1, A. Montanaro1,
G. Roti1
1Medicine and Surgery, University of Parma, Parma; 2University of Ferrara, Ferrara;
3University of Parma, Parma, Italy; 4Max Delbrueck Center for Moleculare Medicine
in the Helmholtz Association (MDC); 5Leibniz-Forschungsinstitut für Molekulare Pharmakologie
(FMP), Berlin, Germany
Background: Gain-of-function NOTCH1 mutations are the most common genetic abnormality
in T-cell Acute Lymphoblastic Leukemia (T-ALL), accounting for 55-60% of the cases.
Consequently, modulators of the Notch pathway, such as γ-secretase inhibitors (GSI),
would be expected to have clinical efficacy (Rao, Cancer Res 2009). However, their
application was limited by an excess of toxicity due to the suppression of wild-type
(WT) NOTCH1 proteins in normal tissue (Deangelo, JCO 2006; Doody, Alzherimer’s Res
Ther 2015).
In the past, we identified the Sarco-Endoplasmic Ca2+ ATPase (SERCA) as a gatekeeper
of the oncogenic Notch1 signaling. Thapsigargin (TG), a potent SERCA inhibitor (SI),
possesses an anti-NOTCH1-leukemia activity both in vitro and in vivo by preferentially
targeting mutated NOTCH1 proteins over WT ones (Roti, Cancer Cell 2013).
Aims: Since SIs display a favorable therapeutic index by targeting mutated NOTCH1
proteins, development of new SIs is under preclinical development (Marchesini, Cell
Chem Biol 2020). Thus, is important to establish molecular mechanisms portending resistance
in order to develop effective therapeutic strategies to overcome or prevent drug resistance.
Methods: We established drug-resistant clones (R) from parental ALL/SIL T-ALL cell
line using a stepwise increase in treatment dose with TG. We quantified the abundance
of differentially expressed (DE) genes (P adj<0.05) in the sensitive or resistant
cell line by RNA-seq and identified enriched pathways in the insensitive SI cell line.
In parallel, we screened a small molecule library of nearly 2500 bioactive compounds
(from the European Chemical Biology Library provided by EU-OPENSCREEN) in ALL/SIL
and ALL/SIL R. Compound hits were marked by their ability to inhibit each cell line
or both. Confirmatory experiments were completed in multiple T-ALL preclinical models.
Results: ALL/SIL R cells displayed a mutation (c.G770T -> p.Gly257Val) in the ATP2A2
gene occurring between the Asp254-Leu260 in the third SERCA2 transmembrane (TM) helix.
This variation avoids the efficient TG binding to the catalytic domain, resulting
in a diminished inhibitory effect. These cells showed an increase of TG IC50 >150-fold
compared to the parental line while remaining partially sensitive to CAD204520, an
inhibitor that binds SERCA in a pocket different from the one occupied by TG (Marchesini,
Cell Chem Biol 2020). This result suggests that part of the resistance mechanism is
transcriptionally mediated and virtually common to several SI. In SI-R cells, transcriptional
analysis of 6241 DE genes revealed enrichment in steroids synthesis and response pathways,
including cholesterol and lipid metabolism. Consistently, among small molecules targeting
the steroid hormone receptors subfamily 3, glucocorticoids (GC) scored among the top
hits in ALL/SIL R. While naïve cells were resistant to GC, R cells showed an enhanced
sensitivity at low nanomolar concentrations of several GC including dexamethasone,
clobetasol and fluticasone. This effect is at least in part mediated by the GC receptors
since the R cells displayed an elevated GC receptors protein level, as showed in western
blotting and RT-PCR, and the effect on viability were reversed by RU486 co-treatment.
Consequently, the association of SI plus GC displayed a synergistic effect in multiple
preclinical models, including steroid-resistant cell lines, primary leukemia cells,
and PDX models.
Summary/Conclusion: These findings suggest that SERCA-Ca2+ modulation mediates GC
and steroid signaling and that innovative SI can pharmacologically modulate glucocorticoid
resistance in T-ALL.
P337: LONG TERM INFECTION-DRIVES CHANGES IN THE BONE MARROW MICRO-ENVIRONMENT OF PAX5+/-
MICE PREDISPOSED TO B CELL PRECURSOR ACUTE LYMPHOBLASTIC LEUKEMIA
W. Liu1, O. Stencel2, Z. Lu2, C. Xu1, P. Lang1, F. Auer3, J. Hauer3, U. Fischer2,
A. Borkhardt2, A. Pandyra2,*
1Department of Molecular Medicine II; 2Department of Pediatric Oncology, Hematology
and Clinical Immunology, Heinrich-Heine University, Düsseldorf; 3Department of Pediatrics,
Technical University of Munich, Munich, Germany
Background: Pax5 heterozygosity (Pax5
+/-) in mice mimics germline or somatic Pax5 dysregulation (resulting in reduced Pax5
levels) observed in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL).
While a link between general non-specific infectious exposure and BCP-ALL has been
demonstrated in the Pax5
+/- mouse model, the effects of a specifically tractable infection are poorly understood.
In particular, little is known about potential interactions of pre-leukemic early
progenitor B cell populations and immune cells present in the bone marrow microenvironment
(BME) during an infection.
Aims: We aimed to characterize the short-term and long-term effects of a viral infection
in the BME of the Pax5
+/- mouse model employing the Lymphocytic choriomeningitis virus (LCMV). LCMV is a
non-cytopathic virus that has been extensively utilized to investigate virus-induced
immunopathology, effector responses and immune tolerance.
Methods:
Pax5
+/- mice backcrossed to the C57BL/6J background (N10) were infected with LCMV. Using
flow cytometry, ELISA and plaque assay, innate and late adaptive immune responses
in infected Pax5
+/- and WT (Pax5
+/+) were assessed. Pre-leukemic early progenitor B cell populations, antigen-specific
CD8+ T cells and regulatory T cells (Treg) were evaluated in the BME. This was complemented
by short-term ex vivo assays where B cells were stimulated with differing immune stimuli.
Results: When we investigated immune responses in a short-term ex vivo culture system
using the bone marrow of Pax5
-/+ and WT mice, we found differences in the induction of the interleukin 7 receptor
(IL-7r) on immature Pro-B cells as well as the production of cytokines (IL-6 and TNF-▫▫)
in response to the toll-like receptor (TLR7) agonist R848 and the lymphocytic choriomeningitis
virus (LCMV) Docile strain. When we infected Pax5
-/+ and WT mice with LCMV-Docile, we found that innate (early interferon production)
and adaptive immune responses (tetramer positive CD8+ T cells and early viral titers)
were not intrinsically impaired in Pax5
-/+ mice. However, at day 90 post-infection, LCMV-specific T cells were present within
the BME and tetramer specific CD8+ T cells raised against the LCMV nucleocapsid protein
(np 396) were increased in the BM of Pax5
-/+ infected mice (Figure 1A). Furthermore, when we evaluated Major Histocompatibility
Complex Class II (MHC-II) expression on different B cell subsets in the BME of infected
and uninfected mice, we found that there was a significant upregulation in all immature
B-cell subsets (Pro-B, PreBI-II) in response to infection in the WT but not in the
Pax5
-/+ infected mice. Pax5
-/+ mice were susceptible to LCMV infection resulting in shorter long-term survival
compared to infected WT mice (Figure 1B).
Image:
Summary/Conclusion: This study demonstrates that the BME environment as well as survival
of pre-leukemic Pax5
-/+ mice is differently affected by LCMV infection supporting the observations that
immune dysregulation could contribute to the emergence of the leukemic clone1.
1Supported by the Carreras foundation project no. DJCLS07R/2019
P338: BRANCHED CHAIN AMINO ACID TRANSAMINASE 1 ASSOCIATES WITH THE KU70/KU86 HETERODIMER
AND MODULATES THE DNA DAMAGE RESPONSE IN T-CELL ACUTE LYMPHOBLASTIC LEUKEMIA CELLS
L. Di Martino1,*, V. Tosello2, S. Dalla Santa1, A. Papathanassiu3, G. Arrigoni4, P.
van Vlierberghe5, E. Piovan1,2
1Surgery, Oncology and Gastroenterology, University of Padova; 2Immunology and Molecular
Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy; 3Ergon Pharmaceuticals,
LLC, Washington DC, United States of America; 4Department of Biomedical Sciences,
University of Padova, Padova, Italy; 5Department of Biomolecular Medicine, Ghent University,
Ghent, Belgium
Background: T-cell acute lymphoblastic leukemia (T-ALL) is a biologically and clinically
heterogeneous disease mostly associated with NOTCH1 mutations that characterize over
60% of cases. Therapy-resistant or refractory T-ALL remains a major clinical challenge.
Branched Chain Amino Acid Transferase 1 (BCAT1), a cytosolic aminotransferase converts
BCAAs into their corresponding branched-chain α-keto acids and vice versa. Aberrant
expression of BCAT1 has been demonstrated in different tumor models, including acute
leukemia. However, its biological role in T-ALL remains to be elucidated.
Aims: The purpose of this study was to dissect the functional role of BCAT1 in T-ALL,
with particular emphasis on a putative link between BCAT1 and NOTCH1 in promoting
T-ALL initiation and progression.
Methods: We evaluated BCAT1 expression using RNAseq data from 264 T-ALL samples (ALL0434
protocol). BCAT1 transcript and protein levels were determined in T-ALL cell lines
(n=13) and patient derived xenografts (PDX; n=12). Metabolomics analysis (using 13C6
labeled Leucine) was performed following NOTCH1 inhibition. Further, five PDX samples
were treated with a γ secretase inhibitor (DBZ) to dampen NOTCH signaling. BCAT1 promoter
occupancy by NOTCH1 was determined by Chromatin Immunoprecipitation (ChIP) coupled
with qPCR. Tandem affinity purification (TAP) and mass spectrometry (MS) analysis
were used to identify BCAT1 interacting proteins from CUTLL1 T-ALL cells. Kinetics
of the DNA damage response (DDR) was evaluated in BCAT1 knock-down cells. Drug combination
experiments using the DNA damaging agent (etoposide) and a specific BCAT inhibitor
(ERG245) were performed in vitro and in vivo in PDX T-ALL models.
Results: We found variable BCAT1 expression levels amongst the different T-ALL molecular
subgroups, with higher expression levels in TLX1 (p<0.001), HOXA-TLX3 (p<0.05) and
NKX2-1 (p<0.01) subtypes. Considering recurrent genetic alterations, we found BCAT1
expression to be significantly higher (p<0.01) in NOTCH1 mutated cases compared to
un-mutated samples. Interestingly, NOTCH1 mutated PDX samples also showed higher BCAT1
expression compared to un-mutated cases. Metabolic studies disclosed that BCAAs oxidation
is decreased following NOTCH1 inhibition (reduction in 3-methyl-2 oxovaleric acid).
Further, inhibition of NOTCH1 signaling with DBZ decreased BCAT1 expression in numerous
T-ALL models. ChIP analysis and luciferase reporter assays suggest that NOTCH1 may
directly regulate BCAT1 expression. Functionally, TAP followed by MS analysis of BCAT1
interacting proteins disclosed that BCAT1 may be implicated in non-metabolic processes
such as DNA replication and repair and rRNA processing. Indeed, we found Ku70/Ku86
proteins to interact with BCAT1. Evaluation of the DDR following DNA damage, disclosed
that BCAT1 deficient cells present an accentuated but defective DDR which translates
into marked DNA damage leading to pronounced cell death. Consistently, a marked synergistic
therapeutic response was found between the DNA damaging drug etoposide and the BCAT
specific inhibitor ERG245 in numerous T-ALL models.
Summary/Conclusion: NOTCH1 may directly regulate BCAT1 expression in T-ALL cells.
BCAT1 is involved in sustaining genomic integrity following DNA damage through its
interaction with the Ku70/Ku86 heterodimer, known to mediate classical non-homologous
end-joining. Our results identify BCAT1 as a novel therapeutic target and suggest
that the combination between DNA double stand break inducing agents (such as etoposide)
and a BCAT inhibitor could be particularly useful in NOTCH1-mutant T-ALL cases.
P339: BOOSTING THE EFFICACY OF CD20-TARGETING IMMUNOTHERAPY IN B CELL ACUTE LYMPHOBLASTIC
LEUKEMIA
M. Poprzeczko1,*, K. Domka1,2, A. Pastorczak3, K. Fidyt2, L. Komorowski1, M. Winiarska2,
A. Dabkowska2, K. Siudakowska2, A. Wojciechowska2, E. Patkowska4, M. Firczuk1,2
1Laboratory of Immunology, Mossakowski Medical Research Institute Polish Academy of
Sciences; 2Department of Immunology, Medical University of Warsaw, Warsaw; 3Department
of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz; 4Department
of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
Background: CD20 is a B cell-specific surface protein that appears at the stage of
pre-B cells during B cell development. Currently, monoclonal antibodies (mAb) against
CD20 are commonly used for immunotherapy of mature B-cell malignancies. B cell precursor
acute lymphoblastic leukemia (BCP-ALL) originates from immature B cells in a bone
marrow. Overall, the effects of treatment with high-dose multi-agent chemotherapy
are good. However, for relapsed/refractory patients, novel treatment options (mainly
immunotherapy-based) targeting B cell-specific antigens are already used in clinics
and further optimized in preclinical models. Recent clinical trials conducted in Philadelphia
(Ph) chromosome-negative BCP-ALL patients with CD20 expression revealed that the outcome
of young adults may be improved by a combination of chemotherapy with rituximab, the
anti-CD20 mAb. However, in most patients, the expression of CD20 on BCP-ALL blasts
is heterogeneous and may be insufficient for effective treatment with CD20 mAb. Also,
the regulation of CD20 in BCP-ALL is not well defined.
Aims: The aim of this study is to identify drugs that are able to upregulate CD20
in BCP-ALL, in order to improve the efficacy of anti-CD20 mAbs in preclinical models
of the disease.
Methods: We tested the impact of 38 anti-cancer drugs used for the treatment of BCP-ALL
as well as tested in clinical trials on the CD20 surface levels after 48hrs of incubation.
For the in vitro studies, we employed cell lines and primary cells representing selected
high-risk subtypes (Ph-positive, Ph-like, B-other). The levels of CD20 were determined
by flow cytometry. Next, the potential of selected drugs to activate the anti-CD20
mAb, rituximab, was tested in a functional assay, Antibody-Dependent Cellular Cytotoxicity
(ADCC) with Human Peripheral Blood Mononuclear Cells (PBMC) or primary NK cells, isolated
from healthy donors.
Results: Based on the initial screening of cell lines, we selected 10 agents which
increased the level of CD20: three histone deacetylases inhibitors, one inhibitor
of MDM2 protein (p53 signaling pathway), one apoptosis inducer, three kinase inhibitors,
one mitosis inhibitor, and one antimetabolic agent. Importantly, we found that many
tyrosine kinases inhibitors and some retinoids significantly decreased CD20 levels
in BCP-ALL cell lines. Some of these findings were also confirmed in primograft BCP-ALL
blasts co-cultured with OP9 murine stromal cells. Furthemore, we found that five out
of ten drugs enhance antibody-dependent cytotoxicity of rituximab, in functional assay.
Moreover, two of the tested drugs increased phagocytosis of primograft BCP-ALL cells
by primary macrophages differentiated from human peripheral blood monocytes.
Summary/Conclusion: In summary, we identified agents upregulating CD20 levels in BCP-ALL
cells in vitro. More importantly, some of the selected drugs improved RTX-mediated
effector mechanisms such as ADCC and immunophagocytosis. Further studies aiming to
elucidate the mechanisms of CD20 upregulation and to validate our in vitro findings
in murine models are ongoing.
P340: IN VITRO AND IN VIVO EFFICACY OF A NOVEL KINASE INHIBITOR TARGETING JAK2 GENE
REARRANGEMENTS IN CHILDHOOD ACUTE LYMPHOBLASTIC LEUKEMIA
M. Quadri1,*, C. Saitta1, S. Palamini1, C. Palmi1, A. Biondi1, G. Cazzaniga1, G. Fazio1
1Centro di Ricerca Fondazione M. Tettamanti, Monza, Italy
Background: Although risk-based treatment is curative for 85% of children with B-cell
precursor acute lymphoblasticleukemia (BCP-ALL), relapse remains a leading cause of
mortality, urging the need of novel molecular targets. The JAK/STAT alterations represent
about 7% of the ‘Philadelphia-like’ cases. JAK2 gene encodes for a non-receptor tyrosine
kinase fundamental for hematopoiesis and itsmutations have been widely studied, whereas
JAK2 fusion genes are still poorly characterized.
Aims: This study aims to identify JAK2 fusion genes among BCP-ALL pediatric patients,
developing a target strategy in preclinical models.
Methods: We applied RNA Next Generation Sequencing to find JAK2 fusion genes in a
cohort of high risk BCP-ALL pediatric patients. Fusions were validated by RT-PCR and/or
FISH. In vivo expansion of patients’ cells has been carried out in NSG mice. After
drug treatments with JAK2 inhibitors, phosphoflow and apoptosis assays were done.
Results: We identified 10 pediatric cases carrying a JAK2 fusion with different partners,
where PAX5 gene was the only recurrent.After in vivo expansion of cells from 3 cases,
carrying PAX5::JAK2, ATF7IP::JAK2 and ZEB2::JAK2, we demonstrated that JAK2 signaling
was activeat basal level, through phosphorylation of JAK2 Y1007-1008 compared to cases
wild type for JAK2 and CRLF2 (+70%, two-tailed P value 0.03); and also compared to
P2RY8::CRLF2 rearrangements and JAK2 mutation(+40%, two-tailed P value 0.16). The
JAK2 downstream effectors pS727-STAT3 and pY694-STAT5 were also activated.
We targeted JAK2using CHZ868, a new class-II tyrosine kinase inhibitor (TKI) (Novartis,
Basel, CH). After 30 minutes and till48h, we appreciated a mean inhibition of -62%
of Y1007-1008 JAK2 residues in PAX5::JAK2, -22% in ATF7IP::JAK2 and -35% in ZEB2::JAK2.
Contemporarily, we observed a decrease of pS727-STAT3 (-35-50%) and pY694-STAT5 (-15-50%)
and the significant reduction of phosphorylation on PI3K pathway,downregulating PDPK1,
AKT, 4pEBP1 and pS6.
After 48h monotherapy treatment by CHZ868, we detected decrease in cell viability
(20-75% at IC50). In combination with dexamethasone, a further decrease of viability
was observed.
In the PAX5::JAK2 case, we also performed treatments with BIBF1120/Nintedanib, LCK
inhibitor (activated downstream to PAX5 fusions) and we observed a 20% reduction of
cell viability. Importantly, combination of BIBF1120 and CHZ868 showed a synergistic
effect (-45%, at IC50). Moreover, ruxolitinib caused autophagy as observed by higher
levels of LC3-II compared to untreated cells (+45%, p<0.01), with reduction of apoptosis.
Indeed, active caspase 3 increased after ruxolitinib and chloroquine (autophagy inhibitor)
combination(+20% vs ruxolitinib alone, p<0.01). Instead, CHZ868 alone or in combination
with chloroquine does not induce autophagy, with no effect on bothLC3-II and active
caspase 3 levels.
Finally, we demonstrated the in vivo efficacy of CHZ868 in PAX5::JAK2, ATF7IP::JAK2
and ZEB2::JAK2 patient-derived-xenografts. After two weeks of 30mg/Kg daily treatment
of CHZ868, we observed a significant reduction of leukemic CD10+/CD19+ cells both
in BM(-43-85%), spleen(-72-89%), CNS (-13-62%) and PB (-46-80%).Moreover, CHZ868 in
vivo treatment significantly reduced the phosphorylation of pJAK2 (-18-46%), pSTAT5
(-23-71%) and pAKT(Ser473) (-18-34%).
Summary/Conclusion: CHZ868 is a promising drugfor the treatment of JAK2 fusionsBCP-ALL.
Further studies will include combination with standard chemotherapy drugs, by reducing
the intensity and toxicity of chemotherapy.
P341: A NOVEL BISPECIFIC T CELL ENGAGER (UMG2-CD3) IS EFFECTIVE AGAINST CORTICAL-DERIVED
ACUTE LYMPHOBLASTIC LEUKEMIA
C. Riillo1,*, D. Caracciolo1, K. Grillone1, N. Polerà1, F. M. Tuccillo2, P. Bonelli3,
G. Juli1, S. Ascrizzi1, F. Scionti4, M. Arbitrio5, M. Lopreiato1, M. A. Siciliano6,
S. Sestito6, G. Talarico7, E. Galea7, M. C. Galati7, M. Rossi7, A. Ballerini8, M.
Gentile9, M. T. Di Martino1, P. Tagliaferri1, P. Tassone1,10
1Clinical and Experimental Medicine, Università Magna Graecia, Catanzaro; 2Istituto
Nazionale Tumori “ Fondazione Pascale” - IRCCS; 3Istituto Nazionale Tumori “ Fondazione
Pascale” - IRCCS, Napoli; 4Institute of Research and Biomedical Innovation (IRIB),
Italian National Council (CNR), Messina; 5Institute of Research and Biomedical Innovation
(IRIB), Italian National Council (CNR); 6Università Magna Graecia; 7Pugliele-Ciaccio
Hospital, Catanzaro; 8biovelocITA srl, Milano; 9Annunziata Hospital, Cosenza, Italy;
10College of Science and Technology,Temple University, Philadelphia, United States
of America
Background: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological
malignancy derived by T cell precursors and characterized by poor prognosis.The immunotherapy
has revolutionized the outcome of B cell acute lymphoblastic leukemia (B-ALL), but
the absence of tumor-specific T cell antigen hampers its efficacy in T-ALL. Therefore,
the development of novel immune-therapeutic options for the management of this orphan
disease is eagerly awaited. CD1a is a glycoprotein expressed on cortical T-ALL and
only on healthy thymocytes and Langerhans cells. Taking into account its safe pattern
of expression, CD1a might represent a valuable therapeutic target for thetreatment
of T-ALL.
Aims: With the aim to provide an effective immune-therapeutic strategy for T-ALL,
we developed a bispecific T cell engager (BTCE) derived from a novel UMG2 mAb that
recognize a previously uncharacterized CD1a epitope.
Methods: To evaluate the specificity of UMG2 binding to CD1a epitope, HEK293T cells
which do not express CD1a endogenously, have been used. In this regards, cells were
transfected with a plasmid that encode for CD1a or with an empty vector and UMG2 reactivity
has been evaluated by flow cytometry. To assess the unicity of UMG2 mAb binding, a
competitive binding assay between UMG2 and commercially available CD1a antibodies
has been performed. The UMG2 expression profile on peripheral blood cells from healthy
donors and on a panel of cortical T-ALL cells has been evaluated. To develop a UMG2-CD3
construct, an asymmetric 2 + 1 UMG2-CD3 bispecific T cellengager (BTCE) has been generated
by using knobs into hole technology. UMG2-CD3 T cell-redirected cytotoxicity has been
evaluated on HEK293T wild type, on HEK293T-CD1a+ and on patient-derived T-ALL cells,
co-cultured with peripheral blood mononuclear cells (PBMCs), CD4/CD8 depleted and
CD56 enriched lymphocytes at different E:T ratio.Moreover, T cell activation has been
assesed by flow cytometry. UMG2-CD3 anti-tumor activity against a CD1a+ T-ALL cells
has been evaluated in vivo. For this purpose,Hu-PBMCs NSG mouse model has been generated
and tumor growth has been assessed by fluorescent imaging probe via IVIS system.
Results: UMG2 mAb recognizes a novel CD1a epitope and does not compete with any of
the commercially available anti-CD1a mAbs with the exception of a partial competition
with NA1/34-HLK clone. A strong UMG2 reactivity has been observed on T-ALL cells,
while no binding has been found on normal blood cells. A concentration-dependent T
cell cytotoxicity on CD1a+ T-ALL cells co-cultured with PBMCs in the presence of UMG2-CD3
has been observed. Minimal UMG2-CD3 residual anti-tumor activity has been observed
in CD4/CD8 depleted and CD56 enriched lymphocytes. CD56 depleted and Fc-blocked BMCs
were able to induce an anti-T-ALL activity comparable to total PBMCs, demonstrating
that UMG2-CD3 could not recruit monocytes and NK cells through Fc-FcyR interaction.
Moreover, the concentration-dependent increase of i) T cell proliferation, ii) cytotoxic
degranulation marker (CD107a), iii) expression of cell surface activation markers
(CD25, CD69),and iv) pro-inflammatory cytokine secretion (IL-2, TNF-α, IFN-γ) has
been observed in the presence of UMG2-CD3. Importantly, in an in vivo immune-humanized
NSG mice model,UMG2-CD3 was able to significantly inhibit tumor growth and then conferring
the survival advantage of treated animals.
Summary/Conclusion: All our findings, demonstrated that UMG2-CD3 BTCE represents a
promising immune-therapeutic agent against T-ALL to be further investigated for clinical
translation.
P342: BASELINE GENE EXPRESSION ANALYSIS OF RELAPSED ACUTE B-LYMPHOBLASTIC LEUKEMIA
PATIENTS TREATED WITH INOTUZUMAB OZOGAMICIN
C. Sartor1,*, I. Vigliotta2, V. Robustelli1, G. Cristiano1, J. Nanni1, L. Zannoni1,
S. Parisi2, S. Paolini2, G. Marconi1,3, G. Martinelli3, A. Curti2, M. Cavo1,2, C.
Terragna2, C. Papayannidis2
1Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Istituto di Ematologia
Seràgnoli; 2IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia
“Seràgnoli”, Bologna; 3Istituto Scientifico Romagnolo per lo Studio e la Cura dei
Tumori (IRST) IRCCS, 47014, Meldola (FC), Italy
Background: Despite introduction of novel agents, relapsed/refractory B-cell acute
lymphoblastic leukemia (R/R B-ALL) carries dismal outcome. Inotuzumab ozogamicin (InO)
is a humanized anti-CD22 monoclonal antibody conjugated to calicheamicin approved
for R/R B-ALL able to obtain high CR rates but of short duration. Mechanisms underlaying
InO resistance are largely unknown.
Aims: To characterize the baseline differentially expressed genes in a series of R/R
B-ALL patients in relation with patient response to InO in order to individuate potential
pathways involved in resistance.
Methods: Gene expression profile of 18 R/R B-ALL patient samples was analyzed with
RNA-seq, before InO exposure. All patients received at least 1 InO course. Patient
population was divided in poor/non-responders (NR) and responders (R), based on InO
response. R were defined as patients with duration of response (DoR) ≥3 months after
bone marrow (BM) complete remission (CR) achievement. NR were defined either refractory
or with a DoR <3 months after CR achievement. The list of significant (p<0.05) differentially
expressed genes (DEGs) in NR, as compared to R whose absolute fold change (FC) was
≥2, was analyzed. P values were corrected with the Benjamini-Hochberg algorithm (false
discovery rate; FDR). Gene expression results were analyzed with QIAGEN Ingenuity
Pathway Analysis (IPA).
Results: Eighteen R/R BM samples of CD22-positive B-ALL were analyzed with RNA-seq.
Eight patients were defined as NR and 10 patients as R. Patient disease characteristics
in NR and R groups were homogeneous for age, sex, number of Philadelphia (Ph)-positive
ALL patients, duration of 1st remission, number of previous therapy lines including
hematopoietic stem cell transplantation (HSCT) and Blinatumomab. At the time of InO
therapy all patients had morphological BM relapse and all were CD22-positive. Median
CD22 expression percentage on leukemic blasts was 100% (range 100-100) in NR patients
and 100% (range 70-100) in R patients (p=0.177). Two patients in R group had a CD22
expression blast percentage of 70% and 76%. Median CD22-fluorescent intensity (CD22-FI)
in NR as compared to R was 75.38 (IQR 59.58, 89.51) and 136.51 (IQR 114.38, 151.57)
respectively, with significantly lower values in NR group (p = 0.04).
Overall, 370 genes were differentially expressed (p<0.05) in NR, as compared to R
patients, of which 32 were significantly differentially expressed (corrected p < 0.05,
FC ≥2). Thirty-one were down- and 1 was up-regulated in NR vs R. DEGs were involved
in basophil differentiation, carbon dioxide transport, erythrocyte and myeloid cell
development, heme metabolic and porphyrin-containing compound biosynthetic processes,
cation and cellular ion homeostasis. Both IPA upstream regulator and regulator effect
analysis identified the serine/threonine homeodomain-interacting protein kinase-2
(HIPK2) as predicted downregulated. The inhibition of HIPK2 was predicted to be the
causal upstream condition of the under-expression of six DEGs from the set (FECH,
ANK1, SCL4A1, EPOR, GATA1, HBZ) with activation Z score of -2.449 and p value of overlap
= 1.02E-09. No difference in terms of HIPK2 expression was appreciated in the two
groups, suggesting downregulation at post-transcriptional level.
Image:
Summary/Conclusion: A unique pattern of gene expression signature based on HIPK2 downregulation
was identified in poor responders to InO, providing potentially important insights
in mechanisms of resistance. HIPK2 downregulation needs to be further validated.
CT and CP contributed equally
P343: GENOMIC CHARACTERISATION OF B-OTHER ALL IN UKALL2003 PATIENTS BY NEXT GENERATION
SEQUENCING
C. Schwab1,*, R. Cranston1, S. Ryan1, E. Butler1, E. Winterman1, Z. Hawking1, M. Bashton1,
J. Murray1, A. Enshaei1, J. Gibson1, A. Vora2, A. Moorman1, C. Harrison1
1Leukaemia Research Cytogenetics Group, Newcastle University, Newcastle upon Tyne;
2Department of Haematology, Great Ormond Street Hospital, London, United Kingdom
Background: Incorporating genetics into risk stratification for the treatment of childhood
acute lymphoblastic leukaemia (ALL) has contributed to increased survival rates. About
30% of patients (B-other-ALL) harbour none of the known major chromosomal changes.
Recently, we estimated that up to two thirds of B-other-ALL can be classified into
novel subtypes using FISH and MLPA alone. We showed that ABL-class fusions and ERG
deletions were linked to prognosis (Schwab et al, BJHaem, 2022).
Aims: To characterise B-other-ALL in the UKALL2003 trial using next generation sequencing
(NGS), to evaluate the added benefits of these techniques and validate the prognostic
significance of B-other-ALL subtypes.
Methods: B-other-ALL patients were tested using whole genome sequencing (WGS) (n=158)
and bespoke targeted NGS (t-NGS) for the detection of abnormalities in 64 genes commonly
mutated or rearranged in B-ALL (n=180), 42 patients had both WGS and t-NGS. Data were
integrated with results from FISH, MLPA and cytogenetics.
Results: A representative cohort of 347 B-other-ALL was classified into subgroups,
including six with ≥20 cases (Figure).
Among patients tested by WGS, 92% (n=146/158) were classified compared to 75% (n=104/138)
tested by t-NGS. NGS-based approaches allowed for the detection of PAX5 P80R, IKZF1
N169Y and ZEB2 H1038R and fusion partner genes. Identification of DUX4-r was not possible
using t-NGS.
PAX5alt was the most frequently observed (n=90), including dic(9;20) (n=27), dic(9;12)
(n=11), PAX5 fusions (n=21), PAX5 mutation (n=11) and PAX5-ITD (n=12). Additionally,
11 patients were observed with PAX5 P80R mutations, classified as a separate subgroup
due to the associated gene expression signature. PAX5alt had an outcome similar to
B-other-ALL overall (overall survival (OS) at 10yrs 82% vs 86%). There was no significant
difference in outcome between PAX5 abnormalities, although an association with age
was observed: dic(9;20) was more commonly observed in children aged 1-4 (p< 0.001),
while both dic(9;12) and PAX5 P80R were seen in older children aged 10-15 years (p=0.005).
One subgroup with DUX4 rearrangements and/or ERG deletions (DUX4-r/ERG-d) (n=78) had
DUX4 involvement confirmed by WGS in 57 patients. In the remaining 21 patients ERG-d
were identified by t-NGS and/or MLPA. ERG-d is exclusive to DUX4-r patients, thus
can be used as a surrogate marker. In line with other studies, patients with DUX4-r
had a lower relapse rate (RR) (5%) and improved OS (96%) compared to other subgroups.
Patients classified as ETV6-RUNX1-like (n=21) were characterised by multiples abnormalities
of ETV6, including rearrangements (n=17) and/or deletion (n=12) as well as rearrangements
(n=7) and/or deletions (n=6) of IKZF1. An ETV6-RUNX1-like gene expression signature
was confirmed by RNA-Seq (n=6). No relapses or deaths were reported within this subgroup.
As previously reported, ABL-class fusions (n=25) were associated with an inferior
outcome compared to other subgroups (RR=63% and OS=50%). Patients with rearrangements
of CRLF2 (n=52) and ZNF384 (n=37) had similar outcomes to the cohort overall.
Image:
Summary/Conclusion: A combination of techniques has classified a representative cohort
of B-other-ALL patients from UKALL2003 into clinically relevant subgroups. The identification
of DUX4-r, subgroup defining mutations and fusion partner genes demonstrates the value
of NGS-based approaches. We have confirmed the good and poor prognostic associations
of DUX4-r and ABL-class fusions, respectively, and identified ETV6-RUNX1-like subgroup
to have a good prognosis.
P344: THE DUAL BCL-2 AND BCL-XL INHIBITOR AZD4320 SHOWS ON-TARGET ACTIVITY IN ALL
AND ACTS SYNERGISTICALLY WITH MCL-1 INHIBITION
M. C. Wichert1, A. Niedermayer1, S. Enzenmüller1, K.-M. Debatin1, L. H. Meyer1, F.
Seyfried1,*
1Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center,
Ulm, Germany
Background: Targeting anti-apoptotic BCL-2 family proteins by BH3-mimetics has become
a promising treatment strategy in acute lymphoblastic leukemia (ALL). Heterogeneous
activity of the selective BCL-2 inhibitor venetoclax has been observed, but other
anti-apoptotic proteins including BCL-XL promote venetoclax insensitivity. In clinical
trials, targeting BCL-XL has previously resulted in a dose-limiting decrease of platelets.
AZD4320 was developed as a dual inhibitor of BCL-2 and BCL-XL, and its dendrimer conjugate
(AZD0466) was recently reported to demonstrate anti-tumor activity in hematological
cancer models, while showing only a transient thrombocytopenia.
Aims: In this study, the anti-leukemia activities of the dual BCL-2 and BCL-XL inhibitor
AZD4320 and of MCL-1-selective AZD5991 were evaluated and compared to the effects
of other BH3-mimetics (BCL-2-selective venetoclax, BCL-XL-selective A-1331852 and
MCL-1-selective S63845). The on-target activity of the inhibitors was functionally
characterized and combination effects were analyzed.
Methods: Cell viability assays were performed in ALL cell lines and patient-derived
xenograft (PDX) samples analyzing half maximal effective concentrations (EC50). Protein
complexes and expression of apoptosis regulators were analyzed by immunoprecipitation
and immunoblotting. Dynamic BH3 profiling using synthetic BH3-peptides was performed
to determine the dependency of ALL cells on BCL-2 family proteins. Combination effects
were assessed by dose-response matrix analyses.
Results: First, we determined the efficacy of the dual BCL-2 and BCL-XL inhibitor
AZD4320 and of the MCL-1 inhibitor AZD5991 for cell death induction in seven B-cell
precursor (BCP) ALL cell lines and in a series of 13 PDX samples, showing heterogeneous
responses. Interestingly, sensitivities of individual samples to both compounds were
not associated with each other. However, we found a significant correlation of sensitivity
to the dual inhibitor (AZD4320) with BCL-2 inhibition (venetoclax; N=13; rs=0.56;
p=0.049), but no association with BCL-XL inhibition (A-1331852). Analyzing activities
of all five BH3-mimetics including venetoclax, we found lowest EC50 values for AZD4320,
indicating particular sensitivity for this dual inhibitor as compared to the single
inhibitors (p<0.001).
Investigating dependencies of ALL cells on BCL-2 family proteins by dynamic BH3 profiling,
we found a shift towards MCL-1-dependence upon exposure to AZD4320, while AZD5991
induced an increased combined dependence on BCL-2 and BCL-XL, indicating potential
synergistic activity of triple inhibition. Using co-immunoprecipitation analyses,
we found that the exposure of ALL cells to AZD4320 reduced binding of both BCL-2 and
BCL-XL to BIM, confirming on-target activity. Moreover, AZD5991 reduced binding of
BIM to MCL-1. Accordingly, combined treatment with both inhibitors results in the
release of BIM and downstream apoptosis signaling. Assessing combinatorial treatment
in a primary PDX sample using multi-dose matrix analyses of both inhibitors revealed
a positive mean Bliss synergy score of +4.8 indicating synergistic activity. Importantly,
the highest synergism was found at low concentrations of both inhibitors, suggesting
efficacy at moderate concentrations, which could potentially be achieved in vivo.
Summary/Conclusion: In summary, our study demonstrates sensitivity, on-target activity
and synergism of the dual BCL-2 and BCL-XL inhibitor AZD4320 with inhibition of MCL-1,
thereby providing strong evidence for further clinical evaluation in ALL.
P345: AN IMMUNE SCREEN IDENTIFIES 5-NONLOXYTRYPTAMINE AS A NOVEL ANTI-CANCER AGENT
CAPABLE OF IMPROVING ANTI-TUMOR IMMUNITY IN COMBINATION WITH ANTI-PD1 THERAPY
P. Stachura1,*, W. Liu1, A. Wlodarczyk2, C. Xu1, S. Bhatia2,3, P. A. Lang1, A. Borkhardt2,3,
A. A. Pandyra2,3
1Department of Molecular Medicine II; 2Department of Pediatric Oncology, Hematology
and Clinical Immunology, Heinrich-Heine-University Dusseldorf; 3German Cancer Consortium
(DKTK), Partner site Essen/Düsseldorf, Dusseldorf, Germany
Background: Exploiting the immune system, particularly CD8+ T cells to eliminate tumors
has revolutionized the treatment landscape. However, despite the evident successes
of CD8+ T cell-targeting immunotherapies such as anti-PD1 checkpoint inhibitors, many
patients fail to respond especially those with poorly immunogenic tumors. Additional
obstacles to successful immunotherapy responses include therapy-induced toxicity,
lack of bio-markers of response, an immunosuppressive environment leading to T cell
dysfunction and exhaustion and poor immune infiltration highlighting a need for novel
combinatorial approaches to augment immunotherapeutic activity.
Aims: We aimed to uncover novel therapeutic agents capable of augmenting the anti-tumoral
responses of CD8+ T cells in ex vivo systems and in vivo syngenetic poorly immunogenic
tumor models.
Methods: To uncover novel potentiators of T cell anti-tumor immunity, we carried out
an ex vivo pharmacological screen, using Lymphocytic choriomeningitis virus (LCMV)-primed
splenic T cells that were combined tumor cells expressing the LCMV gp33 (B16) peptide
CTL epitope and incubated with an NIH 770 compound library. Hits that increased tumor-cell
killing when combined with LCMV-primed splenic T cells were further validated ex vivo.
The key candidate was assessed in vivo in C57BL/6J syngeneic tumor models. Immunocompromised
NSG mice as well as antibody depletion were used validate CD8+ T cell dependence.
In vitro and in vivo immunomodulatory effects were deciphered using flow cytometry,
immunoblot, CRISPR/Cas9 and histological analyses.
Results: We identified 5-Nonyloxytryptamine (5-NL), a serotonin receptor (HTR) agonist,
as increasing the ability of T cells to target tumor cells. In vitro, 5-NL induced
apoptosis in melanoma and childhood B-cell precursor acute lymphoblastic leukemia
(BCP-ALL) at low micromolar levels. 5-NL delayed tumor growth in vivo and the phenotype
was dependent on the hosts’ immune system, specifically CD8+ T cells as their depletion
abrogated the phenotype (Figure 1A-B). 5-NL’s pro-immune effects were attributed to
the upregulation of antigen presenting machinery in melanoma, BCP-ALL and other poorly
immunogenic tumors with low MHC-I/HLAA-C expression (Figure 1C). Importantly, the
upregulation of MHC-I/HLAA-C occurred without concomitant increases in PD-L1 expression
and was linked to upregulation of cAMP Response Element-Binding Protein (CREB) in
vitro and in vivo. As demonstrated through CRISPR/Cas9 HTR1D knockout, the immunomodulatory
and pro-apoptotic effects of 5-NL occurred independently of receptor target signaling.
5-NL was successfully combined with an anti-PD1 antibody in vivo for maximal tumor
growth inhibition.
Figure 1: The serotonin agonist 5-Nonyloxytryptamine (5-NL) was identified as potentiating
T cell mediated anti-tumor immunity. (A) C57BL/6J or (B) CD8 depleted C57BL/6J mice
were subcutaneously injected with 5 x 105 gp33 expressing tumor cells. 7 days post-tumor
injection, mice were randomized and treated daily 6.25 mg/kg of 5-NL or vehicle for
five consecutive days and tumor volume was measured (n = 9-12). H2-Db (MHC-I, murine
cell lines) and HLAA-C (human cells lines) were assessed by FACS analysis following
treatment with 5-NL for 24 hours (n = 3-9). Error bars in all experiments indicate
SEM; *P < 0.05 as determined by a Student´s t-test (unpaired, 2 tailed).
Image:
Summary/Conclusion: This study demonstrates novel therapeutic opportunities for augmenting
immune responses in poorly immunogenic tumors and increasing their responsiveness
to immunotherapies.
P346: THE ROLE OF CYTOKINES, TNF, TGFΒ1 AND NEUTROPHIL-TO-LYMPHOCYTE RATIO IN THE
COURSE OF THE ACUTE LEUKEMIA
V. Barilka1,*, V. Matlan2, S. Prymak3, O. Shalay4
1Laboratory of Immunology and Cytogenetics of Blood Neoplasms, SI “Institute of Blood
Pathology and Transfusion Medicine of National Academy of Medical Science of Ukraine”;
2Associate professor; 3•Department Oncology and Medical Radiology, Lviv National Medical
University named Danylo Galytsky; 4Head of the •Laboratory of Immunology and Cytogenetics
of Blood Neoplasms, SI “Institute of Blood Pathology and Transfusion Medicine of National
Academy of Medical Science of Ukraine”, Lviv, Ukraine
Background: Lymphocytes (Lym) and mature segmental neutrophils (Neu) together with
blasts are most commonly detected in the peripheral blood (PC) of patients (pta) with
acute leukemia (AL). Besides, Neu, Lym and blast cells are an important source of
transforming growth factor β1 (TGFβ1), tumor necrosis factor (TNF) production. Accumulation
of TNF, TGFβ1 in the plasma of AL pts is considered an unfavorable sign in AL. However,
the role of TNF, TGF β1 in the relationship with neutrophil-to-lymphocyte ratio (NLR)
in the course of AL is not definitively established.
Aims: Determine the role of cytokines, TNF, TGF β1 in the relationship with NLR to
assess the course of AL.
Methods: 42 adult AL pts were examined before treatment, who at the time of the examination
had blast cells in the peripheral blood (52.54 ± 5.63 x 109 / L). The NLR was calculated
from peripheral blood cell counts obtained at the time of diagnosis of AL by dividing
the Neu count by the Lym count. The median NLR (0.99) was used to dichotomize patients
into high- NLR and low- NLR groups. The concentration of TNF, TGF β1 was determined
by biological methods using sensitive cell lines L929 and CCL 64, respectively. Statistical
processing of the results was estimated by the value of p ˂ 0, 005. The correlation
between the indicators was estimated by the value of r.
Results: There were 25 people in the low-NLR group. Median age 66 years (36 - 81 years).
Among the pts were 9 people with ALL and 16 people with AML. Number of blasts 55,
83 ± 6.38 x 109 / L; New count was 11.73 ± 2.95 109 / L, Lym count - 21.27 ± 3.64
109 / L. The NLR index is 0.53 ± 0.09. In this group, 88% of pts died in the short
term without achieving remission, the remaining 12% of pts had short-term remission.
The concentration of TNF in plasma was 1.20 ± 0.13 ng / ml, TGF β1 - 4.02 ± 0.48 ng
/ ml, which in both cases was higher than in healthy individuals (p˂0.001 and p˂0.05,
respectively). TNF levels were positively correlated with Lym (r = 0.30) and blasts
(r = 0.40), suggesting a possible role for TNF in neoplastic clone survival.
There were 17 people in the high-NLR group, including 10 pts with ALL and 7 people
with AML who achieved remission. The median age is 38 years (from 16 to 68 years).
The number of blasts was 33.20 ± 9.67 x 109 / L and was significantly lower than in
the low-NLR group (p˂0.05); New count was 22.23 ± 0.10 x 109 / L; Lym count - 22.83
± 3.04 109 / L. The NLR was 1.24 ± 0.16. In this group, 94% of pts had remission.
Plasma TNF and TGF concentrations were lower than in the low-NLR group, apparently
due to lower blast content. However, no statistical difference in cytokine concentration
between the compared groups was found.
Summary/Conclusion: More favorable prognostic features in AL could be NLR, the median
of which is equal to or greater than 0.99. Under these conditions, a decrease in the
number of blast cells, the concentration of cytokines TNF and TGF β1 in the plasma
and the achievement of remission were observed. The data obtained may indicate a prognostic
role of Neu and Lym blood subpopulations in the course of AL and in the relationship
with cytokines, TNF and TGF β1, may be potential targets for the correction of treatment
of AL pts.
P347: COOPERATION OF TLX3 AND FLT3-ITD IN T-CELL ACUTE LYMPHOBLASTIC LEUKEMIA IS ENHANCED
BY TLE4 INACTIVATION
Q. Van Thillo1,2,3,*, L. Lauwereins1,2,3, S. Demeyer1,2,3, S. Provost1,2, N. Mentens1,2,
C. de Bock4, J. Cools1,2,3
1Center for Human Genetics, KU Leuven; 2Center for Cancer Biology, VIB; 3Leuvens Kankerinstituut,
KU Leuven - UZ Leuven, Leuven, Belgium; 4Children’s Cancer Institute, UNSW, Sydney,
Australia
Background: One fifth of T-cell acute lymphoblastic leukemia (T-ALL) cases is defined
by the aberrant expression of TLX3. Being similar to the closely related TLX1, its
expression is mainly associated with mutations in the JAK/STAT pathway (Liu et al.,
Nat Genetics 2017), which have been shown to cooperate with TLX1 in driving T-ALL
(Vanden Bempt et al., Cancer Cell 2018). However, it is not known whether TLX3 has
the same oncogenic role as TLX1. Additionally, TLX3 expression is also regularly found
with FLT3-ITD mutations, which are virtually absent in TLX1-positive cases (Liu et
al., Nat Genetics 2017), suggesting there are some important functional differences
between TLX3 and TLX1.
Aims: To set up a model of cooperation between TLX3 and FLT3-ITD and to elucidate
the oncogenic role of TLX3 in T-ALL.
Methods: We performed in vivo bone marrow transplant (BMT) assays by transducing lineage-negative
cells derived from C57BL/6 mice with the respective oncogenes using retroviruses.
Pro-T cell growth curves were performed in the absence of interleukin-7 (Il7) as described
previously (Van Thillo et al., Nat Communications 2021). Fluorescence activated cell
sorting (FACS) staining was performed on the MACSQuant VYB (Miltenyi Biotec) or Fortessa
(BD). RNA was prepared at the KU Leuven Genomics Core and 3’ prime end sequencing
(QuantSeq) was performed. Differential gene expression analyses were conducted using
the DeSeq2 R package (v1.22.0).
Results: The combination of TLX3 and FLT3-ITD in a constitutive BMT assay resulted
in a rapid leukemia with an immature myeloid immunophenotype (CD11b+, Gr1-, CD4-,
CD8-). The leukemia cells were positive for both FLT3-ITD (GFP) and TLX3 (mCHERRY),
indicating cooperation between the two oncogenes in vivo. Similarly, in our in vitro
pro-T-cell model, the TLX3 and FLT3-ITD double-positive cells outcompeted the single-positive
cells after withdrawal of Il7. Next, we performed RNA-sequencing in CreER (estrogen
receptor) pro-T cells 24 hours after activation of TLX3 expression by adding tamoxifen.
This showed both up- and downregulation of many genes, including Tle4 among the downregulated
genes. Interestingly, TLE4 expression was also lowest in the TLX3-subgroup of patients
(Liu. et al. Nat Genetics 2017). TLE proteins are transcriptional co-repressors that
can interact with the engrailed-homology 1 (Eh1) domain of transcription factors (Riz
et al. Biochem Biophys Res Commun 2009). Therefore, we inserted a point mutation in
the Eh1 domain of TLX3, resulting in a phenylalanine to glutamic acid substitution
at position 18, to impair this interaction. Strikingly, TLX3(F18E) in combination
with FLT3-ITD led to a proliferative advantage in pro-T cells after the omission of
Il7 compared to wild-type TLX3. On top of that, cells with TLX3(F18E) and FLT3-ITD
were able to grow in the absence of stem cell factor (Scf).
Summary/Conclusion: We demonstrate that TLX3 cooperates with FLT3-ITD in vivo and
in vitro in a pro-T-cell context. TLX3 downregulates the co-repressor Tle4 and inhibiting
the interaction between TLX3 and Tle4 reinforces the proliferative capacity of pro-T
cells in the absence of growth factors.
P348: MRK-560 AND DEXAMETHASONE ARE SYNERGISTIC IN THE TREATMENT OF T-CELL ACUTE LYMPHOBLASTIC
LEUKEMIA
C. Vandersmissen1,2,3,*, C. Prieto Fernàndez1, O. Gielen1, K. Jacobs1, I. Govaerts1,
J. Maertens4, H. Segers5, J. Cools1
1Human Genetics, KU Leuven; 2Cancer biology, VIB; 3Leuvens Kanker Institutie (LKI),
KU Leuven - UZ Leuven; 4Microbiology, Immunology and Transplantation, KU Leuven; 5Oncology,
KU Leuven - UZ Leuven, Leuven, Belgium
Background: NOTCH1 activating mutations are found in the majority of T-cell acute
lymphoblastic leukemia (T-ALL) cases and inhibition of NOTCH1 activation is therefore
a potential therapeutic option in T-ALL. Gamma-secretase inhibitors have been shown
to block NOTCH1 activation, but are also associated with dose-limiting toxicity, preventing
their use for T-ALL treatment. We have recently shown that a more selective inhibition
of the gamma-secretases with PSEN1-selective γ-secretase inhibitors, such as MRK-560,
retains a strong anti-leukemia effect with minimal gastrointestinal toxicities.
Aims: We wanted to investigate if MRK-560 would show synergy with currently used chemotherapeutic
drugs, such as dexamethasone, vincristine and doxorubicin, and if it could be safely
combined with chemotherapy for treating T-ALL patients.
Methods: Dose response curves were performed in DND-41 and SUPT-1 T-ALL cell lines,
which were treated with DMSO/MRK-560 alone or in combination with different concentrations
of chemotherapy. Apoptosis assay was conducted with annexin-V/PI staining. Human primary
T-ALL samples were transduced with a luciferase vector to obtain luciferase positive
xenografts in immunodeficient NSG mice that could be used for in vivo treatment. Mice
were treated for 3 weeks (5 days on – 2 days off) with vehicle, dexamethasone (5mg/kg,
IP), MRK-560 (15mg/kg, IP) or the combination. Leukemia progression was followed by
weekly blood sampling and bioluminescent imaging after injection of luciferin.
Results: Treatment with MRK-560 increased the sensitivity to dexamethasone in NOTCH1-dependent
T-ALL cell lines (DND-41 and SUPT-1) and we documented a strong synergy between these
drugs. The combination between MRK-560 and doxorubicin/vincristine showed only an
additive effect. Furthermore, we demonstrated that treatment with MRK-560 and dexamethasone
increased apoptosis 2.5 fold compared to single dexamethasone treatment. Dexamethasone
acts by activating the glucocorticoid receptor (NR3C1) which activates several target
genes including BIM, an inducer of apoptosis. Mechanistically, we found that mRNA
and protein levels for NR3C1 and BIM were significantly more upregulated after combination
treatment compared to single drug treatment. This was associated with strong downregulation
of HES-1 levels. These data indicate that MRK-560 synergizes with dexamethasone via
downregulation of HES-1 (downstream of NOTCH1), resulting in enhanced glucocorticoid
signaling with higher BIM expression levels and apoptosis. Finally, we also studied
the synergy between MRK-560 and dexamethasone in patient-derived xenograft models
of T-ALL. Bioluminescent imaging showed significant reduction in leukemia progression
in combination treatment compared to single treatments. Furthermore, combination treatment
(41 days) significantly prolonged survival compared to single MRK-560 (34 days), dexamethasone
(30 days) or placebo (16 days) treatment.
Summary/Conclusion: We have shown that the PSEN1-selective gamma-secretase inhibitor
MRK-560 can be safely combined with chemotherapy for the treatment of T-ALL patients
thereby demonstrating a synergistic effect between MRK-560 and dexamethasone. Mechanistically,
MRK-560 acts in a similar way as broad spectrum gamma-secretase inhibitors, but does
not show gastrointestinal toxicity.
P349: CLONAL ARCHITECTURE DISSECTING AT SINGLE-CELL RESOLUTION REVEALS MECHANISMS
OF CHEMOTHERAPY RESISTANCE AND RELAPSE IN T CELL ACUTE LYMPHOBLASTIC LEUKEMIA
J. Zhang1,*, Y. Duan1, Y. Zhang1, X. Zhu1
1Institute of Hematology & Hospital of Blood Diseases,CAMS &PUMC, Tianjin, China
Background: T-lineage acute lymphoblastic leukemia (T-ALL) comprises approximately
10-15% of pediatric ALL cases with distinct feature in biology and largely inferior
outcome compared to B-ALL. Growing evidence has reflected pivotal roles of clonal
evolution in T-ALL recurrence, but bulk sequencing may not serve as the perfect model
to reliably infer clonal heterogeneities and their immunomodulatory milieu during
leukemia development. In this study, single-cell sequencing was applied to uncover
leukemic clonal relationships with relapse throughout chemotherapy in T-ALL at a more
accurate resolution.
Aims: We performed bulk and single cell multi omics analysis for diagnostic and relapse
T-ALL, with the aim to dissect mechanisms of chemotherapy resistance and relapse in
T cell acute lymphoblastic leukemia.
Methods: We performed bulk whole-exome sequencing for sorted CD7+ BMMCs from 5 pairs
of diagnosis-relapse (Dx_Rel) samples, revealing a series of well-reported hotspot
mutations in T-ALL.
To dissect clonal diversities within and across the 5 Dx_Rel T-ALL pairs, we carried
out high-throughput droplet-based 5’-single-cell RNA-seq (scRNA-seq) and paired T
cell receptor sequencing (scTCR-seq).
Results: By performing unsupervised clustering of scRNA- seq profiles encompassing
10 samples, we identified 23 distinct T-lineage clusters (Cluster 0-22) based on the
two-dimensional UMAP visualization. In 2 out of 5 patients, diffusion map of T-lineage
sub-clusters between diagnostic and relapsed samples appeared to be almost identical,
while distinct shifts from diagnosis to relapse in the compositions have been observed
in the other 3 out of 5 patients.
We sought to further deconvolute the clonal architecture trajectory for T-ALL Dx_Rel
pairs. We observed that dominant diagnostic clones of 4 patients diminished or vanished
at relapse, sparing newly emerged subclones predominantly substituted at relapse.
We clearly depicted two distinct patterns of evolutionary trajectories in these 4
Dx_Rel pairs by comprehensively mapping hierarchical TCR clonotypes onto leukemic
clonotypes at single cell levels. Specifically, in T956 and T723, we observed significant
outgrowth of incidental diagnostic sub-clones at relapse, whereby surrogate TCR repertoires
correspondingly enumerated, suggestive of dynamic shifts in dominant clone over continuous
chemo-exposure. Whereas in T593 and T856, expanding clones at relapse were showed
up with completely different gene signatures from the diagnostic ones, but dominant
clones at diagnosis and relapse were surprisingly presented with identical TCR repertoires.
This was undoubtedly informative of leukemic “clonal drift” within which hypothetical
intrinsic transformation happened to the same subclones over persistent chemotherapy.
Image:
Summary/Conclusion: Collectively, our presented study accurately distinguished leukemic
cells from normal T cells in T-ALL at a single-cell resolution. By tracking transcriptomic
profiles within and across Dx_Rel T-ALL pairs, we further identified distinct clonal
evolutionary patterns, which may determine diversified fates of leukemic clones in
response to therapeutic pressures, extending significant implications for future precise
chemotherapies.
P350: BLINATUMOMAB AND DONOR LYMPHOCYTE INFUSION (DLI) FOR MOLECULAR RELAPSE AFTER
HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PEDIATRIC PATIENTS
F. Muriano1,*, F. Cacace2, V. Caprioli2, M. R. D’amico2, G. De Simone2, G. Giagnuolo3,
G. Menna3, F. P. Tambaro2
1Hematology, AOU Federico II; 2BMT UNIT; 3Hematology, AORN Santobono Pausilipon, Naples,
Italy
Background: Detection of measurable residual disease (MRD) after Hematopoietic Stem
Cell Transplantation (HSCT) is an early predictor of frank relapse. The role of DLI
as a pre-emptive treatment to prevent relapse is still debated. Blinatumomab, a CD3/CD19
bispecific T-cell engager molecule, is approved in adult patients with MRD and in
children with relapsed or refractory acute lymphoblastic leukemia (ALL).
Aims: We evaluated the safety and feasibility of blinatumomab plus DLI to treat MRD-positive
pediatricALLafter HSCT. Here, we report our experience of three patients with post-transplant
MRD-positive ALL who received blinatumomab and DLI.
Methods: Three pediatric patients with high-risk B-ALLwere treated according to AEIOP
ALL 2009 and AIEOP ALL 2017 protocols and referred for allo-HSCT. Patients became
MRD-positive after allo-HSCT and received blinatumomab with DLI.Patients’ characteristics
are summarized in the Table.
Results: Patient 1 developed CNS relapse during maintenance, then received salvage
therapy according to the INTREALL HR 2010 and 1 course of blinatumomab, achieving
MRD-positive CR1.Patient 2, a 16-year old female, had secondary ALL and achieved CR1
after 1 course of chemotherapy and subsequently received 2 courses of blinatumomab,
due to chemotherapy-associated toxicity and achieved MRD-undetectable CR1. Patient
3, a 17-year old male was primary refractory to 2 lines of chemotherapy and blinatumomab,
received 2 courses of inotuzumab and achieved an MRD-positive complete remission (CR).All
patients proceeded to allo-SCT and were MRD-positive at 8, 31 and 14 weeks post-allo-SCT,
respectively. All patients discontinued immunosuppressive therapy and received 2,
1 and 4 courses of DLI with blinatumomab, respectively. Blinatumomab was started one
week after each DLI, was given at a dose of 15-28cg/kg for a total of 4 weeks each
course. Only patient 3 experienced Grade 2 liver GVHD. None of the patients experienced
serious adverse events requiring blinatumomab discontinuation. Patient 3 developed
isolated CNS relapse after 4 courses of combined DLI plus blinatumomab; patient 2
achieved and remains in MRD-undetectable CR with current follow up and a patient 1
achieved MRD-negative CR after two courses of combined treatment and CNS relapse.
Image:
Summary/Conclusion: The role of cellular therapy with DLI is still debated in ALL.
Most post-transplant relapses are associated withloss of surface HLA bythe leukemia
cells, hence DLI alone has limited efficacy in patients with post HSCT relapse. Blinatumomab
directs T cells to bind CD19 present on malignant B cells and engages CD3 on T cells
causing activation and inducing cytotoxicity against ALL blasts. We, therefore, tested
the efficacy of blinatumomab plus DLI in 3 MRD-positive patients with ALL after allogeneic
HSCT. The use of blinatumomab allowed recruitment of fit donor-derived T lymphocytes
(not exposed to immune suppressive agents) against ALL B cells. This hypothesis is
supported by the fact thatpatient 3, who received blinatumomab pre-HSCT and had disease
progression, probably due to lack of T cells showed by flow cytometry, achieved MRD-undetectablestatus
after receiving DLI plus blinatumomab post-transplant. Twopatients reached stable
MRD-undetectablestatusand 2 subsequently developed CNS relapse; none received CNS
prophylaxis post-HSCT. We hypothesize that blinatumomab plus DLI can clear the hematologic
disease, but it is not effective in preventing CNS relapse. Although very interesting,
this hypothesis should be confirmed in a larger number of patients.
P351: DOSE-ADJUSTED EPOCH + INOTUZUMAB OZOGAMICIN (DA-EPOCH-INO) IS SAFE AND ACTIVE
IN ADULTS WITH RELAPSED/REFRACTORY (R/R) B LYMPHOBLASTIC LEUKEMIA (B-ALL): INITIAL
RESULTS OF A PHASE I TRIAL
R. Cassaday1,2,3,*, K.-L. Garcia1, T. Gooley2, C. Martino3, M.-E. Percival1,2,3, A.
Halpern1,2,3, V. Oehler1,2,3, J. Abkowitz1,2,3, R. Walter1,2,3, C. Ghiuzeli1,3, E.
Estey1,2,3
1University of Washington; 2Fred Hutchinson Cancer Research Center; 3Seattle Cancer
Care Alliance, Seattle, United States of America
Background: Despite new drugs, survival with R/R B-ALL remains poor. Combination therapies
may improve outcomes, but none are proven superior, and toxicities may pose limits.
InO has been added to low-intensity chemotherapy, but the risks/benefits of such combinations
are poorly defined. As we recently found DA-EPOCH to be effective for untreated ALL,
we here tested the addition of InO to DA-EPOCH (#NCT03991884).
Aims: The primary objective was to estimate the maximum tolerated dose (MTD) of InO
when added to DA-EPOCH.
Methods: Eligible adults had R/R CD22+ B-ALL with ≥5% blasts in blood/marrow or ≥1
site of extramedullary disease [EMD] ≥1.5 cm in diameter. No history of sinusoidal
obstructive syndrome (SOS) or chronic liver disease was allowed. Other key eligibility
criteria included bilirubin ≤1.5x upper limit of normal (ULN), AST/ALT ≤2.5x ULN,
creatinine ≤1.5x ULN, QTc ≤500 msec, and ECOG 0-2. All patients (pts) gave written
informed consent.
DA-EPOCH was given on Days 1-5 with G-CSF. Three dose levels (DL) of InO given on
Days 8 and 15 were studied in 28-day cycles: DL1, 0.3 and 0.3 mg/m2; DL2, 0.6 and
0.3 mg/m2; and DL3, 0.6 and 0.6 mg/m2 (respectively). In Cycles 2+, EPOCH was dose-adjusted
based on the hematologic nadir from the prior cycle. Up to 4 cycles were permitted.
Hepatic prophylaxis with ursodiol and CNS-directed therapy with intrathecal chemotherapy
was recommended.
MTD was the dose of InO yielding a true dose-limiting toxicity (DLT) rate of 33%.
Initial dose-escalation plan for InO was a 6 + 6 design, but after 5 pts enrolled,
this was modified to Bayesian Optimal Interval Design (BOIN) with target accrual of
24 pts. Secondary objectives were descriptions of treatment-related adverse events
(TRAEs) per NCI CTCAE v5; efficacy by rate of complete remission without or with incomplete
hematologic recovery (CR/CRi) and response at EMD sites per NCCN; measurable residual
disease negativity (MRD-) by multiparameter flow cytometry (MFC) and high-throughput
sequencing (HTS; clonoSEQ) of bone marrow aspirate; and survival.
Results: To date, 17 pts have been enrolled (Table). All are evaluable for DLT. 5
pts were treated at DL1; then per BOIN, 3 pts at DL2, and 9 pts at DL3. Median number
of cycles given was 2 (range, 1-3). There have been 4 DLTs: 1 at DL2 (20%; grade 4
sepsis) and 3 at DL3 (33%; grade 4 hyponatremia, grade 4 SOS, and prolonged pancytopenia).
The single case (6%) of SOS occurred after allografting and is ongoing. There have
been no treatment-related deaths. Grade 3+ non-heme TRAEs were seen in 13 pts (76%).
Those seen in >1 pt were infections (5 pts; 8 events), neutropenic fever (5 pts; 5
events), oral mucositis (3 pts; 4 events), and increased ALT (2 pts; 3 events); all
these events resolved. Other hepatic toxicity was generally mild. Of 10 pts who received
≥2 cycles, 5 (50%) escalated the dosing of EPOCH, while 2 (20%) de-escalated.
17 pts are evaluable for response. Of 13 with >5% blasts, 12 (92%; 90% confidence
interval 68-100%) achieved CR/CRi, 10 (77%) after 1 cycle. MRD- by MFC was achieved
in 9 (75%); 1 had prior InO. In the 5 pts with EMD, 4 (80%) responded including 3
CRs. To date, 6 pts (35%) have undergone allograft post-study. Median follow-up of
survivors is 7 mo, with 7 pts in ongoing remission beyond 6 mo.
Image:
Summary/Conclusion: DA-EPOCH-InO has a manageable toxicity profile with infrequent
DLTs and only 1 case of SOS in heavily pre-treated adults with B-ALL. Nearly all pts
with >5% blasts at enrollment achieved CR, with most being MRD- by MFC. Responses
in EMD were common. Enrollment is ongoing. Updated follow-up and responses by HTS
will be presented.
P352: SYMPTOMATIC VTE INCIDENCE IN ADULT ALL TREATED WITH PEG-ASPARAGINASE COMPARED
TO THE NATIVE L-ASPARAGINASE IN WITH AN ASPARAGINASE-BASED PROTOCOL
R. Chen1,*, J. Seki1, L. Liu1, E. Atenafu1, K. Yee1, A. Schuh1, V. Gupta1, S. Chan1,
A. Schimmer1, D. Maze1, M. Minden1, H. Sibai1
1University of Toronto, Toronto, Canada
Background: Venous thromboembolism (VTE) is a well-known complication in adults receiving
aspraginase (ASNase)-based chemotherapy for adults in acute lymphoblastic leukemia,
including the ASNase-intensive Dana-Farber Cancer Institute (DFCI) 91-01 protocol.
Previous studies have shown an VTE incidence of up to 41% in patients receiving the
DFCI protocol and up to 29% with prophylactic anticoagulations. Since 2019, native
L-asparaginase is no longer available in the Canada such that pegylated (PEG)-ASNase
has gradually replaced native e.coli ASNase (L-ASNase) as part of the DFCI protocol
at our centre.
Aims: The object of this study is to evaluate the incidence of VTE between patients
who received PEG-ASNase vs L-ASNase, with emphasis on the highest risk periods of
induction and intensification phases.
Methods: A single centred retrospective cohort study of 249 adult patients with Philadelphia
chromosome negative (Ph-) between 2011 to 2021 were conducted. There were 178 patients
received L-ASNase between 2011 to 2019 and 71 patients received PEG-ASNase between
2018-2021. Of note, all patients received enoxaparin for DVT prophylaxis during the
intensification phase.
Results: During the induction phase, 19 out of 178 (10.6%) patients who received L-ASNase
during induction developed VTE, whereas 21 out of 71 (29.6%) patients with PEG-ASNase
developed VTE (p-value = 0.0002, OR = 3.079, 95 CI 1.43-6.64), with multivariable
analysis after adjusting for age, sex, weight, and central line types. Similarly,
during the intensification phase, 19 out of 135 (14.1%) patients had VTE on L-ASNase
while 10 out of 34 (29.4%) patients on PEG-ASNase developed VTE (p-value =0.014, OR=
2.94, 95% CI 1.21-7.13) after adjusting for age, sex, weight, and central line types.
Of note, PICC line was noted to be associated with higher incidence of CVC lines with
during induction (p-value= 0.021, OR= 2.52, 95% CI 1.15-5.51).
Image:
Summary/Conclusion: PEG-ASNase is associated with higher incidence of VTE compared
to L-ASNase, both during induction and intensification, despite the administration
of prophylactic anticoagulation. Further investigations and strategies on reducing
VTE risk associated with PEG-ASNase is needed in adult ALL patients who receive an
asparaginase-based protocol.
P353: FORTY MONTHS UPDATE OF THE GIMEMA LAL2116 (D-ALBA) PROTOCOL AND ANCILLARY LAL2217
STUDY FOR NEWLY DIAGNOSED ADULT PH+ ALL
S. Chiaretti1,*, R. Bassan2, A. Vitale1, L. Elia1, A. Piciocchi3, P. Viero2, F. Ferrara4,
M. Lunghi5, F. Fabbiano6, M. Bonifacio7, N. Fracchiolla8, P. Di Bartolomeo9, P. Fazi3,
M. S. De Propris1, M. Vignetti3, A. Guarini10, A. Rambaldi11, R. Foà1
1Translational and Precision Medicine, Sapienza University of Rome, Rome; 2Hematology,
Ospedale dell’Angelo, Venice; 3GIMEMA Data Center, Fondazione GIMEMA – Franco Mandelli
Onlus, Rome; 4Hematology, Ospedale Antonio Cardarelli, Naples; 5Translational Medicine,
University of Eastern Piedmont, Novara; 6Hematology, Hospital Villa Sofia-cervello,
Palermo; 7Medicine, Verona University, Verona; 8Fondazione IRCCS, Ca’ Granda-Ospedale
Maggiore Policlinico, Milan; 9Hematology, Transfusion Medicine and Biotechnologies,
Ospedale Civile, Pescara; 10Molecular Medicine, Sapienza University of Rome, Rome;
11Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
Background: The outcome of adult Philadelphia-positive acute lymphoblastic leukemia
(Ph+ ALL) patients greatly improved since the introduction of tyrosine kinase inhibitors
(TKIs). A further improvement was achieved with a chemo-free induction/consolidation
strategy based on the sequential administration of dasatinib followed by blinatumomab
- the D-ALBA GIMEMA LAL2116 trial - with overall survival (OS) and disease-free survival
(DFS) rates of 95% and 88% at 18 months (Foa et al, NEJM 2020).
Aims: To provide an updated follow-up of the D-ALBA trial and to document the long-term
management of previously enrolled patients.
Methods: In the D-ALBA trial, after the primary endpoint, patients were followed for
12 months. Data on subsequent treatment and survival are being collected in the ancillary
study GIMEMA LAL2217.
Results: As reported, 63 patients were enrolled (median age 54 years, 24-82; no upper
age limit); the median follow-up is 40 months (0.9-62.5). Since enrollment in the
D-ALBA trial, 9 relapses have been documented: 4 hematologic (1 major protocol deviation
and 1 in an 82-year old woman who rapidly went off study), 4 at CNS and 1 nodal; median
time to relapse was 4.4 months (1.9-25.8). Six deaths were recorded (3 post-allogeneic
transplant) in 1st complete hematologic remission (CHR).
Of the 58 patients who started blinatumomab, 29 continued treatment with TKI: 21 with
dasatinib (85% after receiving all 5 cycles of blinatumomab), 2 switched to imatinib
due to intolerance (all after the 5 cycles of blinatumomab) and 5 switched to ponatinib
for a molecular minimal residual disease increase or medical decision (66% had received
all 5 cycles of blinatumomab). Twenty-nine patients were allografted, including 6
patients in 2nd CHR: 9 from a sibling, 13 from an unrelated donor, 6 from a haploidentical
donor and 1 from a cord blood. Six transplants were performed in 2nd CHR. Before transplant,
8 patients received 2 and 3 cycles of blinatumomab, respectively, 5 4 cycles and 6
5 cycles (2 patients were allografted after 1 cycle). Among grafted patients, 6 deaths
occurred, 3 of which in cases transplanted in 2nd CHR; thus, the transplant-related
mortality in 1st CHR is 10%. When considered in a covariate model, transplant did
not impact on both OS and DFS; similar results were obtained also considering allograft
only in 1st CHR. It must, however, be underlined that the allografted population was
enriched in cases who did not achieve a molecular response (23 pts). Further investigation
is ongoing on this aspect. In the updated follow-up, the estimated 48 months OS is
78% (95% CI: 66-92%) and the DFS is 75% (95% CI: 64-87%). DFS was significantly better
in patients achieving a molecular response upon induction than in those who did not
(100% vs 67%, p=0.016, Fig. 1A), with no events recorded in the group of complete
molecular responders and positive-not-quantifiable cases. Furthermore, we confirmed
the inferior DFS for patients carrying an IKZF1
plus genotype compared to that of cases with no IKZF1 deletions/IKZF1 deletions alone
(85% vs 47%, p=0.012, Fig. 1B).
Image:
Summary/Conclusion: In the updated analysis of the D-ALBA trial and the ancillary
GIMEMA LAL2217 study, with a median OS of 40 months, we confirm the reported very
favorable outcome. Among the few relapsed cases, we observed a rather high incidence
of CNS involvements. These results represent the scientific rationale for the ongoing
phase 3 GIMEMA LAL2820 trial, where dasatinib has been substituted by ponatinib, CNS
prophylaxis has been increased, and transplant allocation is based on genomic features
and minimal residual disease monitoring.
P354: REAL-WORLD ANALYSIS OF THE RISKS OF THROMBOTIC AND BLEEDING EVENTS IN PATIENTS
RECEIVING PEGYLATED ASPARAGINASE FOR TREATMENT OF ACUTE LYMPHOBLASTIC LEUKEMIA
B. Mautner1, B. Brewer2, J. Mort1, E. Pierce1, F. Ghamsari1, S. Dibenedetto1, E. Morse1,
L. Wells1, A. Morris1, C. Allen3, I. Patel4, D. D’Souza4, C. Jackson5, M. Perciavalle6,
B. Yelvington6, R. Miller6, K. Abernathy6, K. Walsh6, H. Wolfe7, S. Locke7, D. Reed5,
V. Duong4, B. Dholaria6, T. LeBlanc7, R. Shallis3, M. Keng1, B. Horton2, F. El Chaer1,1,*
1Hematology and Oncology; 2Public Health Sciences, Division of Translational Research
and Applied Statistics, University of Virginia, Charlottesville; 3Hematology and Oncology,
Yale University, New Haven; 4Hematology and Oncology, University of Maryland Medical
Center, Baltimore; 5Hematology and Oncology, Wake Forest Baptist Medical Comprehensive
Cancer Center, Winston-Salem; 6Hematology and Oncology, Vanderbilt University Medical
Center, Nashville; 7Hematology and Oncology, Duke Cancer Institute, Durham, United
States of America
Background: The prognosis of younger adult patients with acute lymphoblastic leukemia
(ALL) has improved with the adoption of pediatric-inspired chemotherapy regimens.
PEGylated asparaginase (PEG-Asp) is essential to these regimens but is associated
with an increased risk of coagulopathy, venous thromboembolism, and depletion of antithrombin
III (AT3). There are limited data evaluating the risk of clinical coagulopathy (thrombosis
and bleeding) and the optimal antithrombotic interventions in these patients are unknown.
Aims: We aimed to assess the risk of thrombotic and bleeding events in patients with
ALL treated with PEG-Asp and compare the treatment modalities with either AT3 + fibrinogen
repletion vs the addition of prophylactic anticoagulation (AC) with either subcutaneous
heparin 5000 TID or subcutaneous lovenox 30-40mg/day.
Methods: We performed a multicenter retrospective analysis of patients ≥ 18 yo who
received PEG-Asp for B- or T-ALL as part of multiagent chemotherapy from 2015-2020
treated across 6 US academic institutions. Categorical variables were summarized using
means and standard deviations. The former was compared using Fisher’s exact test;
the latter was compared using either the Wilcoxon text (for two groups) or the Kruskal-Wallis
(for more than two groups) as appropriate. One way ANOVA test was used to compare
continuous variables.
Results: Our study included 129 treated patients who met eligibility criteria for
induction and 73 patients who underwent post-induction therapy. During induction,
the majority of the patients (90%) had AT3 and fibrinogen repletion, while only 10%
received additional prophylactic anticoagulation. The rates of any thrombotic events
were lower in the AT3/cryoprecipitate-only treatment group (18% vs 50%, p=0.015).
Venous thromboembolism accounted for the majority of the thrombotic events (91% and
100% in the AT3/cryoprecipitate vs AT3/cryoprecipitate + prophylactic anticoagulation,
respectively). Bleeding rates were numerically higher in patients who received AT3/cryoprecipitate
+ prophylactic anticoagulation (33% vs 14%, p=0.18). The rates of thromboses or bleeding
events were not different when either prednisone or dexamethasone were included in
the induction treatment regimen. During the post-induction phase, the rates of thrombotic
and bleeding events were similar across all groups. The median time to thrombotic
event was 11 days after the last PEG-Asp administration during induction, and 123
days during post-induction.
Image:
Summary/Conclusion: Although the increased thrombotic risk in adult patients receiving
PEG-Asp is well-characterized, rates of reported thromboses vary widely. Studies examining
bleeding risk associated with PEG-Asp regimens are even scarcer and with more variable
outcomes. In our study, there was both a statistically significant increase in the
rate of any thrombotic event as well as a trend towards increased bleeding in patients
receiving AT3/cryoprecipitate + prophylactic anticoagulation versus those receiving
AT3/cryoprecipitate alone. More investigation into the relative thrombotic and bleeding
risks associated with PEG-Asp is needed to determine optimal antithrombotic interventions
in adult patients with ALL.
P355: MINI-HYPER-CVD PLUS INOTUZUMAB OZOGAMICIN, WITH OR WITHOUT BLINATUMOMAB, IN
OLDER ADULTS WITH NEWLY DIAGNOSED B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA: UPDATES FROM
A PHASE II TRIAL
F. Haddad1,*, H. Kantarjian1, N. Short1, F. Ravandi1, N. Jain1, W. Macaron1, T. Kadia1,
Y. Alvarado1, N. Daver1, G. Borthakur1, C. DiNardo1, M. Konopleva1, W. Wierda1, J.
Jacob1, E. Roy1, C. Loiselle1, A. Milton1, J. Rivera1, R. Garris1, E. Jabbour1
1Leukemia, The University of Texas MD Anderson Cancer Center, Houston, United States
of America
Background: Blinatumomab (Blina) and inotuzumab ozogamicin (INO) improve the outcomes
of patients (pts) with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL).
These drugs may allow for use of less chemotherapy and improve remission durations
and overall survival (OS) in older pts with newly diagnosed B-ALL.
Aims: To evaluate the combination of low-intensity chemotherapy and INO with or without
Blina, in older pts with newly diagnosed B-ALL.
Methods: Pts ≥60 years with newly diagnosed Philadelphia chromosome (Ph)-negative
B-ALL were eligible to receive mini-hyper-CVD for up to 8 cycles. Initially, INO was
given at 1.3-1.8mg/m2 on day (D) 3 of cycle 1 and 0.8-1.3mg/m2 on D3 of cycles 2-4.
Rituximab (if CD20+) and prophylactic IT chemotherapy were given for the first 4 cycles.
POMP maintenance was given for up to 3 years. Starting with pt #50, the protocol was
amended and INO was administered in fractionated doses with each of the 4 cycles of
hyper-CVD (0.6 mg/m2 on D2 and 0.3 mg/m2 on D8 of cycle 1; 0.3 mg/m2 on D2 and 8 of
cycles 2-4) followed by 4 cycles of blina. The cumulative doses of INO given before
and after this amendment were 4.3 mg/m2 and 2.7 mg/m2, respectively. Maintenance was
also amended to include 12 cycles of POMP and 4 cycles of blina (1 cycle of blina
after every 3 cycles of POMP).
Results: 80 pts have been treated with a median age of 68 years (range, 60-87); 30
pts (38%) were ≥70 years. 6 pts were in complete remission (CR) at enrollment and
unevaluable for morphological response. Pt characteristics are summarized below (Figure
1A).
The overall response rate (ORR) among 74 evaluable pts was 99% (CR, n=66; CRi, n=7).
MRD negativity by flow cytometry was achieved in 61/76 pts (80%) after 1 cycle and
74/79 pts (94%) overall. The 30-day and 60-day mortality rates were 0% and 3%, respectively.
Among 79 pts who achieved remission, 33 (42%) are still in ongoing remission without
stem cell transplant (SCT), 31 (39%) died in remission, 11 (14%) relapsed without
SCT, and 4 (5%) proceeded to SCT in first remission (1 of whom subsequently relapsed).
Notably, 9 pts (11%) later developed MDS/AML, 7 of whom had a TP53 mutation. Overall,
6 pts (8%) developed VOD, 1 after allogeneic SCT.
With a median follow-up of 55 months (range, 3-119), the 5-year continuous remission
and OS rates were 76% and 47%, respectively (Figure 1B). Pts aged 60-69 years had
better outcomes compared with pts ≥70 years (5-year OS rates: 59% vs 27%, respectively;
P=0.04) and as did those without poor cytogenetics (i.e. complex, KMT2A-rearranged,
low-hypodiploidy/near-triploidy) compared with poor cytogenetics (5-year OS rates:
54% and 25%, respectively; P=0.02). Deaths in remission were more frequent in pts
≥70 years compared with those 60-69 years (70% vs 35%; P=0.005). The 5-year OS rates
for pts age 60-69 without poor-risk cytogenetics (n=37), age 60-69 with poor-risk
cytogenetics (n=13), age ≥70 without poor-risk cytogenetics (n=24), and age ≥70 with
poor-risk cytogenetics (n=6) were 69%, 39%, 36% and 0% respectively (Figure 1C). Presence
of a TP53 mutation did not significantly impact OS (P=0.22).
Image:
Summary/Conclusion: Reduced-intensity chemotherapy plus INO, with or without blina,
resulted in an ORR of 99% and a 5-year OS rate of 47% in older pts with newly diagnosed
Ph-negative B-ALL. Outcomes were particularly favorable in those 60-69 years of age
without poor-risk cytogenetics. Pts ≥70 years were at a higher risk of death in remission.
A chemotherapy-free approach, with INO plus blina only, is therefore now being explored
in this population.
P356: SUBGROUP ANALYSES OF KTE-X19, AN ANTI-CD19 CHIMERIC ANTIGEN RECEPTOR (CAR) T-CELL
THERAPY, IN ADULT PATIENTS WITH RELAPSED OR REFRACTORY B-CELL ACUTE LYMPHOBLASTIC
LEUKEMIA (R/R B-ALL) IN ZUMA-3
B. D. Shah1,*, R. D. Cassaday2, J. H. Park3, R. Houot4, O. O. Oluwole5, A. C. Logan6,
N. Boissel7, T. Leguay8, M. R. Bishop9, M. S. Topp10, D. Tzachanis11, K. M. O’Dwyer12,
M. L. Arellano13, Y. Lin14, M. R. Baer15, G. J. Schiller16, M. Subklewe17, M. Abedi18,
M. C. Minnema19, W. G. Wierda20, D. J. DeAngelo21, P. Stiff22, D. Jeyakumar23, J.
Dong24, S. Adhikary24, L. Zhou24, P. C. Schuberth24, B. Kharabi Masouleh24, A. Ghobadi25
1Moffitt Cancer Center, Tampa, FL; 2University of Washington, Fred Hutchinson, Seattle
Cancer Care Alliance, Seattle WA; 3Memorial Sloan Kettering Cancer Center, New York,
NY, United States of America; 4CHU Rennes, University Hospital Rennes, Inserm & EFS,
Rennes, France; 5Vanderbilt University Cancer Center, Nashville, TN; 6UCSF Medical
Center, San Francisco, CA, United States of America; 7Hôpital Saint-Louis, Paris;
8Service d’hématologie clinique et thérapie cellulaire Hôpital du Haut-Leveque CHU
de Bordeaux, Bordeaux, France; 9The University of Chicago Medicine, Chicago, IL, United
States of America; 10Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg,
Würzburg, Germany; 11University of California San Diego, San Deigo, CA; 12Wilmot Cancer
Institute of University of Rochester, Rochester, NY; 13Winship Cancer Institute of
Emory University, Atlanta, GA; 14Mayo Clinic, Rochester, MN; 15University of Maryland
Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD; 16David
Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America; 17Ludwig-Maximilians-Universität
München, Munich, Germany; 18University of California Davis Comprehensive Cancer Center,
Sacramento, CA, United States of America; 19University Medical Center Utrecht, on
behalf of HOVON/LLPC, Utrecht, Netherlands; 20The University of Texas MD Anderson
Cancer Center, Houston, TX; 21Dana-Farber Cancer Institute, Boston, MA; 22Loyola University
Chicago Stritch School of Medicine, Maywood, IL; 23University of California Irvine
Medical Center, Irvine, CA; 24Kite, a Gilead Company, Santa Monica, CA; 25Washington
University School of Medicine, St Louis, MO, United States of America
Background: Brexucabtagene autoleucel (KTE-X19) is approved in the US for the treatment
of adult patients with R/R B-ALL based on the positive results of the ZUMA-3 study.
In the pivotal Phase 2 portion of ZUMA-3, the overall complete remission (CR) rate
(CR + CR with incomplete hematologic recovery [CRi]) was 71% (N=55; median follow-up
16.4 months; Shah et al. Lancet 2021).
Aims: To assess outcomes in ZUMA-3 by prior number of therapy lines, prior blinatumomab,
prior allogeneic stem cell transplant (alloSCT), and subsequent alloSCT.
Methods: Eligible adults had R/R B-ALL and received a single infusion of KTE-X19 following
leukapheresis and conditioning chemotherapy. Outcomes assessed by independent review
are reported in all patients treated at the pivotal dose (1×106 CAR T cells/kg).
Results: As of 23 July 2021, the median follow-up among all treated patients in Phase
2 (N=55) was 26.8 months (range, 20.7-32.6). At baseline, 10 patients (18%) had 1
prior line of therapy, and 45 patients (82%) had ≥2 prior lines. The overall CR/CRi
rates (95% CI) for patients with 1 or ≥2 prior lines of therapy were 90% (55-100)
and 67% (51-80), respectively. In patients with 1 or ≥2 prior lines of therapy, the
median (95% CI) duration of remission (DOR) was 4.7 months (1.8-not estimable [NE])
and 14.6 months (9.4-NE), and the median (95% CI) overall survival (OS) was not reached
(NR) (2.1-NE) and 25.4 months (14.2-NE), respectively. Grade ≥3 cytokine release syndrome
(CRS) occurred in 10% and 27% of patients with 1 or ≥2 prior lines of therapy, and
Grade ≥3 neurologic events occurred in 30% and 24%, respectively.
Among the 25 patients (45%) who had prior blinatumomab at baseline, the overall CR/CRi
rate (95% CI) was 60% (39-79), and 80% (61-92) in the 30 patients (55%) without prior
blinatumomab. The median (95% CI) DOR was 19.1 months (1.3-NE) and 10.3 months (5.2-NE)
in patients with or without prior blinatumomab, and the median (95% CI) OS was 14.2
months (3.2-26.0) and NR (18.6-NE), respectively. In patients with or without prior
blinatumomab, rates of Grade ≥3 CRS were 24% and 23%, and rates of Grade ≥3 neurologic
events were 20% and 30%, respectively.
The overall CR/CRi rate in the 23 patients (42%) who had prior alloSCT vs the 32 patients
(58%) who did not was 70% (47-87) vs 72% (53-86), respectively. In patients with vs
without prior alloSCT, the medians (95% CI) for DOR were 14.6 months (8.7-23.6) vs
NR (4.7-NE), and the medians (95% CI) for OS were 25.4 months (14.2-NE) vs NR (9.0-NE),
respectively. Grade ≥3 CRS occurred in 17% and 28% of patients with or without prior
alloSCT, and Grade ≥3 neurologic events occurred in 26% and 25%, respectively.
For patients who achieved CR/CRi and did (n=10) or did not (n=29) proceed to subsequent
alloSCT post–KTE-X19 infusion, the median (95%CI) DOR was NR (NE-NE) and 14.6 months
(8.7-23.6), and the median (95% CI) OS was NR (7.6-NE) and 26.0 months (18.6-NE),
respectively.
A pooled analysis of all Phase 1 and 2 patients treated at the pivotal dose (N=78)
was newly conducted, with a median follow-up of 29.7 months (range, 20.7-58.3). The
similar results observed in this expanded data set as shown in the Table further support
the subgroup outcomes described above in Phase 2 patients.
Image:
Summary/Conclusion: Adults with R/R B-ALL benefitted from KTE-X19, with manageable
safety, regardless of prior exposure to blinatumomab or prior alloSCT, though survival
appeared better in patients without these prior therapies and in earlier lines of
therapy, with limited patient numbers in some subgroups.
roups.
P357: MULTICENTER RETROSPECTIVE ANALYSIS OF CLINICAL OUTCOME OF ADULT PATIENS WITH
MIXED-PHENOTYPE ACUTE LEUKEMIA (MPAL) DIAGNOSED AND TREATED IN THE LAST TEN YEARS.
A CAMPUS-ALL STUDY.
D. Lazzarotto1,*, I. Tanasi2, A. Vitale3, M. Piccini4, M. Dargenio5, F. Giglio6, F.
Forghieri7, N. Fracchiolla8, M. Cerrano9, E. Todisco10, C. Papayannidis11, M. Leoncin12,
M. Defina13, F. Guolo14, C. Pasciolla15, M. Delia16, P. Chiusolo17, A. Mulè18, A.
Candoni1, M. Bonifacio2, G. Pizzolo2, R. Foà3
1Clinica Ematologica, Azienda Sanitaria Universitaria Friuli Centrale, Udine; 2Dipartimento
di Medicina, Sezione di Ematologia, Università di Verona, Verona; 3Ematologia, Dipartimento
di Medicina Traslazionale e di Precisione, “Sapienza” Università di Roma, Roma; 4SOD
Ematologica, AOU Careggi, Firenze; 5S.C. Ematologia, Ospedale Vito Fazzi, Lecce; 6Unità
di Ematologia e Trapianto di Midollo Osseo, IRCCS Ospedale San Raffaele, Milano; 7S.C.
Ematologia, Azienda Ospedaliero Universitaria di Modena, Modena; 8U.O. Ematologia,
IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milano; 9S.C. Ematologia
2, AO Città della Salute e della Scienza, Torino; 10Divisione di Ematoncologia, Istituto
Europeo di Oncologia, Milano; 11Dipartimento di Oncologia e Ematologia, Policlinico
S. Orsola-Malpighi, Bologna; 12U.O. di Ematologia, Ospedale dell’Angelo, Mestre; 13UOC
Ematologia, Azienda Ospedaliero Universitaria Senese, Siena; 14Ematologia e Terapie
Cellulari, IRCCS Policlinico San Martino, Genova; 15U.O. di Ematologia, IRCCS Istituto
Tumori Giovanni Paolo II; 16U.O. Ematologia con Trapianto, Azienda Ospedaliero-Universitaria
Consorziale, Policlinico di Bari, Bari; 17Servizio di Ematologia, Policlinico Gemelli,
Roma; 18Divisione di Ematologia ad indirizzo oncologico, A.O. Ospedali Riuniti Villa
Sofia-Cervello, Palermo, Italy
Background: Mixed phenotype acute leukemia (MPAL) is a very rare disease in adults.
Most data derive from small retrospective series, some including also pediatric patients.
Generally, treatment is similar to that of acute lymphoblastic leukemia (ALL), but
the outcomes in adults and the role of allogeneic stem cell transplantation (AlloSCT)
are not well defined.
Aims: Purpose of this retrospective study was to provide data on a large cohort of
adult patients diagnosed and treated in 18 Italian hematology centers in the last
10 years.
Methods: Seventy-seven adult patients diagnosed with MPAL according to the EGIL and
WHO criteria in the last 10 years (2011-2021) and treated with curative intent were
included in the study. Endpoints were the CR rate after induction, overall survival
(OS), disease-free survival (DFS) and rate of AlloSCT.
Results: Forty-eight of the 77 (62%) patients had B/myeloid MPAL, 27 (35%) had T/myeloid
MPAL and 2 (3%) had B/T/myeloid MPAL. Median age at diagnosis was 49 years (range
17 - 77); it was lower in patients with T/myeloid MPAL than in B/myeloid MPAL cases
(39 vs 54 years, P=0.04). Extramedullary involvement was detected in 26/77 (34%) of
patients (mainly lymph nodes and spleen), but none had a central nervous system involvement.
Cytogenetic analysis was normal in 40% of patients; 36% of cases had a complex karyotype,
24% had other cytogenetic abnormalities (including 8% with a BCR-ABL1 rearrangement).
FLT-ITD mutation was detected in 14% of patients. No differences were found between
B/myeloid and T/myeloid MPAL cases.
Thirty/77 (39%) of patients were treated with an acute myeloid leukemia (AML)-like
induction regimen, while 47 (61%) were treated with an ALL-like induction. Globally,
CR rate after induction was 62% and 42% of these cases were also MRD negative. Patients
treated with an AML-like induction had a higher rate of refractory disease after induction
(33% vs 10%, P=0.02). The death during induction (DDI) rate was similar in the 2 groups
(7% vs 8%). In univariate analysis an ALL-like therapy was the only variable associated
with a better CR rate (P=0.0447). Patients refractory to AML-like induction were mainly
salvaged with a subsequent AlloSCT or ALL-like therapy followed by an AlloSCT, and
60% of them achieved a CR.
The median OS and DFS of the entire cohort were respectively 41.9 and 37.6 months,
with an OS and DFS at 5 years of 43% and 39%, respectively. Age, type of induction
therapy, karyotype, presence of FLT3-ITD mutation, extramedullary involvement and
type of MPAL (B/myeloid vs T/myeloid) were tested in univariate analysis for OS and
DFS. Age below 60 years was the only variable associated with a better OS (median
OS of 67 months vs 26 months, P=0.0138), while no variable impacted on DFS.
AlloSCT was performed in 50 patients (65%), 80% of them (40 patients) as part of the
frontline treatment (36/40 patients transplanted in CR1). Among transplanted patients,
we observed a 5-year OS of 54% (median not reached), that improved to 69% in patients
transplanted as part of the frontline treatment.
Summary/Conclusion: This study describes one of the largest cohorts of adult patients
with MPAL. These data outline that this disease responds better to ALL-like induction
therapy than to AML-like therapy and that consolidation therapy should include, whenever
possible, an AlloSCT. Prospective studies are needed to uniform the therapeutic approach
and to identify variables capable of predicting outcome and to guide therapy.
P358: COMPREHENSIVE DIAGNOSTICS OF ACUTE LYMPHOBLASTIC LEUKEMIA BY WHOLE TRANSCRIPTOME
RNA SEQUENCING
J. Li1,*
1SINO-US Diagnostics, Tianjin, China
Background: Acute lymphoblastic leukemia (ALL) has genetic heterogeneity, which is
helpful to clinical diagnosis, prognosis stratification and treatment guidance. Gene
aberrations are structurally diverse and are currently analyzed by different assays.
Aims: This study aims to establish a single and comprehensive platform for ALL diagnosis
by whole transcriptome RNA sequencing (WTRS), which can detect fusion, mutations and
expression.
Methods: We analyzed the data of 70 ALL patients by WTRS.
Results: We found that the fusion coincidence rate of WTRS was 100% (19 / 19) comparing
with quantitative real-time polymerase chain reaction (qPCR). At the same time, six
rare fusion forms were detected, which increased the diagnosis rate of fusion gene
from 27% (19 / 70) to 36% (25 / 70). The expression of CRLF2 gene in 5 cases was consistent
with that of qPCR. Seven cases with IKZF1 gene skipping identified by qPCR were consistent
with the results of WTRS, and a rare IKZF1 deletion subtype was detected. Of the 51
genes associated with ALL, 100% (103/103) gene mutation at the RNA level by WRTS is
consistent with the results at the DNA level by targeted DNA sequencing (except 3
splice site mutations and 4 mutations with poor coverage).
Summary/Conclusion: In conclusion, the standardization of experimental operation and
improvement of biological information pipline of WRTS are very important factors in
its use in standard diagnostic procedures. WRTS has the potential to provide accurate
comprehensive diagnostic information for clinic and further improve the ALL diagnostic
rate.
P359: IMPROVED OVERALL SURVIVAL AND MRD CLEARANCE WITH BLINATUMOMAB VS CHEMOTHERAPY
AS PRE-TRANSPLANT CONSOLIDATION IN PEDIATRIC HIGH-RISK FIRST-RELAPSE B-CELL PRECURSOR
ACUTE LYMPHOBLASTIC LEUKEMIA (B-ALL)
F. Locatelli1,*, G. Zugmaier2, C. Rizzari3, J. Morris4, B. Gruhn5, T. Klingebiel6,
R. Parasole7, C. Linderkamp8, C. Flotho9, A. Petit10, C. Micalizzi11, Y. Zeng4, R.
Desai12, W. Kormany4, C. Eckert13, A. Möricke14, M. Sartor15, O. Hrusak16, C. Peters17,
V. Saha18,19, L. Vinti1, A. von Stackelberg20
1IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome, Italy;
2Amgen Research (Munich), Munich, Germany; 3MBBM Foundation, ASST Monza, University
of Milano-Bicocca, Monza, Italy; 4Amgen Inc., Thousand Oaks, United States of America;
5Universitaetsklinikum Jena, Jena; 6Universitätsklinikum Frankfurt am Main, Frankfurt,
Germany; 7Azienda Ospedaliera di Rilievo Nazionale Santobono Pausilipon, Napoli, Italy;
8Medizinische Hochschule Hannover, Hanover; 9Universitätsklinikum Freiburg, Freiburg,
Germany; 10Hopital Armand Trousseau, APHP.Sorbonne Université, Paris, France; 11Istituto
Pediatrico di Ricerca e Cura a Carattere Scientifico G Gaslini, Genova, Italy; 12IQVIA
Inc., Durham, United States of America; 13Charité Campus Virchow-Klinikum Pädiatrie
m.S. Onkologie/Hämatologie, Berlin; 14Univ.-Klinikum Schleswig-Holstein, Kiel, Germany;
15Westmead Hospital, Sydney, Australia; 16Charles University, Motol University Hospital,
Prague, Czechia; 17St Anna Children’s Hospital, Vienna, Austria; 18Division of Cancer
Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University
of Manchester, Manchester, United Kingdom; 19Tata Translational Cancer Research Centre,
Tata Medical Center, Kolkata, India; 20Charite Universitaetsmedizin CVK Berlin, Berlin,
Germany
Background: Children with high-risk first-relapse B-ALL have a poor prognosis. We
previously reported that, as compared with chemotherapy, treatment with blinatumomab
resulted in prolonged event-free survival (EFS) and overall survival (OS) and higher
rates of minimal residual disease (MRD) remission in a phase 3 trial (JAMA 2021;325:843-54;
ASH 2021:Abs 1231).
Aims: Here we provide follow-up (FU) data as of Sep 2021, with a focus on survival
and MRD response.
Methods: In this phase 3 trial (Amgen NCT02393859), children >28 days and <18 years
old with high-risk first-relapse B-ALL were randomized 1:1 after induction and 2 cycles
of consolidation to receive either a third intensive multidrug chemotherapy consolidation
or blinatumomab (15 μg/m2/day, 4 weeks, continuous IV infusion). Study enrollment
required M1 (<5% blasts) or M2 (≥5% and <25% blasts) bone marrow (BM) at randomization.
Patients with complete remission (CR, ie, M1 BM) after treatment could undergo allogeneic
hematopoietic stem cell transplant (alloHSCT). EFS, the primary endpoint, was calculated
from randomization to whichever occurred first of relapse or M2 BM after CR, failure
to achieve a CR at the end of treatment, second malignancy, or death (any cause).
Survival outcomes and response rates were analyzed by baseline MRD, ie, MRD at enrollment
prior to chemotherapy or blinatumomab. Parents or a legally acceptable representative
provided written informed consent.
Results: Enrollment was stopped based on the interim analysis for benefit of blinatumomab
(7/17/19 primary analysis). The following results reflect an updated analysis as of
Sep 2021. Between Nov 2015 and Aug 2019, 111 patients were randomized: 54 (49%) to
blinatumomab and 57 (51%) to chemotherapy at 47 centers in 13 countries. Baseline
characteristics were comparable in both arms as previously reported. After a median
FU of 44 months, EFS was significantly higher with blinatumomab vs chemotherapy (at
4 years: 59% vs 27%, stratified log-rank p<0.001, hazard ratio [HR]: 0.35, 95% CI:
0.20-0.61). Blinatumomab also showed a strong benefit for the secondary endpoint of
OS (at 4 years: 77% vs 49%, stratified log-rank p=0.002, HR: 0.34, 95% CI: 0.17-0.69).
EFS, OS, and MRD remission (<10-4 blasts) were all improved with blinatumomab, both
overall and by baseline MRD subgroup (< or ≥10-3) (Table 1a-b, Figure). Further, patients
with and without extramedullary disease both had a benefit with blinatumomab. AlloHSCT
in second CR was performed in 51/54 patients in the blinatumomab arm and in 39/57
patients in the chemotherapy arm. No new safety signals were identified. Second relapses
occurred in 16/54 (30%) patients in the blinatumomab arm and 34/57 (60%) patients
in the chemotherapy arm. In the blinatumomab arm, these second relapses were extramedullary
in 5/54 patients, CNS in 5 (9.3%), 2 of which were solely CNS. In the chemotherapy
arm, these second relapses were extramedullary in 8/57 patients, CNS in 3 (5.3%),
2 of which were solely CNS. CD19-negative relapse was seen in 3 patients in the blinatumomab
arm (3/54) and 1 patient in the chemotherapy arm (1/57) prior to any subsequent CD19-directed
therapy (ie, after study therapy).
Image:
Summary/Conclusion: In children with high-risk first-relapse B-ALL, treatment with
1 cycle of blinatumomab vs chemotherapy before alloHSCT resulted in superior EFS and
improved OS and MRD response rates, all independent of baseline MRD. The incidence
of CD19-negative relapse after blinatumomab was low.
P360: EFFICACY AND SAFETY OF DARATUMUMAB IN PEDIATRIC AND YOUNG ADULT PATIENTS WITH
RELAPSED/REFRACTORY T-CELL ACUTE LYMPHOBLASTIC LEUKEMIA OR LYMPHOBLASTIC LYMPHOMA:
RESULTS FROM PHASE 2 DELPHINUS STUDY
A. Vora1,*, T. Bhatla2, D. Teachey3, F. Bautista4,5, J. Moppett6, P. Velasco Puyó7,
C. Micalizzi8, C. Rossig9, N. Shukla10, G. Gilad11,12, F. Locatelli13, A. Baruchel14,
C. M. Zwaan4,15, E. A. Raetz16, N. Bandyopadhyay17, L. Lopez Solano18, R. M. Dennis18,
R. Carson18, L. E. Hogan19
1Department of Haematology, Great Ormond Street Hospital for Children, London, United
Kingdom; 2Department of Pediatrics, Children’s Hospital of New Jersey at Newark Beth
Israel Medical Center, Newark, NJ; 3Division of Oncology, Center for Childhood Cancer
Research, Children’s Hospital of Philadelphia, Perelman School of Medicine, University
of Pennsylvania, Philadelphia, PA, United States of America; 4Princess Máxima Center
for Pediatric Oncology, Utrecht, Netherlands; 5Department of Pediatric Oncology and
Hematology, Hospital Niño Jesús, Madrid, Spain; 6Paediatric Haematology, Bristol Royal
Hospital for Children, Bristol, United Kingdom; 7Servicio de Oncología y Hematología
Pediátricas, Vall d’Hebron Hospital, Barcelona, Spain; 8Clinical Experimental Haematology
Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy; 9University Children’s Hospital
Münster, Münster, Germany; 10Department of Pediatrics, Memorial Sloan Kettering Cancer
Center, New York, NY, United States of America; 11Department of Pediatric Hematology-Oncology,
Schneider Children’s Medical Center of Israel, Petach Tikva; 12Sackler Faculty of
Medicine, Tel Aviv University, Tel Aviv, Israel; 13Department of Pediatric Hematology
and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza University of Rome,
Rome, Italy; 14Pediatric Hematology and Immunology Department, Robert Debré University
Hospital (APHP and Université de Paris), Paris, France; 15Department of Pediatric
Oncology, Erasmus MC-Sophia Children’s Hospital, Rotterdam, Netherlands; 16New York
University Langone Medical Center, New York, NY; 17Janssen Research & Development,
LLC, Raritan, NJ; 18Janssen Research & Development, LLC, Spring House, PA; 19Department
of Pediatrics, Stony Brook Children’s, Stony Brook, NY, United States of America
Background: Approximately 15-20% of pediatric patients with T-cell acute lymphoblastic
leukemia (ALL) or lymphoblastic lymphoma (LL) will be refractory to/relapse after
frontline treatment; relapsed disease is associated with poor outcomes. In a phase
2 study, 2 of 7 (28.6%) patients with T-cell ALL in first relapse achieved a complete
response (CR) using the vincristine, prednisone, PEG-asparaginase, and doxorubicin
(VPLD) reinduction backbone. Daratumumab (DARA), a human IgGκ monoclonal antibody
targeting CD38 approved for treating multiple myeloma, has shown preclinical efficacy
in ALL models.
Aims: Here we report the initial results of DARA plus VPLD in pediatric and young
adult patients with relapsed/refractory T-cell ALL or LL enrolled in the phase 2,
open-label DELPHINUS study.
Methods: Eligible patients were aged 1-30 y, had T-cell ALL or LL in first relapse
or refractory to 1 prior induction/consolidation regimen, and had a performance status
≥70. DARA (16 mg/kg IV QW) was given with VPLD in Cycle 1 and with methotrexate, cyclophosphamide,
cytarabine, and 6-mercaptopurine in Cycle 2. Patients received age/risk-adjusted intrathecal
therapy. Response was measured at the end of each cycle by local bone marrow morphology.
The primary endpoint for ALL patients was CR rate in pediatric patients at the end
of Cycle 1. Patients achieving CR after Cycles 1 or 2 could proceed to allogeneic
HSCT off study. Overall response rate (ORR) was defined as CR or CR with incomplete
hematological recovery (CRi) at any time before start of subsequent therapy or HSCT.
Minimal residual disease (MRD) negativity (<0.01%) at any time before disease progression,
start of subsequent therapy, or HSCT was centrally reviewed by flow cytometry.
Results: Twenty-four pediatric (age 1-17 y) and 5 young adult (age 18-30 y) ALL patients
and 10 LL patients (age 1-30 y) received ≥1 DARA dose. Median (range) age was 10.0
(2-25) y (ALL) and 14.5 (5-22) y (LL); median (range) time from initial diagnosis
to first study treatment was 2.0 (0.1-6.1) y (ALL) and 0.8 (0.5-6.0) y (LL). Pediatric
ALL patients received a median (range) of 2 (1-3) treatment cycles; young adult ALL
patients and LL patients each received 2 (1-2). Among pediatric ALL patients, 10 (41.7%)
patients (90% CI, 24.6-60.3) achieved CR at the end of Cycle 1. ORR was 83.3% (CR,
13 [54.2%] patients; CRi, 7 [29.2%] patients) in pediatric and 60.0% (all CR) in young
adult ALL patients and 40.0% (all CR) in LL patients. Ten (41.7%) pediatric ALL patients
achieved MRD negativity. All pediatric ALL patients had a grade 3/4 TEAE. No pediatric
ALL pt discontinued DARA primarily due to AEs and 1 (4.2%) died due to TEAEs (brain
edema and hepatic failure) attributed to study treatment but unrelated to DARA.
Summary/Conclusion: The addition of DARA to VPLD in pediatric and young adult patients
with relapsed/refractory T-cell ALL or LL showed initial activity, generating improved
response rates compared to those achieved with backbone therapy alone, with a manageable
safety profile.
P361: EARLY MEASURABLE RESIDUAL DISEASE ANALYSIS IS ONLY PREDICTIVE OF SURVIVAL OUTCOMES
AFTER TWO CYCLES OF TREATMENT IN ADULTS WITH B-LYMPHOBLASTIC LEUKEMIA RECEIVING HYPER-CVAD
INDUCTION.
R. Nedumannil1,2,*, D. Ritchie3,4, A. Bajel3,4, A. P. Ng3,5, S. J. Harrison3,4, D.
Westerman1,4
1Department of Pathology, Peter MacCallum Cancer Centre, Melbourne; 2Department of
Diagnostic Haematology, Royal Melbourne Hospital, Parkville; 3Department of Clinical
Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne;
4Sir Peter MacCallum Department of Oncology, The University of Melbourne; 5Immunology
Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
Background: Measurable residual disease (MRD) monitoring using multi-parametric flow
cytometry (MFC) has a well-established role in B-lymphoblastic leukemia (B-ALL). However,
the optimal time-point (TP) for early MRD testing and the associated prognostic impact
remain undefined in adult B-ALL patients receiving Hyper-CVAD induction chemotherapy,
a dose-intensive regimen comprising alternating A cycles (hyperfractionated cyclophosphamide,
vincristine, doxorubicin and dexamethasone) and B cycles (methotrexate and cytarabine).
Aims: To evaluate the utility of MRD analysis by MFC after one cycle (TP1) in comparison
to that after two cycles (TP2) of induction treatment in adult B-ALL patients receiving
frontline Hyper-CVAD chemotherapy.
Methods: A retrospective chart review was performed of consecutive patients older
than 18 years with newly diagnosed B-ALL who received frontline Hyper-CVAD induction
chemotherapy at two Australian tertiary referral centres from 2010-2020. Patient inclusion
required data regarding remission and MRD statuses after each 21-day cycle of induction
treatment (defined as two [A and B] cycles of Hyper-CVAD chemotherapy). Two flow cytometry
panels (a two tube 8-color panel and a single tube 10-color panel) were used; samples
were analyzed using Kaluza Analysis Software (Beckman Coulter) on FACSCanto II (BD
Biosciences) and Navios EX (Beckman Coulter) cytometers, respectively. The sensitivity
of these assays was 0.001%-0.01%. Clinicopathological and survival data [pertaining
to overall survival (OS) and event-free survival (EFS)] were collected from review
of patient records.
Results: 49 of 76 (64%) adult B-ALL patients received frontline Hyper-CVAD chemotherapy
and had available MFC MRD assessments at each of the two TPs. 19 patients (39%) had
Philadelphia chromosome-positive (Ph+) disease and received concomitant tyrosine kinase
inhibitor (TKI). Median times to TP1 and TP2 relative to start of treatment were 21
and 45 days, respectively. At TP1, 40 patients (82%) achieved complete remission (CR),
amongst whom 12 (30%) were MRD negative. At TP2, the majority of patients (48, 98%)
achieved CR, of whom 23 (48%) were MRD negative. Among those who achieved CR after
Hyper-CVAD induction, 25 (52%) received Hyper-CVAD consolidation chemotherapy ± TKI
while 24 (50%) proceeded to alloSCT in first CR.
After cycle 1A, patients with residual morphological disease compared to those who
attained CR had a median OS of 10 months and 44 months, respectively (HR, 6.15; 95%
CI, 1.36-27.94; P = 0.019), while the median EFS was 58 months and not reached, respectively
(HR, 1.31; 95% CI, 0.33-5.17; P = 0.423). MRD negativity at TP1 was not associated
with a significant improvement in either OS (P = 0.426) or EFS (P = 0.335) when compared
to patients with MRD positivity. In contrast, MRD negativity at TP2 was associated
with significantly higher OS (P = 0·005) and EFS (P = 0.047) over patients with MRD
positivity. Multivariate analysis demonstrated that KMT2A-rearrangement and MRD positivity
at TP2 were the only factors (including Ph+ status) that correlated with worse EFS
and OS.
Summary/Conclusion: In the absence of residual morphologic disease, MRD analysis after
one cycle of Hyper-CVAD induction chemotherapy does not correlate with survival outcomes
when compared to MRD testing after two cycles of Hyper-CVAD in adult B-ALL patients.
Although validation with larger studies is required, initial MRD testing after two
cycles of Hyper-CVAD induction (i.e. after day 42) promotes appropriate resource utilization
and ensures no loss in the prognostic power of early MRD monitoring in adult B-ALL.
P362: MINIMAL-LOSS-CYTOMETRY FOR EVALUATION OF CEREBROSPINAL FLUID IN PEDIATRIC ACUTE
LYMPHOBLASTIC LEUKEMIA. SINGLE CENTER STUDY.
M. Nováková1,2,*, I. Janotová2, E. Vodičková3, A. Houdková3, J. Hanzalová4, M. Vášková1,2,
D. Kužílková1,2, E. Mejstříková1,2, L. Šrámková2, J. Starý2, O. Hrušák1,2
1CLIP-Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University;
2Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles
University and University Hospital Motol; 3Department of Clinical Hematology, University
Hospital Motol; 4Department of Immunology, Second Faculty of Medicine, Charles University
and University Hospital Motol, Prague, Czechia
Background: Central nervous system (CNS) involvement at diagnosis of acute lymphoblastic
leukemia (ALL) represents a risk factor for CNS relapse. Currently, examination of
the cerebrospinal fluid (CSF) is performed and evaluated quantitatively using a counting
chamber and qualitatively using cytomorphological analysis of cytospin. Recently,
flow cytometry (FC) evaluation of CSF was shown to provide more reliable results and
to predict relapse risk (de Haas et al., 2021, Thastrup et al., 2020). Independently
of these studies, we applied a lyse-no wash technique aiming at minimizing cell loss
during preparation and at improved cell concentration estimate.
Aims: Our aim was to compare our findings with morphological criteria and published
data and correlate them with clinical outcome in pediatric patients with ALL.
Methods: We included pediatric patients with newly diagnosed ALL (04/2014 – 01/2022),
whose CSF examination was performed in CLIP laboratory (n=208). Patients were classified
as B cell precursor ALL (BCP ALL; 85%) and T ALL (15%) and treated according to following
protocols: AIEOP-BFM ALL 2009 (n=118), AIEOP-BFM ALL 2017 (n=71), EsPhALL(n=8), Interfant06
(n=5), off-protocol (n=6). During follow up, 14 patients relapsed (5 CNS incl. combined,
9 non-CNS). CSF (drawn into tube with no fixative agent and processed within 2 hours)
was concentrated by centrifugation and incubated with combination of antibodies (CD20/CD10/CD45/CD34/CD19/CD3/Syto41
in BCP ALL and CD4/CD99/CD5/CD3/CD7/CD16 + 56/CD8/CD45/Syto41 in T ALL). Residual
erythrocytes were lysed with NH4Cl solution, followed by immediate acquisition on
BD FACS Lyric or BD LSRII cytometers. Blasts were quantified to initial CSF volume.
Cluster of 20 events was required to assign samples as FC positive (FC+). In parallel,
CNS status was concluded according to morphological criteria, being classified as
CNS1 (no blasts), CNS2a/b (≤5 WBC/μL with blasts), CNS2c/CNS3 (≥5 WBC/μL with blasts)
according to treatment protocol guidelines. Continuous variables were compared with
the Mann–Whitney test, survival data were analyzed using Mantel-Cox test. P values<0.05
were considered as significant.
Results: Using FC, we identified atypical blasts in 50 out of the 208 analyzed samples
(24%). Median of analyzed CSF volume was 367µL (range 143-1033µL), which enabled median
sensitivity 0.054 events/µL (range 0.02-0.14ev/µL). Mean blast concentration was 0.47ev/µL
(range 0-25ev/µL) in all samples and 2 ev/µL (range 0.01-25ev/µL, median 0.53ev/µL)
in FC+ samples. We observed significantly higher blast concentration in patients with
T ALL, in patients with CNS2 and CNS3 and those who subsequently relapsed. Patients
with FC+ had significantly lower relapse-free survival than FC- patients (55% vs 94%),
which provided better separation than CNS status (68% vs.85% for CNS2/3 vs. CNS1,
respectively).
Image:
Summary/Conclusion: Using fresh CSF sample with lyse-no wash protocol enabled us to
detect approximately 10x higher blast count (0.24ev/µL in FC+CNS1 and 1.25ev/µL in
FC+CNS2/3) than was recently published (0.015ev/µL in FC+CNS1 and 0.073ev/µL in FC+CNS2/3
in Thastrup et al., 2020), and thus it better correlates to cytomorphological data.
Interestingly, the proportion of FC+ patients in our cohort is comparable to (24%
vs 25%), showing that although in our method we lose fewer cells, we identify the
same proportion of FC+ patients. The presented study shows a higher impact of FC positivity
on relapse risk than described previously. The disadvantage of our method is a need
for a fresh sample, which is investigated locally.
Supported by UNCE/MED/015, NU20J-07-00028.
P363: TREATMENT OF ACUTE LYMPHOBLASTIC LEUKEMIA DURING COVID-19
E. Paramonova1,*, E. Zhelnova1, E. Misyurina1, E. Baryakh1, E. Karimova1, E. Zotina1,
E. Grishina1, D. Gagloeva1, M. Lysenko1
1hematology, moscow city clinical hospital 52, Moscow, Russia
Background: During the coronavirus pandemic, the risk of severe COVID-19 and mortality
are higher in certain groups, in particular in patients with oncohematological diseases.
Acute lymphoblastic leukemia (ALL) is a special group of oncohematological diseases
in which mortality in the era of COVID-19 has increased 2-3 times. Currently, there
is no consensus on the treatment of ALL during coronavirus infection.
Aims: To determine the basic principles and features of the management of patients
with ALL during COVID-19.
Methods: 46 patients with ALL and COVID-19 (men 52.2%, women 47.8%) aged 18-74 years
(median-44.5) were treated at the Moscow City Clinical Hospital 52 on 01.04.20-01.11.21.
B-ALL was 58.7% (27 patients), T-ALL - 34.8% (16 patients), biphenotypic - 4.3% (2
patients), not defined - 2.2% (1 patient), Ph-positive ALL - 17.4% (8 patients). The
status of the disease of patients upon admission to the hospital differed: debut of
ALL - 20 patients (43.5%), remission - 16 patients (34.8%), relapse and refractory
course - 10 patients (21.7%). All patients were treated COVID-19 in accordance with
the current guidelines for the prevention, diagnosis and treatment of COVID-19 (interleukin
6 inhibitor, anticoagulant and antibacterial therapy, glucocorticoids (GCs), human
immunoglobulin (IG) against COVID-19). According to vital indications and with stabilization
of the patient’s condition, 18 patients (39.1%) received chemotherapy (CT).
Results: There were no deaths in the group of patients with remission of ALL. In patients
with the debut of ALL, mortality was 45% (9 patients), in relapse and refractory course
- 50% (5 patients) (p=0.005). Mortality in the group who did not receive CT was 35.7%,
and in the group who received CT - 22.2%. 6 patients with Ph-positive ALL (75.0%)
continued therapy with tyrosine kinase inhibitors (TKI). According to the protocol
for the treatment of ALL, full doses of GCs (100%) and anthracyclines (ATC) (100%)
were used, lumbar punctures (LP) and intrathecal (IT) injections of CT (100%) were
continued. Due to the high risk of thrombotic complications in COVID-19 and asparaginase
therapy, anticoagulant therapy was performed (100%). Prevention of pneumocystis pneumonia
(PCP) (89.1%), antifungal (37.0%) and antibacterial (87.0%) therapy were carried out
in the treatment of COVID-19. With the persistence of COVID-19 and the absence of
antibodies to COVID-19, 2 patients received repeated transfusion of human IG against
COVID-19.
Summary/Conclusion: During the COVID-19 pandemic, patients in remission of ALL coronavirus
infection are treated and controlled. Treatment of COVID-19 in patients with ALL is
carried out according to general protocols for the treatment of COVID-19, taking into
account the peculiarities of nosology (agranulocytosis, high risk of PCP and fungal
infection with long-term therapy of GCs, persistence of COVID-19). When the patient’s
condition is stabilized, the issue of CT should be decided individually in each case,
taking into account all the risks of ALL and COVID-19. During CT, use full doses of
GCs, ATC. In patients with mild and moderate COVID-19, continue LP and IT injections
of CT, therapy with TKI.
P364: DONOR-DERIVED CD19 CAR-T CELLS AND CHEMOTHERAPY PLUS DONOR LYMPHOCYTE INFUSION
IN THE TREATMENT OF RECURRENT CD19+ B-ALL AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL
TRANSPLANTATION
X. Tan1, C. Zhang1, L. Gao1, L. Gao1, P. Kong1, Q. Wen1, S. Lou2, X. Zhang1,*
1Medical center of hematology, Xinqiao hospital, Army medical university; 2Department
of hematology, the second affiliated hospital, chongqing medical universtiy, Chongqing,
China
Background: Donor derived CD19 car T cells plus donor lymphocyte infusion is rarely
used in the treatment of recurrent CD19 positive B-ALL after allogeneic hematopoietic
stem cell transplantation (allo-HSCT)
Aims: To compare the efficacy of donor derived CD19 car T cells and chemotherapy plus
donor lymphocyte infusion in the treatment of recurrent CD19 positive B-ALL after
allogeneic hematopoietic stem cell transplantation (allo-HSCT)
Methods: We retrospectively analyzed 43 patients with B-ALL who relapsed after allo-HSCT
from March 2016 to November 2019,including 22 patients treated with donor derived
CD19-CAR-T cells (CAR-T group) and 21 patients treated with chemotherapy plus donor
lymphocyte infusion (Chemo-DLI group).
Results: The rates of complete remission (77.3%) in the CAR-T group were significantly
higher than those in the Chemo- DLI group (38.1%); The 1- and 2-year leukemia-free
survival(LFS) in the CAR-T group and Chemo-DLI group were 54.5% vs 9.5%, 45.5% vs
4.8%, respectively. The 1- and 2-year overall survival(OS) in the CAR-T group and
Chemo-DLI group were 68.2% vs 19%, 63.6%vs 9.5%, respectively. Six patients (28.6%)
in the Chemo-DLI group developed grade II-IV acute graft-versus-host disease (aGVHD).
Two patients (9.1%) with grade I-II aGVHD were found in the CAR-T group. The incidence
of CRS in the CAR-T group was 86.4%, the grade I-II CRS was59.1%, the grade III CRS
was 27.3%. There was no grade 4 CRS. Two subjects developed ≤grade-2 immune effector
cell-associated neurotoxicity syndrome (ICANS).
Summary/Conclusion: Compared with chemotherapy plus DLI, donor-derived CD19 CAR-T
is more effective and safe in the treatment of recurrent B-ALL after allogeneic HSCT.
P365: INCORPORATION OF NELARABINE (NEL), PEGYLATED ASPARAGINASE (PEG) AND VENETOCLAX
(VEN) IN THE FRONTLINE THERAPY OF ADULT PATIENTS WITH T-ACUTE LYMPHOBLASTIC LEUKEMIA/T-LYMPHOBLASTIC
LYMPHOMA (T-ALL/LBL)
F. Ravandi1,*, E. Jabbour1, N. Jain1, T. Kadia1, B. Gautam1, M. Konopleva1, W. Wierda1,
J. Burger1, G. Issa1, A. Maiti1, H. Balkin1, M. Kelly1, R. Garris1, P. Kebriaei1,
A. Ferrajoli1, G. Garcia-Manero1, Y. Alvarado1, N. Short1, H. Kantarjian1
1The University of Texas - MD Anderson Cancer Center, HOUSTON, United States of America
Background: NEL is effective for patients (pts) with relapsed T-ALL/LBL but its incorporation
into the front-line therapy has not been investigated. In children, addition of NEL
to the induction regimen improved disease-free survival (DFS) but not overall survival
(OS) (Dunsmore KP, JCO, 2020). VEN has shown preclinical and clinical activity in
relapsed T-ALL/LBL.
Aims: We investigated addition of NEL and PEG into the hyperCVAD (HCVAD) regimen in
adult pts with T-ALL/LBL. We then explored the safety and efficacy of addition of
VEN to this regimen.
Methods: Pts with previously untreated or minimally pre-treated T-ALL/LBL were eligible
if they had an ECOG PS ≤3, creatinine ≤ 2 mg/dL, bilirubin ≤ 2 mg/dL and ALT/AST ≤
4 x ULN. Pts received 8 cycles of HCVAD (cycles 1,3, 5, 7) alternating with high dose
ara-C and methotrexate (MTX) (cycles 2, 4, 6, 8) at approximately 3 weeks intervals.
2 cycles of NEL (650 mg/m% daily x 5) were initially administered after the 8 cycles
(cohort 1) but later moved to after cycle 4 (cohort 2) and then PEG (1500 IU/m2 capped
at 3750 IU) was added on day 5 of NEL cycles (cohort 3). More recently, VEN 400 mg
daily on the first 7 days of each of 8 cycles of therapy was added (cohort 4) and
this was later adjusted to be given for 7 days on the induction cycle and reduced
to 3 days per cycle in pts with ETP-ALL or with persistent MRD with no VEN after cycle
1 in all other pts (cohort 5) Pts with ETP-ALL were referred for allogeneic stem cell
transplant in CR. After the completion of the intensive phase, all pts in all cohorts
received 30 cycles of maintenance with monthly POMP and early intensification with
NEL/PEG on cycles 6 and 7 and late intensification with MTX/PEG in cycle 18 and HCVAD
on cycle 19. All pts received 8 intrathecal doses of MTX alternating with ara-C and
mediastinal radiation was considered in pts with balky mediastinal disease.
Results: Between 8/2007 and 12/2021. 120 pts were enrolled in the 5 study cohorts
sequentially (cohort 1= 30, 2= 49, 3=17, 4=16, 5=8)(Table). 74 pts had T-ALL, 45 T-LBL
and 1 biphenotypic T/myeloid leukemia. Median age was 35 yrs (range 18-78), 93 pts
were male (78%) and 106 pts (88%) had PS 0-1. In pts with T-ALL, median bone marrow
blast % was 81% (range, 20-97%). 6 pts (5%) had CNS disease. Cytogenetics was diploid
in 77 pts (64%), abnormal in 34 (28%), and unavailable in 9 pts (7.5%). Pts were further
characterized as thymic (n=53), early T-cell precursor (ETP; n=23), near ETP (N=19)
mature (n=11), not otherwise specified (NOS; n=11), and early-non-ETP (n=3). 25 pts
had received 1-2 prior cycles of therapy prior to enrollment with 18 having achieved
CR. 30-day mortality was 0%. 106 pts (88%) had ≥ one grade 3/4 non-hematological treatment-emergent
adverse events (TEAE) with the most frequent grade 3/4 TEAEs being neutropenic infections
in 92 pts (77%) and elevated liver enzymes in 25 pts (21%). Overall response rate
(CR/CRi/PR) was 97% with CR/CRp in 95/102 pts (93%), PR in 4 (4%), and no response
in 3 (3%). The median no. of cycles to response (including negative PET scans) was
1 (range 1-10). CR/CRp rate in T-ALL was 88% and T-LBL was 100%. At a median follow-up
of 48 months (mo; 2-168), 79 pts (66%) are alive with a median PFS and OS of 135 and
135 mo (Figure) and 73 pts (61%) remain in CR.
Image:
Summary/Conclusion: Addition of NEL/PEG cycles to the HCVAD regimen is feasible and
may be beneficial. Addition of VEN to each cycle is associated with significant myelosuppression.
Lower doses of VEN may benefit selected pts.
P366: BLINATUMOMAB ADDED TO PREPHASE AND CONSOLIDATION THERAPY IN NEWLY DIAGNOSED
PRECURSOR B-ALL IN ADULTS. A PHASE II HOVON TRIAL
A. Rijneveld1,*, P. Gradowska2, M. Bellido3, O. de Weerdt4, A. Gadisseur5, D. Deeren6,
L. van der Wagen7, Y. Jauw8, R. Fijnheer9, D. van Lammerren10, D. Selleslag11, S.
Halkes12, B. Biemond13, D. Breems14, I. Moors15, G. van Sluis16, M. Bakkus17, C. Homburg18,
V. Janda19, V. van der Velden20, J. Cornelissen21
1Hematology; 2HOVON data Center, Erasmus MC, Rotterdam; 3hematology, Univerisity Medical
Center Groningen, Groningen; 4Hematology, Sint Antonius Hospital, Nieuwegein, Netherlands;
5Hematology, Universitair ziekenhuis Antwerpen (UZA), Antwerp; 6Hematology, AZ Delta,
Roeselare, Belgium; 7Hematology, University Medical Center, Utrecht; 8Hematology,
Amsterdam UMC, VU University Medical Center, Amsterdam; 9Hematology, Meander Medical
Center, Amersfoort; 10Hematology, Haga Teaching Hospital, The Hague, Netherlands;
11Hematology, St Jan hospital, Brugge, Belgium; 12Hematology, Leiden University Medical
Center, Leiden; 13Hematology, Amsterdam UMC, Amsterdam Medical Center, Amsterdam,
Netherlands; 14Hematology, ZNA Stuivenberg, Antwerpen; 15Hematology, Universiteits
ziekenhuis Gent, Gent, Belgium; 16Hematology, Isala Clinic, Zwolle, Netherlands; 17Hematology,
University Hospital Brussels, Brussels, Belgium; 18Immunocytology, Sanquin Diagnostic
Services, Amsterdam, Netherlands; 19Immunology, UZ Gent, Gent, Belgium; 20Immunology;
21Hematology, Erasmusm University Medical Center, Rotterdam, Netherlands
Background: The bispecific antibody blinatumomab (blina) is approved for patients(pts)
with relapsed/refractory precursor B-cell ALL (ALL). It has also shown to be highly
effective in MRD+ pts in first line treatment.
TABLE Pt characteristics
Total, N(%)
71(100%)
Age(y), median(range)
53(18-70)
Age (y), N(%)
≤40
22(31%)
40-60
29(41%)
>60
20(28%)
BM blasts (%), N(%)
≤50%
4(6%)
>50%
62(87%)
Unknown
5(7%)
Karyotype, N(%)
Ph+
26/71(37%)
KMT2A
2/67(3%)
Complex
23/67(34%)
Hypodiploidy
5/67(7%)
Aims: To evaluate whether blina added in upfront therapy to prephase and after consolidation
(cons)-1 would increase MRD negativity measured by qPCR or flowcytometry at a cut-off
of <10-4 (primary endpoint). Secondary endpoints included CR, EFS, OS, adverse events
and treatment-related mortality.
Methods: Pts, 18-70 years(y) old, with newly diagnosed CD19+ ALL (incl. Ph+), were
included. Treatment was based on a pediatric inspired protocol (HOVON 70, Rijneveld
et al., 2011) with reduced doses of anthracyclines, MTX, etoposide and PEG-ASP for
pts ≥40y old. Prephase consisted of 10 days steroids, from day 5 combined with 14
days blina in the standard step-up dosing schedule. After cons-1 and after intensification
2, two 4-week blina courses were added irrespective of MRD. The protocol was amended
twice due to toxicity. First, in 2018 the first PEG-ASP administration was omitted.
Second, in 2021 doxorubicin, dexamethasone and PEG-ASP were reduced during intensification
1. Rituximab (if CD20+), prophylactic ITs and imatinib (if Ph+) were standard. AlloHSCT
was offered to intermediate and high-risk pts. Trial was registered with ClinicalTrials.gov,
identifier NCT03541083.
Results: Seventy-one pts were enrolled. Pt characteristics are presented (Table).
Fifteen pts discontinued treatment before blina cons-1 due to refractory disease (n=8),
toxicity (n=7) or death (n=2). In the total study population, 55/71 pts (77%) achieved
CR after (blina) cons-1. Among pts still on treatment after cons-1, 55/56 (98%) pts
achieved CR, 50/55 (91%) reached MRD negativity. After prephase, CR was already reached
in 63% and MRD negativity in 53%. Blina related AEs in prephase were as expected (83%
of pts had ≥1 AE and 10% had ≥1 SAE (hepatotoxicity 3 pts, pain lymph node 1, CRS
1, pneumonia 1, renal insufficiency 1)). CRS was observed in 35% of pts, 32% of whom
experienced grade 3 and no grade ≥4. During prephase 5 pts discontinued blina; during
blina cons-1, 4 pts stopped blina. With a median follow-up of 17,6 months, the estimated
2-y EFS was 64% standard error (SE) ± 7% (≤60y 71% SE ± 9% and >60y 47% SE ± 12%).
Overall, 14 (20%) pts died. OS after 2y was 73% SE ± 7% (≤60y 82% SE ± 8% and >60y
52% SE ± 14%) (Figure 1). For pts with Ph+ ALL, 2-y EFS was 88% SE ± 6%and OS also
88% SE ± 7%. For Ph- ALL, 2-y EFS and OS were 53% SE ± 9% and 68% SE ± 9%, resp. Among
pts who reached CR on protocol (n=60), 5 (8%) had relapse, 6 (10%) died and 6 (10%)
discontinued treatment due to toxicity. Until now, 22 pts proceeded to alloHSCT and
11 with maintenance.
Image:
Summary/Conclusion: Blina can safely be added to prephase of an intensified pediatric
schedule for newly diagnosed ALL up to 70y of age, albeit with dose reductions for
PEG-ASP, doxorubicin and dexamethasone. The combination increases CR and MRD negativity
rate. The early addition of blina resulted in very early achievement of MRD negativity
with 53% after prephase and 91% after blina cons-1. Further reductions of chemotherapy
should be explored (especially for Ph+) if these results are maintained with longer
follow-up.
P367: LONG TERM OUTCOMES OF NEWLY DIAGNOSED CRLF2 REARRANGED B-CELL ACUTE LYMPHOBLASTIC
LEUKEMIA
J. Senapati1,*, E. J. Jabbour1, N. J. Short1, F. Ravandi1, P. Kebriaei1, T. M. Kadia1,
G. Borthakur1, N. Pemmaraju1, R. S. Garris1, D. Bansal1, S. Konoplev1, S. Wang1, W.
Wang1, G. Tang1, K. P. Patel1, M. Konopleva1, N. Jain1
1Leukemia, MD Anderson Cancer Center, Houston, United States of America
Background: CRLF2 rearranged B cell acute lymphoblastic leukemia (B-ALL), a subtype
of Philadelphia (Ph) -like ALL, constitutes a high-risk subset of B-ALL with poor
outcomes with chemotherapy. Targeted therapies such as blinatumomab (blina) or inotuzumab
(ino) may improve treatment outcomes for these patients (pts).
Aims: To study the outcomes of newly diagnosed CRLF2 rearranged B-ALL treated with
hyper CVAD based regimens and the impact of targeted therapies like inotuzumab and
blinatumomab
Methods: We retrospectively analyzed pts with newly diagnosed B-ALL (diagnosed between
01/2001 and 12/2021) at our center who had documented CRLF2 overexpression. Initial
therapy, including use of ino and blina in CR1 were noted. Outcomes measures included
CR/CRi, MRD response, relapse free survival (RFS) and overall survival (OS). RFS was
censored at allogeneic stem cell transplantation (ASCT).
Results: A total of 76 pts with a median age of 38 years (yrs) (range, 18-80) were
identified, of which 70% were males and 81% were of Hispanic ethnicity. All pts had
overexpression of CRLF2 documented by flow cytometry or gene expression profile. A
subset of pts (n=37) had a concomitant CRLF2 FISH performed with all confirming CRLF2
rearrangement. Baseline disease parameters, treatment and outcomes are detailed in
Table 1. Sixty-five pts (85%) received Hyper-CVAD based induction therapy [HCVAD-based
(n=51); mini-CVD-based (n=14)] and 11 (15%) received augmented BFM. We focus on the
outcomes of pts treated with HCVAD/mini-HCVD (n=65); among these pts, 24/65 (37%)
received blina in CR1 during consolidation at a median of 3.6 months after starting
induction therapy, 22/65 (34%) received ino in CR1 (in C1 as part of mini-HCVD-ino,
n=14; in C2 as part of HCVAD, n=8). A total of 14/65 (22%) received both ino and blina
in CR1; 32/65 (49%) received ino and/or blina. The median follow-up was 39 months
(mos). CR/CRi rate after C1 was 52/65 (80%) with 25/47 (53%) MRD evaluable pts and
25/65 (38%) overall achieving MRD-neg post C1. The median RFS and OS was 17.6 and
26.6 mos, respectively (Fig. 1). Among the 32 pts who received ino and/or blina, the
median RFS and OS was 17.6 mos and 38.8 mos respectively. CR/CRp rate after C1 among
pts who received mini-HCVD-ino in C1 was 100% (14/14) with 79% MRD-neg. On landmark
analysis to the time to blina initiation, blina treated pts had similar RFS and trended
for improved OS. A total of 19/65 (29%) had ASCT in CR1; all were MRD-neg prior to
ASCT. Landmark analysis for OS based on the time of ASCT (6 mos) in CR1 favored ASCT
in CR1 (47.2 vs. 17.6 mos, p=0.04).
Table 1:
Disease and treatment parameters
Parameters
N (%) or median [range] (N=76)
Age, yrs
38 [18-80]
Gender, male
53 (70)
Ethnicity
Hispanic
62 (81)
Baseline parameters
WBC (x 109/L)
17 [1-602]
Platelets (x 109/L)
39 [3-195]
PB blasts (%)
72 [0-98]
BM blasts (%)
89 [29-98]
CNS positive
10 (80)
Cytogenetics and molecular
Diploid karyotype
28 (37)
CRLF2 mutation (n=31)
4 (13)
JAK2 mutation (n=67)
25 (37)
JAK1 mutation (n=31)
9 (29)
Frontline regimen
Augmented BFM
11 (15)
HCVAD
51 (67)
Chemotherapy alone
33/51 (65)
with ino alone
2/51 (4)
with blina alone
10/51 (20)
with ino + blina
6/51 (12)
Mini-HCVD-ino
14 (18)
with ino alone
6 (43)
with ino + blina
8 (57)
Image:
Summary/Conclusion: Despite improvements in treatment options, CRLF2 overexpressed
B-ALL continue to have inferior outcomes. Earlier initiation of targeted therapies
might improve outcomes.
P368: A PHASE II STUDY OF INOTUZUMAB OZOGAMICIN FOR THE TREATMENT OF MEASURABLE RESIDUAL
DISEASE-POSITIVE B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA
J. Senapati1,*, H. Kantarjian1, N. Short1, Y. Alvarado1, J. Burger1, N. Jain1, M.
Konopleva1, F. Ravandi1, C. DiNardo1, L. Masarova1, K. Sasaki1, P. A. Thompson1, A.
Ferrajoli1, J. O. Jacob1, E. D. Mayor1, A. M. Milton1, C. Loiselle1, R. S. Garris1,
E. J. Jabbour1
1Leukemia, MD Anderson Cancer Center, Houston, United States of America
Background: Persistence or re-emergence of measurable residual disease (MRD) in B-cell
acute lymphoblastic leukemia (B-ALL) is strongly associated with shorter relapse-free
survival. Inotuzumab ozogamicin (INO) is an anti-CD22 antibody-drug conjugate with
the potential to eradicate MRD in B-ALL.
Aims: We aimed to evaluate the efficacy of INO in clearing MRD in patients (pts) with
persistent or recurrent MRD positivity after treatment with conventional chemotherapy.
Methods: This is a single arm phase II trial of pts with B-ALL in complete remission
(CR) who did not achieve MRD negativity (MRD-ve) or had MRD-positive (MRD+ve) relapse
after at least 3 months (mos) from the start of frontline therapy (i.e. CR1) or 1
month from the start of any salvage therapy (i.e. ≥ CR2). Eligibility was defined
by MRD+ve at ≥ 0.01%. MRD-ve was defined as undetectable MRD by flow cytometry at
a minimum sensitivity of 10-4 for Philadelphia (Ph) negative (Ph-) B-ALL and undetectable
MRD by both flow and PCR at 10-4 for Ph positive (Ph+) B-ALL. INO was given at a dose
of 0.6 mg/m2 on D1 and 0.3 mg/m2 on D8 of C1 and 0.3 mg/m2 on D1 and 8 of subsequent
cycles (up to 6 total cycles, given every 21-28 days) along with ursodiol prophylaxis.
Pts with Ph+ ALL received concomitant TKI, the choice of which was decided by the
treating physician.
Results: Between 11/2018 and 1/2022, 20 pts with MRD+ve B-ALL, with a median age of
40 years (range, 19-68) were treated. Twelve pts (60%) had Ph+ B-ALL, 11 of whom received
concurrent ponatinib and 1 dasatinib. Eight pts were Ph- (including 3 Ph-like ALL).
Fourteen pts (70%) were in CR1, and 6 pts (30%) were in ≥ CR2 (3 in CR2, 2 in CR3
and 1 in CR4). Eleven pts (55%) had received prior blinatumomab (blina) and 4 pts
(20%) had prior stem cell transplantation (SCT). The median BCR-ABL1 level in Ph+
B-ALL was 0.58% (range, 0.001-17.5%) and median MRD level by flow cytometry in Ph-
B-ALL was 0.2% (range, 0.05-1.24%). Twelve pts (60%) became MRD-ve (responders), 10
after cycle 1 and 2 after cycle 2. In the Ph+ group, 6 pts (50%) responded; another
3 pts attained MMR as best response. Six of 8 (75%) Ph- pts responded. The median
number of cycles were 3 (range, 1-6). Seven of 9 pts (78%) without prior blina exposure
became MRD-ve, compared with 5/11 (45%) pts with prior blina exposure (p=0.22). Patient
disposition is shown in Fig. 1A. Amongst the 12 responders, 5 pts were consolidated
with ASCT after a median of 3 cycles (range, 1-3) of INO, all in MRD-ve CR pre-ASCT.
Three responders (25%) relapsed (including 1 relapse after ASCT). At a median follow-up
of 17 mos, 15 pts (75%) are alive, 8 of whom are in continued MRD-ve CR. The estimated
18 mos RFS and OS were 63% and 73% respectively, for the whole group (Fig. 1B), and
the 18 mos OS was 87% and 55% for responders and non-responders, respectively. Outcomes
were similar between pts with Ph+ ALL vs. Ph- ALL, prior blina exposure vs. no prior
exposure, and pts in CR1 vs. ≥ CR2.
INO was overall well-tolerated with no grade 4 non-hematological toxicities. Grade
3 non-hematological toxicities included elevated transaminases in 1 pt and veno-occlusive
disease in 1 pt without history of prior ASCT. Ten pts (50%) had grade 3-4 hematological
toxicities (primarily neutropenia).
Image:
Summary/Conclusion: Low-dose, fractionated INO is a well-tolerated option for MRD
eradication in Ph+ and Ph- B-ALL pts, including in those with prior blina exposure.
P369: A PHASE II STUDY OF MINI-HYPER-CVD PLUS VENETOCLAX IN PATIENTS WITH PHILADELPHIA
CHROMOSOME-NEGATIVE ACUTE LYMPHOBLASTIC LEUKEMIA
J. Senapati1,*, H. Kantarjian1, N. Short1, M. Konopleva1, F. Ravandi1, N. Jain1, P.
A. Thompson1, N. Pemmaraju1, W. G. Wierda1, G. Borthakur1, T. M. Kadia1, G. Garcia-Manero1,
M. Yilmaz1, J. Thankachan1, M. Zhao1, C. Loiselle1, M. T. Talley1, M. I. Kwari1, R.
S. Garris1, E. J. Jabbour1
1Leukemia, MD Anderson Cancer Center, Houston, United States of America
Background: Venetoclax (VEN), an orally active Bcl-2 antagonist, has shown activity
in acute lymphoblastic leukemia (ALL) in preclinical models and in combination with
navitoclax (BCL-XL inhibitor) in early clinical studies. Combining VEN with other
active agents in ALL might synergize this antileukemic effect.
Aims: We aimed to evaluate the efficacy and tolerability of VEN added to mini-hyper
CVD (mHCVD) chemotherapy in patients (pts) with relapsed/refractory (R/R) ALL.
Methods: Pts ≥18 years of age with R/R Philadelphia chromosome negative (Ph-) B- or
T-cell ALL/lymphoblastic lymphoma (LBL) were eligible. Pts were required to have a
PS of ≤3, bilirubin ≤1.5 mg/dl, AST/ALT ≤3 x ULN and creatinine ≤2 mg/dl. Treatment
consisted of mHCVD for up to 8 cycles. VEN was given at a dose of 400 mg daily on
D1-14 of C1 and on D1-7 of C2-8. Rituximab (if CD20+) and prophylactic IT chemotherapy
x 8 doses were given in C1-C4. Pts with T-cell ALL received an additional 2 cycles
of nelarabine (650 mg/m2 daily on D1-5) and peg-asparaginase (1,500 IU/m2 [capped
at 3750 IU] on D5), without VEN, during consolidation and another 2 cycles during
maintenance. Responding pts received vincristine and prednisone maintenance with VEN
daily on D1-14 of each 28-day cycle for up to 2 yrs.
Results: From 6/2019 to 2/2021, 20 pts with R/R ALL were treated, 15 (75%) with B-ALL,
4 (20%) with T-ALL (including 1 ETP-ALL) and 1 (5%) with T-LBL. The median age was
45 yrs (range, 20-70). The median lines of prior therapy was 2 (range 1-5). Of the
15 B-ALL pts, 6 had previously received both blinatumomab (blina) and inotuzumab (INO),
7 had received blina without INO, and 2 had received neither. Overall, 11 pts (55%)
had undergone prior stem cell transplantation (SCT).
Pt disposition is shown in Fig. 1A. One pt was in measurable residual disease (MRD)-positive
CR at trial enrollment; all others had active disease and were evaluable for response
assessment. Pts received a median of 2 cycles of mHCVD-VEN (range, 1-6). Overall,
12 of 19 evaluable pts (63%) responded (9 CR, 3 CRi), 9 (75%) of whom after 1 cycle.
Response in B-ALL was 9/15 (60%) and in T-ALL was 3/4 (75%). Six of the 12 responders
(50%) attained MRD negativity (MRD-ve). Of the 12 responders, 4 (33%) underwent SCT,
3 of whom subsequently relapsed and 1 of whom is in continued MRD-ve CR. All of the
8 responders (67%) who did not undergo SCT, subsequently relapsed and died, with a
median duration of response of 2.1 mos. Six of the 7 non-responders have died. With
a median follow up of 15 months (mos), 5 pts are alive with a median RFS and OS of
6.2 and 7.1 mos respectively (Fig. 1B. The median OS in responders was 8.8 mos (Fig.
1C).
Fig.1: (A) Patient disposition and survival, (B) OS and RFS for the entire cohort,
(C) OS of responders and non-responders
The therapy was generally well-tolerated. There were no grade 4-5 adverse events.
Four pts had a grade 3 related event (hyperbilirubinemia and mucositis in 1 pt and
transaminitis, fatigue and tumor lysis syndrome in 1 pt each). One pt required VEN
dose reduction due to cytopenias. The median times to neutrophil and platelet recovery
in C1 in responding pts were 18 and 27 days, respectively.
Image:
Summary/Conclusion: Low dose combination chemotherapy with VEN in a population of
heavily pretreated R/R ALL was well-tolerated and resulted in a response rate of 63%.
This study has now been amended to add navitoclax in an attempt to further improve
outcomes.
P370: EARLY RESULTS OF A SAFETY AND EFFICACY STUDY OF ALLOGENEIC TRUUCAR™ GC502 IN
PATIENTS WITH RELAPSED/REFRACTORY B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA (R/R B-ALL)
S. Li1,*, Z. Yuan1, L. Liu1, Y. Li1, S. Martina2, J. Liu2, Z. Li2, X. Wang2, J. He2,
W. Zhao2, L. Shen2, X. Zhang3, S. Wang1
1920th Hospital, Kunming; 2Gracell Biotechnologies Ltd, Shanghai; 3The Second Affiliated
Hospital of Army Medical University, Chongqing, China
Background: CD19 targeted autologous CAR-T therapies have been approved for the treatment
of r/r B-ALL and greatly improved outcome. However, some patients may not be eligible
to receive autologous CAR-T. TruUCAR™ GC502 is an allogeneic, universal CAR-T product
with CD19/CD7 dual directed CAR. Preclinical data of GC502 were reported at ASH 2021
(Abstract 148500).
Aims: Here, we report early clinical results from a phase I open-label, non-randomized,
prospective investigator initiate trial (IIT) of GC502 in r/r B-ALL patients to evaluate
safety and preliminary efficacy
Methods: GC502 is manufactured using leukopaks from HLA-unmatched healthy donors.
It contains a 4-1BB based CD19/CD7 dual directing CAR, a T cell enhancer, and genetically
disrupted TRAC and CD7 loci to avoid GvHD and fratricide.
Patients (pts) with r/r B-ALL were enrolled and treated with one of two different
formulations (A or B) in escalating dose levels ranging from 1.0x107 (DL1) to 1.5x107
(DL2) cells/kg. Prior to infusion of GC502, pts received a Flu/Cy based lymphodepletion
regimen. Adverse events, disease response and expansion kinetics were evaluated in
this study.
Results: At date cut off of Feb. 22, 2022, 4 pts (15-34 yrs) had been enrolled into
the investigator initiate study of GC502 (NCT05105867). All of patients were heavily
pretreated, and had received either autologous or donor derived CD19 or CD19-CD22
targeted CAR-T in prior lines of therapy. Baseline marrow blast levels ranged from
19.5% to 92% (median 48.1%). 1 pt had extramedullary (EM) involvement.
At data cut-off all pts had received a single dose of GC502: 1 pt at DL 1 1.0x107cells/kg
in formulation A and 3 patients at DL 2 1.5x107cells/kg – out of which 2 were treated
with formulation B. At day 28 post CAR-T infusion, 3 out of 4 response evaluable patients
had achieved CR/CRi; 1 pt with EM achieved PR at month 1 and subsequently received
allo-HSCT on day 39.
TEAEs presented as Gr 3 febrile neutropenia (4/4), Gr 4 thrombocytopenia (1/4) and
Gr 3 anemia (3/4). All TEAE resolved after treatment with SOC. Non-hematological TEAE
presented as Gr≥3 γ-GT increase (3/4), Gr≤3 AST increase (2/4) and Gr≤3 ALT increase
(3/4). 2 pts received formulation A and experienced Gr 3 CRS with a duration of 7
and 10 days respectively (CRS was graded according ASTCT Consensus Grading). CRS presented
as Gr 2 in the 2 patients with formulation B with a duration of 9 and 15 days respectively.
CRS in all pts was manageable and resolved after treatment with Ruxolitinib, SOC and
supportive care. No ICANS or aGvHD were observed. The pt treated in DL 1 did not show
adequate GC502 cellular expansion. Peak expansions of GC502 in peripheral blood were
observed between week 1-2 in DL 2. Median peak CAR copies were 149,945 copies/ug DNA
(range 10,849-195,400).
Summary/Conclusion: TruUCAR™ GC502 demonstrated promising early results with a manageable
safety profile. Robust CAR-T cell expansion was observed in DL2 at 1.5x107cells/kg
in heavily pretreated r/r B-ALL patients, including those previously treated with
CD19 or CD19-CD22 CAR-T therapies. The study is ongoing and continues accruing patients.
P371: HYPER-CVAD WITH SEQUENTIAL BLINATUMOMAB, WITH OR WITHOUT INOTUZUMAB OZOGAMICIN,
IN ADULTS WITH NEWLY DIAGNOSED PHILADELPHIA CHROMOSOME-NEGATIVE B-CELL ACUTE LYMPHOBLASTIC
LEUKEMIA
N. Short1,*, H. Kantarjian1, F. Ravandi1, M. Yilmaz1, T. Kadia1, P. Thompson1, X.
Huang2, M. Konopleva1, A. Ferrajoli1, N. Jain1, K. Sasaki1, Y. Alvarado1, G. Borthakur1,
C. Dinardo1, M. Ohanian1, W. Macaron1, S. Kornblau1, M. Zhao1, M. Kwari1, C. Loiselle1,
R. Delumpa1, A. Milton1, J. Rivera1, S. Lewis1, R. Garris1, E. Jabbour1
1Department of Leukemia; 2Department of Biostatistics, The University of Texas MD
Anderson Cancer Center, Houston, United States of America
Background: Blinatumomab and inotuzumab ozogamicin (INO) are highly effective in relapsed/refractory
B-cell acute lymphoblastic leukemia (ALL); blinatumomab is also the only approved
therapy for eradication of persistent or recurrent measurable residual disease (MRD)
after initial ALL therapy. We hypothesized that early incorporation of blinatumomab
and INO in patients (pts) with newly diagnosed Philadelphia chromosome (Ph)-negative
B-cell ALL would lead to deeper and more durable responses, reduce relapses, and improve
survival.
Aims: We evaluated the efficacy and safety of hyper-CVAD with sequential blinatumomab,
with or without INO, in pts with newly diagnosed Ph-negative B-cell ALL.
Methods: Pts 14-59 years of age with newly diagnosed Ph-negative B-cell ALL, including
pts who had received no more than 1 prior cycle of chemotherapy, were eligible. Pts
were required to have a performance status of ≤3, total bilirubin ≤2 mg/dl, creatinine
≤2 mg/dl, and no significant CNS pathology (with the exception of CNS leukemia). Pts
received hyper-CVAD alternating with high-dose methotrexate and cytarabine for up
to 4 cycles, followed by 4 cycles of blinatumomab at standard doses. Pts with CD20+
disease (≥1% cells) received 8 doses of ofatumumab (2000 mg) or rituximab (375 mg/m2).
Eight doses of prophylactic IT chemotherapy were given. Maintenance was with alternating
blocks of POMP (given in maintenance cycles 1-3, 5-7, 9-11, and 13-15) and blinatumomab
(given in maintenance cycles 4, 8, and 12). Those with high-risk disease features
started blinatumomab after 2 cycles of hyper-CVAD. Beginning with pt #39, INO at a
dose of 0.3 mg/m2 on day 1 and 8 was added to the 2 cycles of MTX/Ara-C and to 2 cycles
of blinatumomab consolidation (4 total cycles with INO).
Results: As of February 2022, 58 pts have been treated (38 without INO and 20 with
INO). Pt characteristics are summarized in Table 1. Median age was 34 years (range,
17-59 years). 13 pts were in CR at enrollment.
Among 45 pts with active disease at study entry, 100% achieved CR, with 80% achieving
CR after the first cycle. MRD negativity by 6-color flow cytometry was achieved in
32/42 evaluable pts (76%) after 1 cycle and 55/58 evaluable pts (95%) overall. Two
of the 3 pts who did not achieved MRD negativity were later found to have a NUP214::ABL1
fusion.
The median duration of follow-up is 26 months (range, 3-61 months). Overall, 5 pts
(9%) did not undergo stem cell transplant (SCT) and subsequently relapsed, 18 pts
(34%) underwent SCT in first remission (including 2 additional pts who relapsed post-SCT),
2 pts (3%) died in CR, and 33 pts (57%) remain in continuous remission without SCT.
All relapses occurred in pts with ≥1 poor-risk feature(s), and no relapses have occurred
beyond 2 years from the start of treatment.
For the entire cohort, the 3-year continuous remission and OS rates were 84% and 85%,
respectively (Figure 1). No relapses or deaths have occurred in the INO group, and
the estimated 1-year OS rate is 100%.
Treatment was overall well-tolerated. One pt discontinued blinatumomab due to a related
adverse event (grade 2 encephalopathy and dysphasia). No pts discontinued INO due
to toxicity, and no cases of veno-occlusive disease have been observed.
Image:
Summary/Conclusion: Hyper-CVAD with sequential blinatumomab, with or without INO,
is highly effective as frontline treatment of Ph-negative B-cell ALL, with an overall
MRD negativity rate of 95% and a 3-year OS rate of 85%. Early outcomes with the addition
of INO to this regimen are encouraging, with no relapses or deaths observed to date.
P372: DISMAL OUTCOMES OF PATIENTS WITH RELAPSED/REFRACTORY B-CELL ACUTE LYMPHOBLASTIC
LEUKEMIA AFTER FAILURE OF BOTH INOTUZUMAB OZOGAMICIN AND BLINATUMOMAB
W. Macaron1,*, E. Jabbour1, M. Konopleva1, F. Ravandi1, N. Jain1, G. Issa1, T. Kadia1,
K. Sasaki1, P. Kebriaei1, M. Yilmaz1, P. Thompson1, K. Takahashi1, H. Abbas1, W. Wierda1,
R. Garris1, H. Kantarjian1, N. Short1
1Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston,
United States of America
Background: The development of novel monoclonal antibodies such inotuzumab ozogamicin
(INO) and blinatumomab (Blina) results in superior response rates and survival in
patients (pts) with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia
(ALL). However, outcomes of pts after failure of both INO and Blina are not well-established,
and effective treatment options for these pts are limited.
Aims: To determine response rates to subsequent therapies and overall survival (OS)
in pts with R/R Philadelphia chromosome (Ph)-negative B-cell ALL after failure of
both INO/Blina.
Methods: We conducted a retrospective analysis of outcomes in adult pts with Ph-negative
B-cell ALL who relapsed or were refractory to both INO/Blina, at least one of which
given as salvage therapy.
Results: The baseline characteristics of the 65 pts included are shown in Table 1.
Of 29 pts with paired flow cytometry for CD19 and CD22 prior to receipt of either
INO/Blina, 4 pts (14%) lost CD19 expression and 3 pts (10%) lost CD22 expression at
time of INO/Blina failure.
After INO/Blina failure, 54 pts (83%) received subsequent therapy, with a median of
2 therapies (range, 1-6). Of 53 evaluable pts, 16 pts (30%) achieved CR/CRi with the
first salvage therapy received after INO/Blina failure (CR=9%, CRi=21%), and 22 pts
(42%) achieved CR/CRi with at least one salvage therapy (CR=17%, CRi=25%). Of the
22 responders, 16 pts (73%) achieved measurable residual disease (MRD) negativity
by flow cytometry (sensitivity=0.01%) at best response. Nine responses (41%) were
achieved with an investigational drug/regimen and 13 (59%) were achieved with commercial
agent(s). Among 54 pts who received at least one subsequent therapy, 12 pts (22%)
received CAR T-cells, and 9 pts (17%) proceeded to hematopoietic stem cell transplant
(HSCT).
With a median follow-up of 22.3 months, the median OS from the time of INO/Blina failure
for the entire population was 3.8 months, and the 1-year OS was 22% (figure 1A). Median
relapse-free survival (RFS) was 3.5 months, with a 1-year RFS of 12%. Median duration
of response (DOR) to subsequent therapy was 5.0 months, with a 1-year DOR of 17%.
Median OS from INO/Blina failure in pts who received salvage therapy compared to those
who did not were 4.7 months and 1.4 months, respectively (figure 1B). In the 9 pts
who underwent HSCT, 2 (22%) are still alive and in continuous remission. The median
OS from time of HSCT was 13.4 months, and the 1-year post-HSCT OS was 59%. In the
12 pts who received CAR-T cells as a subsequent therapy, 6 (50%) responded and 2 were
bridged directly to HSCT. The median OS from time of CAR T-cell therapy was 3.9 months,
and the 1-year OS was 36%.
Outcomes of pts who received both INO and Blina as salvage therapy (n=50), excluding
those who received 1 of these agents in the frontline or MRD setting, were also analyzed.
Median OS from the time of INO/Blina failure was 2.6 months. Thirty-nine (78%) of
these 50 pts received subsequent therapy after INO/Blina failure. Of 38 evaluable
pts, 15 pts (39%) achieved CR/CRi with at least one subsequent salvage therapy (CR=13%,
CRi=26%). Median RFS and OS in pts who underwent subsequent therapy were 5.9 months
and 3.9 months, respectively.
Image:
Summary/Conclusion: This study highlights the very poor outcomes of pts with R/R B-cell
ALL after failure of both INO/Blina. For pts who respond to subsequent salvage therapy,
a consolidative approach with SCT may improve outcomes. These data provide a historical
reference for expected outcomes that may serve as a benchmark for the evaluation of
novel drugs and combinations in the setting of INO/Blina failure.
P373: CIRCULATING ENDOTHELIAL PROGENITOR CELLS AND METABOLIC FACTORS IN CHILDHOOD
CANCER SURVIVORS
E. Athanasopoulos1,*, G. Martimianaki1, P. Iordanis1, M. Stratigaki1, N. Katzilakis1,
E. Stiakaki1
1Department of Pediatric Hematology-Oncology and Laboratory of Blood Diseases and
Childhood Cancer Biology, University Hospital of Heraklion, Medical School, University
of Crete, Heraklion Crete, Greece
Background: Circulating Endothelial Progenitor cells (cEPCs) participate in the regulation
and maintenance of vascular integrity, balancing the coagulation–anticoagulation mechanisms,
as well as the immune response. Obesity and hypertension constitute late effects of
chemotherapy in children and numerous studies have shown a decrease in cEPCs number,
underlining poor vascular repair capability.
Aims: The determination of cEPCs in children treated for Acute Lymphoblastic Leukemia
(ALL), Lymphomas (LYM) and solid tumors (ST) and the study of their levels in correlation
with patients’ Body Mass Index (BMI) and blood pressure (BP) in different post-treatment
time points.
Methods: Peripheral blood from children with ALL (n=166), LYM (n=37), ST (n=109) and
children without malignancies (Control, n=191) were studied. The cEPCs population
was determined by Flow cytometry based on CD34, CD45, CD133, VEGFR2 expression. The
BMI and the blood pressure (BP) values were registered and the corresponding percentiles
calculated. Patients were divided into three groups: <1 year post-treatment, ≤1-3
years post-treatment, and ≥3 years after completion of treatment. Statistical analysis
was performed using t-test (Holm-Sidak) and 2way ANOVA (Tukey’s multiple comparisons
test).
Results: In ALL group decreased levels of CD34+VEGFR2+ and CD34+CD133+VEGFR2+ were
estimated in contrast with the LYM, ST and Control group (p=0,0144, 0,0262 respectively).
Decreased levels of both populations were observed immediately after treatment completion
in ALL, while in the post treatment period the levels gradually increased. On the
contrary, in the LYM and ST group, at completion of treatment higher level of cEPCs
were measured which gradually decreased (p= 0,0405 & 0,0026 respectively). The obesity
was correlated with higher levels of cEPCs in the ST and LYM compared to the ALL group.
The hypertensive patients had elevated cEPCs.
Image:
Summary/Conclusion: Conditions causing vascular damage such as obesity and hypertension
are correlated with increased cEPCs in patients treated for ALL, LYM and ST. The effect
of treatment seems to have a different impact in these groups, possibly not only related
with the biology of the disease but with the differences of treatment protocols as
well. The increase of cEPCs during the 1st post-treatment year probably reflects an
effort to repair vascular damage caused by the treatment. Further investigation is
needed to clarify these results.
P374: BLINATUMOMAB AS CONSOLIDATION FOR ADULT B-CELL PRECURSOR ACUTE LYMPHOBLASTIC
LEUKEMIA. A REAL-WORLD STUDY
I. Urbino1,*, E. Lengline2, M. Cerrano1, F. Rabian2, R. Kim3, M. Sebert2, R. Itzykson2,
E. Audisio1, L. Ades2, H. Dombret2, E. Raffoux2, D. Ferrero4, E. Clappier3, N. Boissel2
1Hematology Department, AOU Città della Salute e della Scienza di Torino, Torino,
Italy; 2Hematology Department; 3Hematology Laboratory, Saint-Louis Hospital - Université
de Paris, Paris, France; 4Department of Molecular Biotechnology and Health Sciences,
Division of Hematology, University of Torino, Torino, Italy
Background: Blinatumomab is a CD3/CD19 bispecific T-cell engager approved for the
treatment of relapsed/refractory (R/R) or minimal residual disease (MRD)-positive
B-cell precursor acute lymphoblastic leukemia (B-ALL). Recent phase 3 studies in children
and young adults with B-ALL in first relapse suggested that blinatumomab used as consolidation
after chemotherapy salvage could be more beneficial than given as single agent in
overt relapse. Whereas current strategies aim to demonstrate the benefit of blinatumomab
in first line, the optimal use of blinatumomab in adult patients with first B-ALL
relapse still deserves to be further explored
Aims: To evaluate the efficacy of blinatumomab given as consolidation in adults with
B-ALL in a real-world setting
Methods: We retrospectively included 115 consecutive patients with B-ALL treated with
blinatumomab from April 2012 until June 2021 at Saint-Louis Hospital, Paris, France
(n=100), and at AOU Città della Salute e della Scienza, Turin, Italy (n=15). Patients
included in clinical trials were excluded. Patients were divided in three subgroups:
68 patients treated in 1st complete remission (CR1), 31 patients treated in 2nd CR
after chemotherapy-base salvage therapy (CR2), and 16 patients treated in overt relapse
(R/R). Patients in CR1 received blinatumomab for MRD persistence (n=59/68, 87%) or
due to inability to receive standard consolidation (n=9/68, 13%, off-label use). The
number of blinatumomab cycles along with the use of chemotherapy and/or of allogeneic
hematopoietic stem cell transplant (alloHSCT) after blinatumomab was up to physician
choice.
Results: The median age of patients was 37 years (range, 15-84). Among the 115 patients,
24% (n=28) were Philadelphia (Ph)-positive. Age, sex, baseline disease features and
genetic risk categories did not differ between subgroups. After blinatumomab a complete
MRD-response was achieved in 83% of CR1 and 86% of CR2 patients (p=.99). A complete
remission was reached by 9/15 R/R patients (60%). In the 3 subgroups, the median number
of blinatumomab cycles given was 2 (range, 1-6). Forty-six patients (42%) treated
in CR (41% CR1, 45% CR2) and 4 R/R patients (25%) were bridged to allo-HSCT in continuous
CR after blinatumomab. With a median follow up of 3.1 years, 3-year DFS was 68% in
CR1 and 67% in CR2 patients (p=0.41); 3-year OS was 80% in CR1 and 71% in CR2 patients
(p=0.32). In R/R patients, 3-year DFS and OS were 13% and 20% respectively. Considering
patients who received blinatumomab in CR (CR1+CR2), univariate analysis showed that
higher MRD levels both before and after blinatumomab were associated with shorter
DFS, while only MRD response to blinatumomab was associated with OS. Both pre- and
post-blinatumomab MRD levels retained significance in bivariate analysis for DFS
Image:
Summary/Conclusion: The present study underlines the efficacy of blinatumomab in consolidation
after chemotherapy-based salvage, showing comparable outcomes between patients treated
in CR2 and CR1. In CR2 patients, promising DFS and OS were observed as compared to
historical cohorts of patients treated with chemotherapy alone or with blinatumomab
in overt relapse. Our data suggest that blinatumomab should be preferably used in
consolidation rather than as salvage therapy in patients with B-ALL in first relapse
P375: CHALLENGES IN MANAGEMENT OF ACUTE LYMPHOBLASTIC LEUKAEMIA IN A RESOURCE CONSTRAINED
SETTING IN LOWER-MIDDLE-INCOME COUNTRIES (LMICS)
T. Vaid1,*, R. Dhawan1, M. Aggarwal1, P. Kumar1, J. Dass1, G. VIswanathan1, T. Seth1,
S. Tyagi1, M. Mahapatra1
1Hematology, All India Institute of Medical Sciences, Delhi, India
Background: Improvement in supportive care, introduction of more intensive chemotherapy
regimens and strategic use of allogenic stem cell transplant in some patients has
resulted in significant improvement in outcomes of patients with acute lymphoblastic
leukemia (ALL). However, this improvement is largely restricted to the developed world.
We explore these challenges encountered in treating ALL in one of the largest government
hospitals in India.
Aims: This study aims to assess the baseline characteristics, outcomes and challenges
in management of acute lymphoblastic leukaemia in a resource constrained setting in
lower-middle-income countries (LMICs)
Methods: Consecutive patients diagnosed with acute lymphoblastic leukaemia in the
department of Hematology from 1 January 2017 to 31 December 2019 were included. Pediatric
patients were treated with standard or augmented BFM protocols depending on risk stratification.
Adolescent and yound adults (AYA) were treated with pediatric inspired protocols or
GMALL like protocols. Adult patients were treated with GMALL like protocol.
Results: Of the 273 patients diagnosed with ALL, only 197 (72%) were able to get admitted
for treatment at our institute. Median time from diagnosis to admission was 12.5 days
(range: 0 -190 days) and the median distance of the patient’s permanent residence
and our institute was 217 km (range 3-2400km). Baseline characteristics of these 197
patients are mentioned in Table 1.
Of the 197 patients treated at our centre, only 165 (83.7%) were able to receive induction
chemotherapy. Of the remaining 32 patients, 26 (13.2%) died before treatment could
be initiated and 6 (3%) were lost to follow up. Patients who died prior to starting
chemotherapy had significantly higher incidence of baseline infection and organ dysfunction
compared to those receiving induction.
Neutropenic fever was present in 50.56% patients at the time of admission. Focus of
infection was lungs in 68.5%, sinusitis in 6.8%, skin & soft tissues in 5.6%, neutropenic
enterocolitis in 2.2%, pyelonephritis in 1.1%, splenic abscess in 1.1% & bacteraemia
with no obvious focus in 14.7% patients. 58.6% of the patients developed a new episode
of neutropenic fever during induction chemotherapy. Post induction, a cumulative of
70 episodes of neutropenic fever and other infections were noted in 57 patients, of
these 19 patients needed hospitalization.
At the end of induction, 64.5% (n=127) patients achieved complete remission. Induction
mortality was 27.9% (n=55) and 4.1% (n=8) patients were lost to follow up. Primary
refractory disease was noted in 3.5% (n=7) patients.
After a follow up period of 4 years, only 22.8% (n=45) patients were alive and on
regular therapy, 32% (n=63) patients died & 25.4% (n=50) patients were lost to follow
up. CNS relapse & medullary relapse was noted in 7.1% (n=14) & 9.1% (n=18) patients
respectively.
4-year event free survival was 57% for both standard risk and high risk paediatric
ALL patients respectively, 44% & 35% for standard risk and high risk AYA patients
respectively & 20% and 27% for standard risk and high-risk adult ALL patients.
Image:
Summary/Conclusion: ALL patients in our study had a high incidence of infection and
organ dysfunction at baseline that complicated therapy. With limited access to healthcare,
poor socio-economic status and high incidence of infection, developing countries face
a unique set of challenges. Outcomes of ALL patients in our cohort are at odds with
those reported from the developed world.
P376: CD38: A FUNCTIONING TARGET IN RELAPSED/REFRACTORY ACUTE LYMPHOBLASTIC LEUKEMIA.
LIMITATIONS IN TREATMENT AND DIAGNOSTICS.
B. Vakrmanova1,*, M. Novakova1, P. Riha2, M. Zaliova1, E. Fronkova1, E. Mejstrikova1,
L. Reznickova Rezkova1, J. Stary2, O. Hrusak1, L. Sramkova2
1Department of Paediatric Haematology and Oncology, CLIP, Second Faculty of Medicine
and Motol University Hospital; 2Department of Paediatric Haematology and Oncology,
Second Faculty of Medicine and Motol University Hospital, Prague, Czechia
Background: The prognosis of relapsed T-acute lymphoblastic leukemia (ALL) is dismal
and there is a need for new treatment options. Daratumumab, a monoclonal kappa chain
antibody against CD38 is routinely used in multiple myeloma treatment. CD38 is also
expressed in malignant cells of most cases with pediatric ALL. Accordingly, daratumumab
can be used experimentally in treatment of relapsed ALL but data about efficacy of
such a treatment is limited. The loss of CD38 described in myeloma patients can be
one of the reasons of treatment failure.
Aims: Does daratumumab provide a benefit in relapsed ALL?
Can we observe the inability of anti-CD38 mAbs to bind to leukemia cells after daratumumab
treatment as described in myeloma patients?
Methods: We treated five patients with relapsed ALL with daratumumab between 10/2019
– 10/2021 (four of them for a first relapse of T-ALL and one for a second CD19neg
relapse of B cell precursor (BCP) ALL). In three patients, daratumumab was used in
combination with chemotherapy, in one patient, chemotherapy was early discontinued
for toxicity and the remaining patient received it in monotherapy due to clinical
condition. Blast positivity of CD38 by a diagnostic monoclonal antibody (mAb) at relapse
was confirmed in all patients before treatment started.
Results: Three of five patients responded to daratumumab plus chemotherapy and were
in second complete remission (CR2) and underwent stem cell transplantation (SCT).
CD38 expression on blasts after daratumumab was not evaluated due to blast elimination
in these cases. Two of these patients relapsed with CD38pos ALL 5 and 7 months after
SCT, respectively. Third patient is in CR2 one year after SCT. In two of five patients
(one with T-ALL and one with a CD19neg BCP ALL) blasts were still detected by flow
cytometry after daratumumab. In patient 4 disease progressed under daratumumab treatment.
Under the progressing disease patient died 52 days after the relapse. In the other
patient, the amount of blasts decreased by one log after the first course of treatment.
Nevertheless, the blasts remained on the same log level. Therefore, daratumumab treatment
was stopped and replaced by inotuzumab and subsequent SCT, which led to a molecular
CR3. We could not detect binding of a diagnostic anti-CD38 (clone T16, HIT2) in neither
of the two cases early after the start of daratumumab treatment. We then tested whether
the lack of anti-CD38 binding could be caused by steric hindrance with daratumumab
molecules as had been described in myeloma patients. We proved our patients had blasts
CD38 positive on mRNA level and intracellularly (patient 4). We also detected daratumumab
directly on the blasts by detecting its kappa chain on the blasts (patients 4). Moreover,
adding daratumumab to blasts of a patient freshly diagnosed with CD38pos BCP ALL blocked
anti-CD38 antibody (clone T16) binding. Collectively, the data showed that like in
myeloma, daratumumab may block binding of some diagnostic antibodies. We are currently
testing the feasibility of multiepitope CYT-38F2 antibody in children with ALL on
daratumumab treatment.
Summary/Conclusion: In conclusion, daratumumab can lead to CR2 in relapse/refractory
ALL, however, the effect is often temporary. Daratumumab administration may result
in weeks lasting inability of anti-CD38 mAbs to bind to leukemia cells.
Supported by Ministry of Health of the Czech Republic, grants nr. NU20-03-00284 and
NU20J-07-00028.
P377: GENETIC CHARACTERISTICS AND CD7-CAR-T THERAPY OF TRAD::MYC TRANSLOCATION POSITIVE
ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS
T. Wang1,2, J. Ni3, H. Zhang3, S. Hui1, T. Liu3, X. Zhang4, G. Zhang4, X. Ma3, X.
Wang1, Y. Zhang1, Q. Zhang1, H. Liu1,2,*
1Division of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Langfang; 2Beijing
Lu Daopei Institute of Hematology, Beijing; 3Division of Laboratory Medicine, Hebei
Yanda Lu Daopei Hospital, Langfang; 4department of chemotherapy, Hebei Yanda Lu Daopei
Hospital, Langfang, China
Background:
TRAD::MYC is a recurrent but rare translocation in leukemia and lacks relevantreports.
Aims: To investigate the genetic characteristics of acute lymphoblastic leukemia (ALL)
with TRAD::MYC translocation and the clinical outcome of chimeric antigen receptor
T cells (CAR-T) therapy and allogeneic hematopoietic stem cell transplantation (allo-HSCT).
Methods: A retrospective analysis was performed of 5,450 patients with ALL admitted
to our hospital from Apr. 1, 2016, to Dec. 31, 2021. Cases positive for t(8;14)(q24.1;q11.2)
in karyotype analysis were selected, TRAD::MYC translocation and CDKN2A/B gene deletion
were confirmed by fluorescence in situ hybridization (FISH). The TRAD::MYC positive
cases were further screened for a panel of 36 common leukemia fusion genes (FGs),
RNA-seq for comprehensive analysis of FGs, and mutation screening of a panel of 86
leukemia driver genes.
Results: A total of 12 cases were TRAD::MYC positive, 9 males and 3 females, aged
3-28 years (median 15 years), 9 children (<18 years old) and 3 adults. The diagnosis
included 11 T-ALL and 1 B-ALL, and 7 cases (63.6%) manifested high white blood cells
(180-486.18×109/L) at the onset. Nine cases had karyotype results at the initial onset,
of which three were positive for t(8;14)(q24.1;q11.2), and the rest 6 acquired this
translocation at relapse. In the 3 cases with no karyotype results at the initial
onset, t(8;14)(q24.1;q11.2) was detected in relapse specimens. Of the 12 cases, 11
showed additional karyotype abnormalities in t(8;14)(q24.1;q11.2) positive specimens.
FISH confirmed the involvement of TRAD and MYC genes corresponding to t(8;14)(q24.1;q11.2)
in all 12 cases, and CDKN2A/B gene loss was detected in 4 (33%) cases. All cases were
screened for fusion genes, and RNA-seq was performed in five cases. Six cases were
positive for driver FGs, including 3 STIL::TAL1, 1 KMT2A::AF9 in the B-ALL case, 1
CTCF::NFATC3, and 1 ZC3HAV1::ABL2. Six cases (50%) were positive for PTEN mutation
and 3 positive for FBXW7 mutation, other mutated genes including NOTCH1, TP53, CCND3,
etc. Eleven cases were followed up for 6-67 months (median 11 months), and 8 died
during the follow-up period. Eight cases (72.7%) experienced central nervous system
(CNS) leukemia, and 3 had mediastinal invasion. All patients relapsed after chemotherapy,
and the leukemia cells proliferated rapidly. Six cases received CD7-CAR-T therapy,
and all achieved disease remission after CAR-T cell infusion, of which 1 died from
secondary severe lung infection and 2 died from relapse. Four cases underwent allo-HSCT,
including 2 salvage transplantation and 2 bridging transplantation after CD7-CAR-T
regimen. All 3 cases that remained alive received CD7- CAR-T therapy, 2 of which were
bridged to allo-HSCT.
Image:
Summary/Conclusion: Recurrent t(8;14)(q24.1;q11.2)/TRAD::MYC have a low incidence,
mainly in T-ALL cases, but can also be seen in B-ALL. More than half of cases acquired
the translocation at relapse and in some cases accompanied with other driver FGs in
T-ALL such as STIL::TAL1, KMT2A fusions, and ABL family gene fusions. TRAD::MYC translocation
confers aberrant MYC expression and hyperproliferative of tumor cells. Half cases
were also accompanied by PTEN mutation, which further adversely affected the prognosis.
In these cases, the response to conventional chemotherapy is extremely poor and prone
to early relapse, and CNS infiltration is common. CD7-CAR-T regimen bridging to allo-HSCT
might be a promising therapeutic strategy for better survival in this subgroup, and
the long-term outcomes remain to be further investigated.
P378: THE PROGNOSIS FACTORS OF CAR-T THERAPY IN PATIENTS WITH RELAPSED/REFRACTORY
B-ALL
Y. Wang1, X. Hu1,*, D. Zhang1, Q. Gao1, X. Zhai1, H. Wang1, Y. Gao1, Y. Miao1, Y.
Guo1, W. Zhang1, X. Ru1, X. Li2, F. Guan2
1Department of Hematology, Shaanxi Provincial People’s Hospital, Xi’An; 2School of
Medicine, Northwest University, Xi’An, China
Background: CAR-T therapy showed good clinical efficacy on relapsed/refractory B-ALL
(R/R B-ALL) patients. However, some latest studies revealed that most patients tend
to relapse during long-term follow-up after CAR-T infusion. Few studies have examined
the factors associated with the prognosis of R/R B-ALL patients.
Aims: The aim of the study is to investigate the factors associated with the long-term
survival of R/R B-ALL patients after CAR-T therapy.
Methods: Thirty-eight R/R B-ALL patients were included in the study. Patients received
CAR-T cells infusion from May 2015 to August 2018 in Shaanxi Provincial People’s Hospital.
Peripheral blood mononuclear cells were collected, and CD3+ cells were sorted. CAR-T
cells were prepared by lentivirus. These cells were transfused 3 to 5 days after lymphocyte
removal. Bone marrow smears, flow cytometry, and QT-PCR were used to assess the development
of disease between 2 and 4 weeks after infusion. The endpoint of follow-up was the
time of death or Feb 15, 2022. Cox regression models were used to analyse prognosis
factors.
Results: The median age of the 38 patients was 25 years (6-59 years), including 21
males and 17 females. 26.3% (10/38) of patients had leukemic cells higher than 30%
in their bone marrow smears. There were 34.21% (13/38) of patients with Ph(+), 34.21%
(13/38) with extramedullary disease (EMD), 10.52% (4/38) with MLL-AF4 fusion gene
(+), and 5.26% (2/38) had received HSCT prior to CAR-T therapy. Within 4 weeks of
CAR-T cells infusion, 86.84% (33/38) of patients achieved MRD (-). 13 patients were
disease-free by Feb 15, 2022. For all the 38 patients, the median OS and DFS were
19 months and 16.5 months. We found that a higher proportion of leukemic cells in
bone marrow, MRD (+) after CAR-T infusion and the presence of the MLL-AF4 fusion gene
could predict a worse prognosis. We found that sex, Ph (+), and HSCT after CAR-T infusion
is not associated with the long-term survival of patients. Patients who had received
maintenance therapy after CAR-T therapy showed better OS (49 months vs 9 months, P<0.001)
and DFS (49 months vs 6 months, P<0.001) compared to those who received no maintenance
therapy. In addition, we unexpectedly found that patients with EMD showed better OS
(49 months vs 15 months, P=0.019) and DFS (49 months vs 12 months, P=0.045) than those
without EMD.
Summary/Conclusion: R/R B-ALL patients could achieve long-term survival with CAR-T
cell therapy. The proportion of leukemic cells in bone marrow, MRD(+), and the presence
of the MLL-AF4 fusion gene could predict a worse prognosis of R/R B-ALL patients.
We found that maintenance treatment after CAR-T therapy could significantly improve
patients’ long-term survival and disease-free time, while HSCT showed no significant
benefit to long-term prognosis.
P379: MUTATIONAL LANDSCAPE IN THERAPY-RELATED ACUTE LYMPHOBLASTIC LEUKEMIA
K. D. Hofer1,*, E. Haralambieva2, C. Fritz2, M. Roncador1, C. Ruetsche3, M. Buehler2,
J. Tchinda4, U. Schanz1, C. C. Widmer1,5
1Department of Medical Oncology and Hematology; 2Institute of Pathology; 3Hematology,
University Hospital Zurich; 4Oncology Laboratory, University Children’s Hospital Zurich,
Zurich; 5Hematology, University Hospital Basel, Basel, Switzerland
Background: There is increasing evidence that therapy-related acute lymphoblastic
leukemia (t-ALL) with a reported incidence of 3% to 9% is a distinct entity, which
is associated with inferior survival compared with de novo ALL (dn-ALL). It appears
that this results from a poor cytogenetic predisposition, but data are still very
limited due to the rarity of the disease and the mutational landscape has not been
adequately explored, as molecular analyses are sparse in this unique patient cohort.
Aims: This study attempted to investigate the molecular landscape of patients with
an initial diagnosis of ALL and a prior history of malignancy with consecutive genotoxic
therapy and to evaluate the cytogenomic results and survival.
Methods: Data were collected from 131 adult patients with an initial diagnosis of
ALL treated at the Department of Medical Oncology and Hematology, University Hospital
Zurich, Switzerland. Patients with prior treatment of another malignancy with alkylating
agents, topoisomerase inhibitors, radioactive iodine ablation (for thyroid cancer)
and/or irradiation were classified as t-ALL.
Results: Within our ALL cohort, 14% of the patients met the criteria for t-ALL. The
median latency between genotoxic therapy and diagnosis of t-ALL was 6.5 years, with
breast cancer being the most common neoplasm. KTM2A rearrangement was not significantly
more common in t-ALL than in dn-ALL (15.7% vs 11%, p = 0.15), a finding which was
not as expected from the literature. In therapy-related myeloid neoplasms large chromosomal
deletions, such as del(5q) and del(7q) are known to be associated with alkylating
agents and topoisomerase II inhibitors. In 12% of our t-ALL patients deletions in
chromosome 7 were found, but none in chromosome 5. On the other hand, a high proportion
of Philadelphia chromosome (Ph) 36.8% was present in the t-ALL group. On a molecular
level, the most frequently observed mutation was KMT2D, followed by myeloid neoplasm-associated
mutations of CDKN2A, KRAS and DNTM3A, but no TP53 mutation was found. KRAS mutation
was never present in combination with Ph+, but was associated with KMT2A rearrangement.
Outcome was particularly poor in Ph+ t-ALL patients compared to Ph+ dn-ALL, an effect
that was mitigated by allogeneic stem cell transplantation.
Image:
Summary/Conclusion: In addition to KMT2A rearrangement as a high-risk genomic subtype,
a high proportion of Ph+ was observed in the t-ALL group. Ph+ t-ALL showed a different
mutational landscape than KTM2A-mutated patients, suggesting that the development
of t-ALL is driven by multiple pathways with particularly bad outcome for patients
with Ph+. Although there is overlap in the molecular profile of t-ALL and myeloid
neoplasms, no TP53 mutation was found in our t-ALL cohort. The heterogeneity of the
genetic aberrations in ALL render molecular genome sequencing of somatic variants
an important step to advance our understanding of the disease, especially in those
patients without KTM2A rearrangement but Ph positivity. Our findings help further
define therapy-associated ALL as a distinct entity, but also highlight the need for
a better molecular understanding of its specific disease evolution.
P380: EX VIVO IMMUNE ACTIVATION WITH THE MACROPHAGE-TARGETING IMMUNOTHERAPY, ANTI-CLEVER-1
ANTIBODY BEXMARILIMAB, IN ACUTE MYELOID LEUKEMIA AND MYELODYSPLASTIC SYNDROME
S. Aakko1,2,*, A. Ylitalo3, H. Kuusanmäki2, M. Björkman1, J. Mandelin1, J. Jalkanen1,
M.-L. Fjällskog1, C. Heckman2, M. Hollmén3, M. Kontro2,4
1Faron Pharmaceuticals Ltd, Turku; 2Institute for Molecular Medicine Finland, HiLIFE,
University of Helsinki, Helsinki; 3MediCity Research Laboratory, University of Turku,
Turku; 4Department of Hematology, Helsinki University Hospital Comprehensive Cancer
Center, Helsinki, Finland
Background: Despite the recent approvals of targeted therapeutic agents for acute
myeloid leukemia (AML), the treatment options remain few and for myelodysplastic syndrome
(MDS), even fewer. The potential of immunotherapies in these myeloid malignancies
remains under investigation. To this end, bexmarilimab, an anti-Clever-1 antibody,
aims to harness the therapeutic potential of macrophages. Clever-1 (STAB1) is a scavenger
receptor, expressed on the surface of a subpopulation of immunosuppressive macrophages.
By inhibiting Clever-1, bexmarilimab demonstrates potential to turn the anti-inflammatory
macrophages to pro-inflammatory and activate CD8+ T cells (Virtakoivu R. et al. 2021.
Clin. Cancer Res) along with promising anti-tumor activity against solid tumors in
patients with multiple lines of previous treatment (Bono P. et al. 2019. Ann. Oncol.).
As Clever-1 is expressed in myeloid cells and high STAB1 levels associate with poor
survival in AML (Lin S.Y. et al. 2019. Mol. Ther. Nucleic Acids), bexmarilimab may
possess therapeutic potential in AML and MDS.
Aims: The aim of this study was to profile Clever-1 expression in AML and MDS and
to test in preclinical models bexmarilimab’s growth inhibitory and immunomodulatory
potential, as a single agent and in combination with azaciditine and venetoclax.
Methods: AML cell lines (n=11) and frozen mononuclear cells, extracted from AML (n=27)
and very high risk MDS (n=4) patient bone marrow (BM) aspirates provided by the Finnish
Hematology Registry and Biobank, were used in this study. Samples were treated for
48h with bexmarilimab alone, or in combination with azacytidine and/or venetoclax.
Flow cytometry was used to detect different myeloid and TBNK cell populations along
with Clever-1, HLA-DR, PD-(L)1 and Granzyme B protein expression.
Results: Our results confirm Clever-1 protein expression in AML cell lines with high
STAB1 level and most importantly, in AML and MDS patient-derived BM monocytes and
blasts. AML samples from FAB M4/M5 subtypes exhibited highest Clever-1 levels, along
with a FAB M2 sample collected prior to allogeneic stem cell transplantation and consequent
rapid relapse. Clever-1 expression correlated negatively with monocyte MHC class II
molecule, HLA-DR, expression, and BM T cell frequency, in line with the immunosuppressed
state associated with high Clever-1.
Ex vivo treatment of the primary AML cells with bexmarilimab resulted in a notable,
5-10x fold increase of monocyte HLA-DR in samples with low basal HLA-DR and high Clever-1.
The combination of azacitidine with bexmarilimab augmented HLA-DR induction by 20-110%
(mean 44%). On the other hand, combination with bexmarilimab overcame the HLA-DR suppressing
effect of venetoclax.
The BM TBNK cell populations and thus bexmarilimab’s effect on the effector cells,
showed great inter-sample variability. FAB M1/M2 AML showed increase of activation
markers, such as Ki67, CXCR3 and Granzyme B after ex vivo bexmarilimab treatment in
CD8+ T cells. Furthermore, bexmarilimab reduced PD-1 expression in NK- and CD8+CXCR3+
T cell populations of FAB M0-M2 AML and MDS-EB2.
Summary/Conclusion: Our results confirm that Clever-1 is expressed in AML and MDS
patient BM blasts and monocytes. Ex vivo treatment with bexmarilimab, alone or in
combination with azacytidine or venetoclax, demonstrated increased HLA-DR expression,
indicating enhanced antigen presentation capability. Furthermore, NK and CD8+ T cells
showed decreased PD-1 and increase of activation markers. These results confirm the
therapeutic potential of bexmarilimab in myeloid malignancies and await further validation
in clinical trials.
P381: DNA METHYLATION PROFILING OF MESENCHYMAL STROMAL CELLS ISOLATED FROM FEMURAL
HEAD BONE MARROW VERSUS BONE MARROW ASPIRATES: RELEVANCE FOR AML STUDY BASED CONTROLS
A. Abdul-Aziz1,*, C. Weigel2, A. Kovacs2, Y.-Z. Wu2, J. Byrd1, E. Hertlein1, C. Oakes2
1Department of Internal Medicine, University of Cincinnati, Cincinnati; 2Department
of Internal Medicine, The Ohio State University, Columbus, United States of America
Background: Bone marrow mesenchymal stromal cells (BMSCs) are important precursors
for multiple differentiated cell types. Alterations in BMSCs contribute to several
disease states, including acute myeloid leukemia (AML) and other hematologic cancers.
As BMSCs are used as controls for research and obtained from various bone tissue locations
with active hematopoiesis, it is important to understand the developmental capacity
and potential differential phenotypes of BMSCs derived from various anatomical sites.
Epigenetic regulation plays an essential role in cell lineage specification and development;
carefully controlled transcription factor (TF) activity and other chromatin-associated
proteins modify the genomic landscape to generate stable, cell type-specific global
patterns of gene expression.
Aims: We examined genome-wide DNA methylation patterns in BMSCs derived from either
femoral head bone marrow or bone marrow aspirates of the iliac crest of healthy donors
to identify the optimal control cell population for hematological malignancy research.
Methods: Hip-derived BMSCs were obtained from femur heads obtained during hip replacement
surgeries (n=19). All patients provided informed consent in accordance with an IRB-approved
protocol and the Declaration of Helsinki. Aspirate-derived BMSCs (n=7) were expanded
from bone marrow mononuclear cells obtained from a commercial source (AllCells). For
genome-wide DNA methylation analyses, the EPIC/850K array platform was used following
manufacturer’s protocols (Illumina). Analyses included data obtained from the Gene
Expression Omnibus (GEO, GSE79695 and GSE124390) generated from 450K (n= 12) and 850K
array data (n=10), respectively. Overlapping probes between the arrays that passed
QC were used for downstream analysis. Sex chromosomes and non-CpG probes were removed.
The data was reduced to 5,000 most-variable CpGs followed by clustering analysis as
described in results.
Results: Unsupervised clustering using principal component analysis (PCA) and hierarchical
clustering revealed two groups that segregated perfectly between hip-derived versus
aspirate-derived BMSCs. There were no observable differences in aspirate-BMSCs generated
by our group and those obtained from GEO. Two group supervised analysis (hip versus
aspirate) revealed 4,568 CpGs (probes) hypomethylated in hip-derived samples (q<0.05,
difference>20%) and 1,433 probes hypermethylated in hip-derived samples (q<0.05, difference>20%).
DNA sequence motif enrichment analysis using the HOMER software revealed that hypomethylation
in hip-derived BMSCs involved activation of CRE, HOX and AP-1 family TFs. We also
observed parallel hypomethylation of the promoters of CREB5, genes from the HOXB and
HOXC clusters, and several genes of the MAPK pathway. Hypermethylation in hip-derived
BMSCs conversely involved RUNX motifs, corresponding with hypermethylation of the
RUNX3 gene. We also observed robust DNA methylation changes at the promoters of other
lineage-specifying genes including TBX3, TBX5, PITX2, NR2F2, and EN1. Pathway analysis
of genes demonstrating promoter hypomethylation in hip-derived samples revealed strong
enrichment for adipocyte progenitors and development.
Summary/Conclusion: These findings demonstrate that hip-derived BMSCs are phenotypically
distinct from aspirate-derived BMSCs, which may affect their potential use in both
therapeutic and research settings where stem-like non-primed BMSCs are required. Our
study highlights the importance of using proper selection of control cells to study
the role of BMSCs in hematologic malignancies.
P382: PTK2B MEDIATES TYROSINE KINASE INHIBITOR DRUG RESISTANCE IN AML ASSOCIATED WITH
ALTERED MIGRATION AND ADHESION PROPERTIES
C. Allert1,2,*, S. Zimmermann2,3, S. Göllner1, D. Heid1,4,5, M. Bruckmann1, M. Janssen1,
B. Besenbeck1, J. Krijgsveld2,3, C. Müller-Tidow1,2,4, M. F. Blank1,3,4
1Medical Department V Hematology, Oncology and Rheumatology, Heidelberg University
Hospital; 2University of Heidelberg Medical Faculty; 3Division Proteomics of Stem
Cells and Cancer, German Cancer Research Center (DKFZ); 4Molecular Medicine Partnership
Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL);
5European Molecular Biology Laboratory EMBL, Heidelberg, Germany
Background: Tyrosine kinase inhibitor (TKI) therapy is a well-established therapy
approach in FLT3-mutated AML. However, TKI resistance occurs frequently while the
underlying mechanisms remain incompletely understood. One concept of chemotherapy-resistance
in leukemia is cell adhesion mediated drug resistance (CAM-DR).
Aims: In this study we aimed to investigate the role of CAM-DR at an early stage of
therapy resistance in FLT3-ITD mutated AML.
Methods: We characterized FLT3-ITD mutated AML cell lines upon shorter and longer
TKI exposure. Emerging TKI resistance was assessed by transcriptome and total proteome
analyses, as well as nascent proteomics and MS-based protein stability/degradation
assays. Further, we investigated the impact of emerging TKI-resistance on cell adhesion
and migration. After identification of a highly expressed protein in TKI resistant
cells with implications in cell adhesion, i.e. PTK2B, we investigated the effects
of available inhibitors of this protein on AML cell lines in the presence or absence
of other TKIs or chemotherapeutic agents and confirmed the effects in primary AML
patient samples (n=6).
Results: We discovered that protein tyrosine kinase 2b (PTK2B) was induced during
early TKI resistance development. Interestingly, PTK2B, a member of the focal adhesion
kinase superfamily, was upregulated at the transcriptional and translational level.
PTK2B was also post-translationally stabilized in TKI resistant cells. Of note, PTK2B
was highly expressed in leukemic stem cells (LSCs) from FLT3-ITD mutated patients.
The upregulation of PTK2B was accompanied by increased migration of TKI-resistant
cells with concomitant loss of cell adhesion, suggesting altered niche interactions.
Notably, treatment with a PTK2B inhibitor, such as PF-431396, reverted the resistance-associated
phenotypes. Enhanced migration was abolished upon PF-431396 treatment. Significantly
altered pathways in resistant cells, assessed by nascent proteomics, were largely
reverted upon treatment of resistant cells with a PTK2B inhibitor.
The FAK/PTK2B inhibitors PF-431396 and Defactinib synergized with several TKIs, such
as midostaurin or gilteritinib, and commonly used chemotherapeutic agents, such as
daunorubicin, in FLT3-ITD mutated AML cells. Synergism was restricted to FLT3 mutated
cells. This synergism was significantly more pronounced in TKI-resistant cells and
the addition of a PTK2B inhibitor could overcome TKI resistance to various extent.
Also in primary AML patient samples, Defactinib showed lower IC50s specifically for
FLT3-ITD mutated patients.
Summary/Conclusion: These data establish that FLT3-ITD AML resistance towards TKI
depends on PTK2B which alters leukemia cell adhesion and migration. PTK2B inhibitors
might be synergistic with TKI in FLT3-ITD AML therapy. PTK2B inhibition could thus
prevent the outgrow of drug-resistant clones at an early stage and may improve the
outcome of FLT3-ITD mutated AML.
P383: USING CF-DNA AS STARTING MATERIAL FOR MRD STUDIES BY NGS IMPROVES THE SENSITIVITY
OF THE METHOD AGAINST CTCS.
N. Álvarez Sánchez-Redondo1,*, S. Dorado2, A. Martín3, L. Rufián1, V. Garrido1, A.
Giménez1, L. Moreno1, Y. Heredia4, J. Martínez-López5, S. Barrio6, R. Ayala7
11. Department of Translational Hematology, Research Institute Hospital 12 de Octubre
(i+12), Hematological Malignancies Clinical Research Unit H120-CNIO, CIBERONC; 23.Computer
Science and 4.Altum Sequencing Co., Madrid, Spain. Engineering Department. Carlos
III University, Madrid and; 34.Altum Sequencing Co., Madrid, Spain.; 44. Altum Sequencing
Co., Madrid, Spain.; 51.Department of Translational Hematology, Research Institute
Hospital 12 de Octubre (i+12), Hematological Malignancies Clinical Research Unit H120-CNIO,
CIBERONC, and 2.Complutense University of Madrid; 61.Department of Translational Hematology,
Research Institute Hospital 12 de Octubre (i+12), Hematological Malignancies Clinical
Research Unit H120-CNIO, CIBERONC, and 4.Altum Sequencing Co., Madrid, Spain.; 71.Department
of Translational Hematology, Research Institute Hospital 12 de Octubre (i+12), Hematological
Malignancies Clinical Research Unit H120-CNIO, CIBERONC, and 2.Complutense University
of Madrid., Madrid, Spain
Background: The quantification of Minimal Residual Disease (MRD) has become a relevant
marker as it can detect acute myeloid leukemia (AML) patients with a high risk of
relapse. Although the reference MRD method in AML is to quantify tumor cells present
in bone marrow (BM), the study of circulating tumor cells (CTC) in peripheral blood
(PB) is also being incorporated into clinical protocols. However, cfDNA as an MRD
biomarker in AML has not yet been validated.
Keywords: AML, MRD, Liquid Biopsy, NGS.
Aims: The purpose of the study is to evaluate the use of cfDNA by NGS test as a new
method for MRD quantification in AML and compare it application with the use of PB
CTCs in the follow-up of the disease.
Methods: Three AML patients treated at the Hospital Universitario 12 de Octubre and
with cfDNA available have been included. The mutational profile was defined by NGS
(Ion Torrent System) at diagnosis using a panel of 42 genes involved in myeloid pathologies.
Then, somatic mutations were selected, and MRD quantification was performed in liquid
biopsy combining whole blood cells (WBC) and cfDNA. Both fractions were informatically
differentiated by including molecular tags in the first PCR amplification. A total
of 18 follow-up samples were analyzed with a threshold for EMR positivity set at 1x10^
(-4) as we previously described (Onecha E et al. Haematologica 2019). An average of
66ng of cfDNA and 660ng of gDNA from WBC were used. The results were compared with
those obtained by multiparametric flow cytometry (MFC) in BM.
Results: In the first patient, four variants affecting the genes CBL (VAF =7.4%),
MPL (50%), JAK2 (51.5%) and ETV6 (10,6%) were detected at diagnosis in BM. The Leukemia-associated
variant affecting ETV6 was selected as an MRD biomarker and detected in cfDNA (0.86%)
post-consolidation 1. On this time-point (TP), the result of MRD by MFC on BM was
also positive (0.002%), being negative in CTC-PB by NGS. The Non-leukemia-associated
variants (JAK2, MPL, CBL) were also followed-up. Of interest, those alterations showed
constant VAF levels in pre-transplant remission. After transplant, the VAF of JAK2
and MPL correlated with the donor chimerism studied by conventional methods after
transplantation. On the other hand, CBL VAF was below the threshold sensitivity limit
(0.01%) during all follow-ups. This result that CBL was also associated with leukemia,
likely affecting a treatment-sensitive subclone.
In patient 2, variants affecting IDH2 (92%), RUNX1 (43%), and DNMT3A (49%) genes were
detected by at diagnosis. IDH2 mutation was selected as MRD biomarker, showed a VAF
of 9.4% at relapse, correlating with the blast count of 10% defined by the morphology
study of BM. Of note, in this TP, MRD-BM by MFC was 0.011%.
In patient 3, the SF3B1 (43%) and EPAS1 (49%) mutations were detected by NGS in BM
at diagnosis. The leukemia-associated variant (SF3B1) was observed in the cfDNA sample
(0,0003) at the first follow-up sample but was negative at the following TPs. MRD
by CTCs and MFC-BM were also negative. This patient has not relapsed to date (follow-up
time six months after diagnosis).
Image:
Summary/Conclusion: MRD quantification based on the use of cfDNA by NGS offers promising
results and, in the future, could be a good option for disease monitoring or early
detection of relapse. The results of the whole series will be presented at the meeting.
P384: THE ROLE OF THE MANNOSE RECEPTOR C-TYPE 2 (MRC2) IN LEUKEMIC STEM CELL MAINTENANCE
IN AML
M. Arnone1,*, E. Görsch1, S. Pöschel1, P. Godavarthy1, A. Stanger1, S. Rudat1, C.
Lengerke1
1Innere Medizin 2, University Hospital Tuebingen, Tuebingen, Germany
Background: Therapy-resistant leukemia-initiating cells, so called leukemic stem cells
(LSC), are responsible for disease relapse and a major cause of death in patients
with acute myeloid leukemia (AML). Effective identification and targeting of LSCs
are thus important goals. Our previous RNA sequencing analyses revealed that the MRC2
gene, which encodes a transmembrane receptor known for its ability to bind and degrade
collagen as well as for its pro-migratory and -invasion functions, is enriched in
LSCs.
Aims: We aim to gain a better understanding of the function(s) of MRC2 in AML in general
and LSCs in particular.
Methods: We used flow cytometry to characterize MRC2 expression in primary AML samples
and healthy cells from the hematopoietic system. Imaging flow cytometry experiments
were performed to study the role of MRC2 in gelatin uptake.
Results: We found that mRNA expression of the MRC2 gene was increased in the LSC compartment
of 5 AML patients compared to their non-LSC counterparts. Analyzing MRC2 cell surface
presentation by flow cytometry, we found that AML cell lines (n=10) displayed the
highest levels of MRC2 expression followed by healthy CD34+ cord blood (CB) cells
(n=7), while primary AML patient samples (n=21) were characterized by an intermediary
MRC2 cell surface presentation with the lowest levels of MRC2 presentation in differentiated
healthy hematopoietic cells. Interestingly, high MRC2 expression correlated with increased
clonogenic activity as only sorted MRC2+ but not MRC2- primary AML cells from the
same sample were able to form colonies in a methylcellulose assay; and siRNA-mediated
knockdown of MRC2 in the bulk of primary AML cells led to a reduction in colony formation
when compared to control siRNA transfected cells. Furthermore, using CRISPR/Cas9 we
generated a MRC2 knock-out AML cell line and observed in in vitro assays that MRC2
expression is required for migration, invasion and adhesion to collagen and laminin
421. Finally, we could show by imaging flow cytometry (ImageStream) that AML cells
lacking MRC2 expression were unable to internalize fluorescently-labelled gelatin.
Summary/Conclusion: Together, our results suggest that AML cells benefit from MRC2
expression due to its multiple functions involving migration, invasion and the interaction
with extracellular matrix components. Additionally, MRC2 has a potential role in LSC
biology such as the maintenance of stemness-associated properties. Further in vivo
studies will improve our understanding of MRC2, which could then inform us on novel
therapeutic approaches.
P385: DETECTION OF KMT2A PARTIAL TANDEM DUPLICATIONS IN ACUTE MYELOID LEUKEMIA PATIENTS
BY NGS
A. Artigas-Baleri1,*, G. Oñate2, A. Brell2, J. Esteve3, S. Vives4, M. Tormo5, M. Arnan6,
A. Garcia7, R. Coll8, A. Sampol9, J. Bargay10, F. Vall-llovera11, O. Salamero12, J.
Nomdedéu2, M. Pratcorona2
1Hematology, Insitut de Recerca de Hospital de la Santa Creu i Sant Pau; 2Hematology,
Hospital de la Santa Creu i Sant Pau; 3Hematology, Hospital Clínic, Barcelona; 4Hematology,
Institut Català d’Oncologia, Hospital Germans Trias i Pujol, José Carreras Leukemia
Research Institute, Badalona; 5Hematology, Hospital Clínico Universitario, Biomedical
Research Institute INCLIVA, Valencia; 6Hematology, Insitut Català d’Oncologia, Hospital
Duran i Reynals, Institut Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat;
7Hematology, Hospital Universitari Arnau de Vilanova, Lleida; 8Hematology, Institut
Català d’Oncologia, Hospital Josep Trueta, Girona; 9Hematology, Hospital Son Espases;
10Hematology, Hospital Son Llàtzer, Palma de Mallorca; 11Hematology, Mutua Terrassa,
Terrassa; 12Hematology, Hospital Vall d’Hebron, Barcelona, Spain
Background: Acute myeloid leukemia (AML) is a heterogeneous hematologic disease consisting
of an aberrant proliferation and differentiation of hematopoietic stem cells in the
bone marrow. In the last years the identification of cytogenetic and molecular biomarkers
has improved the AML classification considering prognosis, and allowing a better risk-adapted
treatment strategy. Regarding molecular biomarkers, NPM1, RUNX1 and ASXL1 mutations,
FLT3-ITD (internal tandem duplications), CEBPAbi or KMT2A-PTD (partial tandem duplications)
have been shown to have clinically significant prognostic value and they define specific
AML disease subtype. Our study focuses on the detection of PTD in KMT2A which is involved
in the regulation of gene expression during hematopoiesis. KMT2A-PTDs are found in
5-10% of normal karyotype patients and have been associated with poorer disease-free
and overall survival rates. Mainly, PTDs in this gene span exons 2 to 8-10 and their
detection is based on long-distance genomic PCR with extracted DNA from bone marrow.
This is a time-consuming technique, and that is probably the main reason to explain
why this marker is not widely performed at the time of diagnosis.
Aims: We aimed to analyze the potentiality of Next Generation Sequencing (NGS) in
the identification of KMT2A-PTD, a technique that is being implemented in many laboratories
and could be a good strategy to detect this marker.
Methods: This study included patients from CETLAM group (Grupo Cooperativo de Estudio
y Tratamiento de las Leucemias Agudas y Mielodisplásicas) with a diagnosis of AML
in 2021 (from January to October) and age≤70. The study of KMT2A-PTDs was carried
out with DNA samples through two different techniques: PCR and NGS. DNA was extracted
from bone marrow with the AllPrep DNA/RNA Mini Kit (Qiagen®). For the PCR amplification
we used both custom 8-exon forward primer and 2-exon reverse primer. NGS was performed
by hybridization capture technology (Nonacus®) using a custom panel which includes
43 genes. MiSeq (Illumina®) system was used for sequencing and Datagenomics for analysing
the results. Hybridization capture allows the comparison of the number of reads per
position between samples to identify structural variants as PTDs. This approach permits
to know exactly which exons have been duplicated.
Results: We included a total of 72 AML patients who were genotyped for KMT2A-PTD with
both PCR and Hybridization capture NGS. We identified 4 and 7 patients with a partial
tandem duplication in KMT2A gene by PCR and NGS, respectively. The four KMT2A-PTDs
identified with PCR were also detected by NGS. In contrast, three PTDs were observed
only with NGS. In two of them the PTD spanned exons 2 to 8 (Figure 1), while in the
other it spanned exons 4 to 8. The first two PTDs were confirmed with the classic
long-distance genomic PCR but performed with cDNA, skipping the presence of a large
non-coding DNA in the rearrangement which could not be amplified by conventional PCR.
To confirm the third PTD a new reverse primer in exon 4 was designed. All three NGS
observed KMT2A-PTDs could be verified.
Image:
Summary/Conclusion: Our results show that Hybridization capture NGS is a good technique
to detect KMT2A-PTDs, with a better sensibility than PCR. NGS is a cost-effective
method as it is becoming an essential tool for AML classification at diagnosis.
P386: CELL FATE DETERMINING TRANSCRIPTION FACTORS AS VULNERABILITIES IN CHILDHOOD
AND ADULT MLL-REARRANGED AND T(8;16) ACUTE MYELOID LEUKEMIA
S. Arza Apalategi1,*, B. Heuts2, S. Bergevoet1, S. van Heeringen3, J. Jansen1, J.
Martens2, B. van der Reijden1
1Laboratory Medicine, Laboratory of hematology, Radboudumc; 2Molecular Biology; 3Molecular
Developmental Biology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
Background: The MLL-AF9 protein contributes to acute myeloid leukemia (AML) development
through transcriptional activation of oncogenic gene programs. Remarkably, MLL-AF9
AML consists of two subgroups, one with high oncogenic MECOM expression and dismal
outcome; the other with a unique gene expression profile lacking MECOM expression
and a favourable outcome. How the differences between these two clinically and biologically
different subgroups are brought about in the context of the same MLL-AF9 genetic background
is currently unknown.
Aims: To identify vulnerabilities in AML beyond the MLL-AF9 oncofusion gene we determined
cell fate determining transcription factors pertinent to the two clinically and biologically
different subgroups.
Methods: Transcription factors key to cell fate were determined by first analysing
differential transcriptional start sites of genes and gene enhancers by genome-wide
“Cap Analysis Gene Expression (CAGE)”-sequencing of three primary MECOM positive and
negative MLL-AF9 AML samples. Sequencing data were subsequently analysed using a new
bioinformatic tool that builds gene regulatory networks based on differential gene
expression and enhancer activities to predict key transcription factors in cell fate
determination. For predicted factors AML cohort-wide gene expression patterns, transforming
potential and downstream gene programs were determined.
Results: Bioinformatic analyses of CAGE-seq data identified the transcription factor
MECOM as top hit for cell fate determination in MECOM+ MLL-AF9, confirming the power
of this approach. In the group lacking MECOM expression we identified HMX3 as most
influential transcription factor. HMX3 is essential for neuronal development and not
expressed in any healthy blood cell type. In a large adult and childhood AML cohort
(totalling 630 cases), high HMX3 expression was observed in 4% and 11% cases, respectively.
HMX3 positive cases clustered tightly together in unsupervised clustering analyses,
and 80% of cases were positive for MLL-rearranged (MLL-AF9, AF10, ENL) and t(8;16)
AML. None were positive for other known recurrent AML mutations except for RAS mutations.
In line with these observations, MLL-AF9 positive human cell lines expressed HMX3
(eg. THP1 and MOLM13). HMX3 silencing in THP1 and MOLM13 cells caused pronounced cell
cycle arrest and apoptosis evidenced by PI, Annexin and 7AAD staining in flow cytometry.
Forced lentiviral HMX3 expression in primary human GCSF-mobilized CD34+ cells resulted
in an over 70% clonogenic growth inhibition compared to control transduced cells.
To identify relevant gene programs we silenced HMX3 in MOLM13 cells followed by RNA-sequencing.
Gene ontology analyses of over 3000 differentially expressed genes (p<0.01) suggested
a role for HMX3 in cell cycle progression, sister chromatid separation, and immune
response suppression (p<0.05).
Summary/Conclusion: Bioinformatic analyses of CAGE-sequencing of primary leukemia
samples predicted the transcription factors MECOM and HMX3 key to poor and good outcome
MLL-AF9 AML, respectively. High HMX3 expression associated with a unique gene expression
profile and associated strongly with MLL-rearranged and t(8;16) AML. HMX3 only was
unable to transform healthy human immature blood cells but appeared essential for
survival of MLL-AF9 leukemia cell lines, potentially by positively regulating cell
cycle progression and immune evasion. Insight into the factors that activate HMX3
expression in AML could provide potential targets for therapies in MLL-rearranged
and t(8;16) AML.
P387: UNRAVELING LEUKEMIC STEM CELLS MITOCHONDRIAL DEPENDENCY IN PEDIATRIC ACUTE MYELOID
LEUKEMIA
M. Benetton1,*, A. Da Ros1, G. Borella1, G. Longo1, C. Tregnago1, F. Locatelli2, M.
Pigazzi1
1Department of Women’s and Children’s Health, Haematology-Oncology Clinic and Laboratory,
University of Padova, Padova; 2Department of Pediatric Hematology and Oncology, IRCCS
Ospedale Pediatrico Bambino Gesù and Sapienza University of Rome, Roma, Italy
Background: Relapse still represents an unsolved problem for children with acute myeloid
leukemia (AML), therefore more effective therapeutic strategies are needed for a complete
blasts eradication. Leukemic stem cells (LSCs) have been proposed as the therapy-resistant
reservoir of cells responsible for failure of antiproliferative chemotherapy and disease
recurrence in AML due to their self-renewal capacity and quiescence allowing evasion
from chemotherapy. Thus, the identification of LSCs-specific vulnerabilities is needed
for more efficacious treatments.
Aims: We aim to dissect novel LSCs characteristics to unravel mechanisms of chemoresistance
and disease recurrence, and characterize biological properties of LSCs that can be
targeted to completely eradicate AML.
Methods: To study LSCs derived from primary pediatric AML samples, we based on reactive
oxygen species (ROS) content since it was shown that functionally defined LSCs have
relatively low levels of ROS. We isolated the lowest 20% of cells fluorescing with
CellROX probe (ROS low) and the highest 20% (ROS high) by cell sorting, and analyzed
cell cycle, engraftment in mice and RNA sequencing to identify deregulated pathways.
Results: After isolating ROS low and ROS high cells, we first assessed classical stemness
features. By colony-forming assay, we observed that ROS low and ROS high cells have
similar colony-forming capacity, but only ROS low cells preserve this capacity after
a second replating whereas the ROS high population does not (p=0.002). By BrdU assay,
we found that ROS low cells are relatively dormant, being more in G0/G1 cell cycle
phase (41.9%) than the ROS high counterpart (7.9%, p=0.001), also confirmed by CFSE
staining, revealing that ROS low cells proliferate more slowly (at day 4, 31% undivided
cells in ROS low vs 8% in ROS high). According to our previously published data, we
confirmed that ROS low cells express higher levels of CDK6-AS1 (RQ=1.38) and CDK6
genes (RQ=2.69; RQ=1 for ROS high) and that, being more quiescent, are characterized
by lower levels of mitochondrial membrane potential by TMRE and lower content of mitochondrial
ROS, byproducts of mitochondrial activity, by MitoSOX staining. We also verified that
ROS low cells have a lower mitochondrial mass by MitoTracker and TOMM20 staining (mitochondria
to nucleus ratio 1.18 vs 1.35 for ROS high, p=0.0005), suggesting that mitochondrial
mass and functions might be involved in quiescent cells properties. However, engraftment
experiments did not reveal different capacity of ROS low or ROS high cells in recreating
leukemia in NSG mice, neither in terms of human CD45+ cells in mice peripheral blood
(23.3% vs 41.9% respectively, p=0.12) nor bone marrow infiltration (73% vs 84.8%,
p=0.44) at 8 weeks post-engraftment, opening for further dissection of the presence
of LSCs within ROS high cells. By RNA sequencing, we found that the top 150 differentially
expressed genes between ROS low and ROS high cells are involved in focal adhesion
(logP=-13.51), regulation of hemopoiesis (logP=-12.31), cell response to stress (logP=-8.73)
and cytokine production (logP=-8.67). Further analyses will confirm the role of these
pathways in maintaining ROS low LSCs. Moreover, we identified 5 differentially expressed
surface antigens that are under investigation as putative ROS low LSCs markers.
Summary/Conclusion: In conclusion, the ROS low strategy permits to identify a quiescent
AML subpopulation with peculiar mitochondrial features and transcriptome allowing
to pinpoint novel molecular pathways that might be targeted to enhance LSCs clearance.
P388: PREFERENTIAL SYNERGISTIC EFFECT OF INDISULAM IN COMBINATION WITH VENETOCLAX
AND AZACYTIDINE IN AML CELLS WITH SF3B1 MUTATIONS.
P. Bernardo1,2,*, M. Pereira1, B. Galvão1, J. Desterro1,3, A. Nunes3, M. D. J. Frade3,
J. Lobato3, M. G. da Silva3, M. Lemos4, M. Silva4, P. Ribeiro4, S. Matos5, N. Custódio1,
M. Carmo-Fonseca1
1Instituto Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade
de Lisboa; 2Serviço de Hematologia Clínica, Hospital da Luz Lisboa; 3Serviço de Hematologia
Clínica, Instituto Português de Oncologia de Lisboa Francisco Gentil; 4Serviço de
Hematologia Clínica, Centro Hospitalar Lisboa Central - Hospital de St. António dos
Capuchos; 5GenoMed - Diagnósticos de Medicina Molecular SA, Lisbon, Portugal
Background: Acute myeloid leukemia (AML) is an aggressive hematological malignancy
with poor prognosis characterized by a high rate of relapse after conventional combination
chemotherapy, highlighting the need for improved therapeutic interventions. Genes
involved in pre-mRNA splicing, namely SF3B1, U2AF1 and SRSF2, are frequently mutated
in AML.Recent studies suggest that pharmacological targeting of the splicing machinery
results in preferential lethality of splicing mutant AML cells, potentially providing
a novel strategy for treatment of this disease. Existing anti- cancer sulfonamides
have been shown to interfere with splicing and induce preferential killing of splicing
mutant AML cell lines. A particular sulfonamide drug, indisulam, was previously studied
in cancer clinical trials where it was found to be safe but demonstrated limited efficacy,
most likely because neither the mechanism of action nor potential biomarkers of response
were known.
Aims: We investigated, in this study, the efficacy of indisulam, either alone or in
combination with ABT-199 (venetoclax) and azacitidine, in cultured primary human AML
cells.
Methods: Bone marrow cells were collected from adults newly diagnosed with AML, non-treated.
Cells were cultured and incubated for 48 and 72 hours with indisulam at a range of
nanomolar concentrations. Additionally, cells were incubated with ABT-199 (10nM) and
azacitidine (1μM), alone or in combination with indisulam. Cell viability and cell
death were measured. For mutation profiling, a targeted myeloid gene panel was used.
Results: We found that treatment with indisulam resulted in lethality of primary AML
cells harboring mutations in SF3B1, but had minor or no effect on AML cells with other
mutations including mutations in splicing factors U2AF1 and SRSF2. In SF3B1-mutated
cells, a clear synergistic effect was further observed when indisulam was combined
with ABT-199 and azacitidine. The correlation between efficacy of indisulam and synergy
with ABT-199 and azacitidine in AML cells with different mutations will be shown and
discussed.
Summary/Conclusion: Our results indicate that indisulam is preferentially active and
synergistic with venetoclax and azacitidine in SF3B1-mutated cells from newly diagnosed,
non-treated AML patients.
P389: BISPECIFIC ANTIBODIES ACTIVATE AUTOLOGOUS LEUKEMIA-REACTIVE T CELLS IN ACUTE
MYELOID LEUKEMIA
D. Böhm1,*, D. Primo2, J. Ballesteros2, K. Wennerberg1
1BRIC - Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen,
Denmark; 2Vivia Biotech, Tres Cantos, Madrid, Spain
Background: Acute myeloid leukemia (AML) is a highly immunosuppressive disease with
a high risk of relapse. This makes the finding of effective immunotherapy-based treatments
important, but also difficult. T cell-engaging bispecific antibodies (TCEs) are designed
to bind to both T cells and target cells, activate the T cells and thus induce an
immune response against the leukemia cells without employing the MHC-T cell receptor
(TCR) interaction. Several therapies of this kind are under development for treatment
of AML. Surprisingly, both in vitro and in vivo, a subset of these TCE immunocoached
T cells (ICTs) can subsequently target autologous AML cells and mediate efficient
TCR-MHC-dependent AML cell killing in the absence of the TCE. This points to that
a) a portion of the T cells present in an AML are leukemia-reactive but have been
immunosuppressed - a suppression that can be reversed by the addition of a TCE, b)
that this anti-AML T cell reactivation may play an important part of the mechanism
of action of AML-targeted TCEs and c) that by understanding this reactivation mechanism,
we will be able to design improved AML immunotherapies in the future.
Aims: To understand the mechanisms by which a TCE induces reactivation of leukemia-reactive
T cells (LRTs) in AML.
Methods: T cells in biobanked AML patient bone marrow samples were activated with
either an anti-CD3 x anti-CD123 TCE or a conventional anti-CD3 antibody for 5 days
before isolating the ICTs. ICT assay: ICTs were washed to remove the TCE, and isolated
ICTs were co-cultured with additional autologous bone marrow cells at different E:T
ratios. Killing efficiency and T cell expansion were determined over several days
using flow cytometry. Trogocytosis assay: ICTs were co-cultured with stained additional
autologous AML cells for 1 h and T cell populations of interest were isolated using
FACS. T cell repertoires were analyzed by targeted RNAseq of the TCR.
Results: ICTs generated with a TCE kill autologous AML cells efficiently without further
addition of the TCE. Adding additional TCE at this stage also does not increase the
AML killing efficiency of the ICTs. ICTs generated with an activating anti-CD3 antibody
show a similar AML killing efficiency and a similar trogocytotic behavior as TCE ICTs.
Most of the TCR clones present in the AML bone marrow pre-stimulation were also represented
among the ICTs and trogocytotic ICTs (ICTs that are directly targeting the AML cells
after either TCE or anti-CD3 stimulation; Figure 1), suggesting that the bulk of T
cells in an AML bone marrow represent heterogeneous immunosuppressed LRTs which can
be equally well reactivated by a TCE or an anti-CD3 antibody. Trogocytotic ICTs generated
from both types of T cell activation have a similar TCR distribution. ICTs derived
from the bone marrow target autologous AML cells more efficiently than ICTs from peripheral
blood.
Image:
Summary/Conclusion: Similarities between TCE ICTs and CD3 ICTs in regards to TCR distributions
and killing efficiencies suggest that the TCE reactivates leukemia-reactive T cells
via a CD3-dependent mechanism rather than a more complex proximity mechanism. Our
data further suggests that T cells in the AML bone marrow are highly heterogeneous
and leukemia targeted to a very high degree. In summary, AML bone marrow ICTs are
targeting and killing leukemic cells efficiently in a polyclonal fashion and our results
highlight that boosting this mechanism can leverage the clinical impact of AML TCEs
and be utilized in other novel AML immunotherapy approaches.
P390: ACUTE MYELOID LEUKEMIA SUPPRESSION BY PALBOCICLIB AND PONATINIB IN PATIENT-DERIVED
XENOGRAFT
D. Busa1,*, M. Culen1,2, T. Loja3, I. Jeziskova1,2, A. Folta1,2, J. Mayer1,2,3
1Department of Internal Medicine - Hematology and Oncology, Masaryk University; 2Department
of Internal Medicine - Hematology and Oncology, University Hospital Brno; 3Central
European Institute of Technology, Brno, Czechia
Background: Palbociclib, a CDK4/6 inhibitor approved for breast cancer treatment,
and ponatinib, a BCR/ABL1 inhibitor with a multi-kinase activity approved for chronic
myeloid and acute lymphoid leukemia, were previously shown to be effective in vitro
against different cancer types including acute myeloid leukemia (AML).
Aims: To test palbociclib and ponatinib, along with reference drugs – venetoclax,
azacitidine, cytarabine+doxorubicine (chemotherapy) on 2 primary AML samples in a
patient-derived xenograft model.
Methods: Two newly diagnosed AMLs (AML #1: myelomonocytic AML, intermediate cytogenetic
risk; AML #2: AML with myelodysplasia-related changes, poor cytogenetic risk) were
xenotransplanted into NOD SCID gamma mice. Treatment was initiated at detection of
approximately 5-20% hCD45+ cells in mouse peripheral blood (PB). Chemotherapy (cytarabine+doxorubicine,
AraC/Dox) was administered as a 5 + 3 regimen. Azacitidine was administered subcutaneously,
five days per week in 3 cycles – 1 week on, 1 week off. Palbociclib, ponatinib, venetoclax,
and vehicle were administered orally for 3 weeks, 5 days per week. Gene sequencing
was performed using targeted 37 gene Archer Variantplex Core Myeloid Kit panel.
Results: From the reference treatments, significant AML reduction and prolonged overall
survival (OS) was achieved with venetoclax in both AMLs, and with chemotherapy in
AML #1. No statistically significant effect was observed with azacitidine, but interestingly
in AML #2 the azacitidine treatment induced the longest remission in the whole study,
almost 10 weeks (<1% hCD45+ cells in PB) in 2/4 mice.
Of the tested drugs, palbociclib showed the same effect as venetoclax, with significantly
reduced AML and prolonged OS in both AMLs. Ponatinib led to prolonged OS only in AML
#1 (Fig. 1A-D).
Overall, none of the treatments led to complete AML eradication and all PDX groups
gradually relapsed.
No phenotype changes were observed in the relapsed cells in palbociclib and ponatinib
groups compared to vehicle (SSClowCD45dim blasts, SSCdimCD45high monocytes, CD34+
and CD34+CD38- cells). Azacitidine group showed altered monocyte, CD34+, and CD34+CD38-
fractions in AML #1. Azacitidine, chemotherapy, and venetoclax showed altered blast
and monocyte fraction in AML #2 and venetoclax also altered CD34+ fraction in AML
#2.
Mutational status in AML #1 remained unchanged (FLT3-TKD and NRAS mutated) in all
PDX groups at relapse. In AML #2, KRAS mutation from the primary tumor remained conserved,
whereas all PDXs lost an IDH1 mutation and gained a WT1 mutation. This indicates that
mutation selection was induced by the xenotransplantation rather than treatment. In
addition, a second WT1 mutation was gained only in the palbociclib group.
Treatment toxicity manifested by weight decrease after the end of treatment was only
seen with chemotherapy and was accompanied by early mouse mortality in 3/4 mice for
AML #2.
Image:
Summary/Conclusion: Palbociclib demonstrated in vivo AML suppression that was comparable
to venetoclax and superior to chemotherapy. Effect of ponatinib was only limited.
This encourages further investigation of treatment efficacy in different AML subtypes
and in combination with other drugs.
Funding: MUNI/A/1330/2021
P391: FLT3-ITD MEASUREMENT AT DIAGNOSIS AND FOR THE ASSESSMENT OF MINIMAL RESIDUAL
DISEASE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH
ACUTE MYELOID LEUKEMIA: CDNA VS. DNA
D. Carbonell1,2,*, M. Chicano1,2, A. Cardero-Illán1, G. Rodriguez-Macías1, P. Muñiz1,2,
R. Bailén1,2, G. Oarbeascoa1,2, I. Gómez-Centurión1,2, J. Anguita1,2, M. Kwon1,2,
J. L. Díez-Martín1,2,3, I. Buño1,2,4,5, C. Martínez-Laperche1,2
1Hospital General Universitario Gregorio Marañón; 2Instituto de Investigación Sanitaria
Gregorio Marañón; 3Department of Medicine; 4Department of Cell Biology, Complutense
University of Madrid; 5Genomics Unit, Instituto de Investigación Sanitaria Gregorio
Marañón, Madrid, Spain
Background: DNA is standardly preferred to cDNA for the analysis of FLT3- internal
tandem duplication (ITD). Although its screening is mandatory at diagnosis in clinical
routine, its use as minimal residual disease (MRD) marker after allogeneic hematopoietic
stem cell transplantation (allo-HSCT) remains controversial due to the poor sensitivity
of its detection method and its apparent instability during the disease.
Aims: To compare FLT3-ITD mutation analysis in DNA and cDNA samples at diagnosis and
to prove the usefulness of its expression measurement as MRD marker after allo-HSCT.
Methods: Forty-six diagnosis samples from 46 patients and 80 samples from 34 patients
who underwent an allo-HSCT (34 pre-HSCT samples, 34 at day 30 after infusion and 12
before relapse) were analyzed. DNA was purified using a Maxwell RSC Blood DNA Kit
(Promega), RNA was isolated using TRIzol reagent (Invitrogen) and cDNA synthesis was
performed using the First Strand cDNA Synthesis kit (Roche). FLT3-ITD was analyzed
in both DNA and cDNA samples by fragment analysis using an ABI3130xl DNA sequencer
(Applied Biosystems). Fragment analysis was analyzed through Peak Scanner Software
(Thermo Fisher). Allelic ratio (AR) quantification was performed by dividing the area
under the curve of the mutant allele by the area under the curve of the wild type
allele. Every clone was analyzed as an event, comparing its DNA ratio with its cDNA
ratio.
Results: Median AR of the 58 FLT3-ITD clones at diagnosis were 0.54 [0-9.47] in DNA
samples and 0.63 [0.012-13] in cDNA samples, (Wilcoxon test, p<0.001, Figure A). In
six patients, FLT3-ITD AR was <0.5 in DNA and ≥0.5 in cDNA, changing their prognosis
based on ELN algorithm. One of them was not candidate for chemotherapy and of the
five remaining, four were refractory or relapsed after intensive treatment. In terms
of sensitivity, cDNA was better than DNA, quantifying higher AR values in most cases,
optimizing the detection of minor clones and the prognostic classification. In respect
of HSCT samples, seven patients were positive for FLT3-ITD mutation pre-HSCT in cDNA
and two in DNA. Of the seven patients which were positive by cDNA, four relapsed after
HSCT. The three patients who did not relapse had FLT3-ITD AR lower than 0.05. As for
the two patients who tested positive in the DNA samples, only the one who also tested
positive for cDNA relapsed. Despite the small sample size, these results reveal that
the analysis of pre-HSCT cDNA samples could have a predictive value for relapse. No
patient was positive on day 30 after HSCT, suggesting that it may not be useful, whether
the DNA or cDNA sample is studied. However, the analysis at this time point could
be crucial in those patients who may relapse early. On respect of the last sample
before relapse (a median of 22 days before relapse, range 7-85), of the 12 cases,
three were positive for FLT3-ITD mutation in DNA samples and nine were positive in
cDNA samples. Great differences were observed between AR of both type of samples (Wilcoxon
test, p<0.001, median 0 [0-0.04] vs. 0.15 [0-0.57], Figure B). In three patients FLT3-ITD
cDNA measurement did not anticipate the relapse, due to the loss of the mutation or
because the day on which the sample was analyzed was too early to detect the mutation,
issues that can be solved by complementing with other MRD markers.
Image:
Summary/Conclusion: In conclusion, cDNA fragment analysis of FLT3-ITD by capillary
electrophoresis is an easy-to-implement technique that could be a great alternative
approach in AML patients at diagnosis and in allo-HSCT monitoring.
P392: PHARMACOLOGICAL INHIBITION OF SYK CONFERS ANTI-PROLIFERATIVE AND NOVEL ANTI-TUMOR
IMMUNE RESPONSES IN AML
L. A. Carvajal1,*, B. Robinson2, Y. Kosaka2, T. Jacob2, J. Lee2, T. Hood1, K. Baker2,
A. Kaempf2, S. N.-A. Amara1, J. Pucilowska2, E. Lind2, C. Tognon2, J. Tyner2, P. Kumar1,
T. Vu2, J. DiMartino1
1Kronos Bio, Inc., San Mateo; 2Oregon Health & Science University (OHSU), Portland,
United States of America
Background: Spleen tyrosine kinase (SYK) acts as a key integrator of signals from
cell surface receptors containing an immunoreceptor tyrosine-based activation motif
to boost cellular proliferation. In acute myeloid leukemia (AML), SYK serves as a
relay to an oncogenic transcriptional regulatory network linked to NPM1, HOXA9 and
MEIS1.
The selective, orally bioavailable SYK inhibitor entospletinib (ENTO) has demonstrated
clinical activity and tolerability in HOXA9/MEIS1-driven AML. ENTO is currently being
investigated in a global Phase 3 trial, AGILITY (NCT05020665), in combination with
intensive induction/consolidation chemotherapy in patients (pts) with treatment-naive
NPM1-mutated (NPM1m) AML. Lanraplenib (LANRA) is a next-generation SYK inhibitor with
similar potency and selectivity to ENTO but with more favorable pharmacologic properties
that is currently being evaluated in combination with gilteritinib in pts with relapsed
or refractory FLT3-mutated AML (NCT05028751).
Aims: To investigate the effects of ENTO and LANRA in T-cell responses in AML.
Methods: All pt-derived bone marrow (BM) and peripheral blood samples were obtained
in accordance with IRB (OHSU#004422) approval. Results from gene expression profiling
studies (RNA sequence [RNA-seq]) in bone marrow/peripheral blood mononuclear cells
(MNCs) derived from 152 pts with AML, for which matching NPM1 status and ex vivo sensitivity
to ENTO was available, previously published by Tyner et al (2018), were reanalyzed
using trimmed mean of M values normalization and differential expression using edgeR.
ENTO/LANRA sensitivity was assessed ex vivo after 72 hours in culture at 37°C by MTS
assay. T-cell functional assays were performed by culturing MNCs with anti-CD3 and
treating with ENTO or LANRA, singly and in combination with anti-PD1. CD3+ T-cell
proliferation as measured by Ki67 expression and pSYK activation were assessed by
quantitative single-cell immunofluorescence microscopy. Six formalin-fixed, paraffin-embedded
BM biopsies from newly diagnosed AML pts were assessed for spatial expression of mRNA
using a digital spatial profiling method.
Results: In AML, mutations in NPM1 with co-expression of HOXA9/MEIS1 at baseline predicted
anti-proliferative activity to ENTO in ex vivo drug sensitivity studies. Accordingly,
treatment of leukemic cells with either ENTO or LANRA inhibited SYK auto-phosphorylation
in a dose-dependent manner. Pathway analysis of archival RNA-seq data from pts enrolled
in the Leukemia and Lymphoma Society BEAT AML Master Protocol (NCT03013998 [BAML-16-001-S6])
revealed gene expression signatures at baseline associated with the observed sensitivity
to ENTO, including significant enrichment in the expression of genes associated with
leukemogenesis, myeloid differentiation and immune regulation. This was supported
by T-cell functional studies, which demonstrated that ENTO and LANRA, singly as well
as in combination with anti-PD1, could restore T-cell proliferation in primary AML
pt samples that exhibited suppression. Lastly, Cancer Transcriptome Atlas analysis
on archival BM biopsies obtained from these AML pts showed a strong correlation between
immune checkpoint response and gene expression associated with immune pathway signaling.
Summary/Conclusion: Inhibition of SYK is a promising therapeutic approach in NPM1m
AML. Our studies illustrate that ENTO and LANRA may restore T-cell proliferation in
a subset of AML pts with dysfunctional T-cell responses, suggesting a novel mechanism
of action. Additional studies are required to fully understand the mechanism of SYK-mediated
antitumor immune responses in AML.
P393: SYNERGISTIC EFFECT OF EZH2 INHIBITOR WITH BCL2 INHIBITOR BY ENHANCING PIK3IP1
EXPRESSION IN ACUTE MYELOID LEUKEMIA
C. Yang1, C. Song2,3, Z. Ge1,*
1Department of Hematology, Zhongda Hospital,Medical School of Southeast University,
Institute of Hematology Southeast University, Nanjing, China; 2Hershey Medical Center,
Pennsylvania State University Medical College, Hershey; 3Division of Hematology, The
Ohio State University Wexner Medical Center, the James Cancer Hospital, Columbus,
United States of America
Background: Acute Myeloid Leukemia(AML) is a highly heterogeneous blood cancer with
poor outcomes. PI3K Interacting Protein 1(PIK3IP1) binds to the p110 catalytic submit
and downregulate PI3K activity. EZH2 is the catalytic subunit of polycomb repressive
complex 2(PRC2). Inhibition of EZH2 activity is a therapeutic vulnerability in cancer
cells. Combination of the BCL2 inhibitor Venetoclax(Ven) with hypomethylating agents
has been approved for treatment in AML with substantial benefits, however, most patients
develop resistance. Thus, a more efficacious treatment by targeting the drug resistance
paradigm based on BCL2 inhibitor is required.
Aims: To investigate the anti-leukemic activity of the combination of the EZH2 inhibitor
DZNeP with BCL2 inhibitor Ven and to evaluate the association of PIK3IP1 with clinical
characteristics of patients with AML.
Methods: EZH2 and PIK3IP1 mRNA level was examined by qPCR in newly-diagnosed AML patients
and healthy control from Feb. 1, 2016, to Jan. 30, 2022, at our institute with approval
of the Ethics Committee. Cell Counting Kit-8 assay(CCK8) was used for cell proliferation
and cytotoxicity in U937 and MV-4-11 AML cells treated with vehicle control, EZH2
inhibitor(DZNeP), Ven, and Combination (Ven+DZNeP). Synergistic effect was analyzed
with Calcusyn. RNA-seq was performed with total RNA isolated from U937 cells treated
with 2μM DZNeP for 48 hours. Apoptosis was measured by cell staining with Annexin
V+propidium iodide (PI) following flow cytometry analysis. Lentiviral PIK3IP1 shRNA
knockdown was performed in AML cells. Gene expression was analyzed by qPCR and Western
blot.
Results: EZH2 mRNA level was significantly increased in 53 newly diagnosed patients
with AML compared to 20 healthy bone marrow control(p<0.0001)(Fig.1A). Both DZNeP
and Ven had a dose-dependent and time-dependent effect on cell proliferation arrest
in AML cells. DZNeP significantly sensitized the effect of Ven on cell proliferation
arrest compared to single drug-only control. CalcuSyn analysis showed the synergistic
effect of the Ven+DZNeP(Fig.1B). The total apoptosis rate increased significantly
in the Ven+DZNeP group(Fig.1C). RNAseq analyses showed that PIK3IP1 is dramatically
upregulated(Fig.1D). GSEA enrichment analyses showed that the Differentially Expressed
Genes mainly upregulated apoptosis and downregulated cell cycle pathway(Fig.1E). Moreover,
the PIK3IP1 mRNA level was significantly decreased in the aforementioned AML cohort
compared to the control(P<0.005)(Fig.1F). The AML patients were divided into two PIK3IP1High
expression groups(n=29) and PIK3IP1Low expression groups(n=24). PIK3IP1low expression
is significantly associated with a lower overall response rate (96.55% vs 79.17%,
p=0.047), higher relapse or death rate(58.33% vs 24.14%, p=0.029). A moderately negative
correlation between EZH2 and PIK3IP1 was observed(r=-0.376, p=0.006)(Fig.1G). In addition,
qPCR data showed that PIK3IP1 is significantly upregulated by DZNeP or Ven+DZNeP(Fig.
1H). PIK3IP1 is efficiently knocked down in U937 and MV-4-11(Fig.1I), and PIK3IP1
knockdown significantly blocked the combination of DZNeP and Ven mediated cell proliferation
arrest and apoptosis(Fig.1J-K).
Image:
Summary/Conclusion: The combination of EZH2 inhibitor DZNeP and BCL2 inhibitor Venetoclax
exhibited a strong synergistic cell proliferation arrest and apoptosis of AML cells
in PIK3IP1 dependent manner. PIK3IP1low expression is associated with oncogenic markers
and poor outcomes in AML. Our results highlight the likelihood of further in vivo
pre-clinical study for the potential clinical trial of the novel combination in AML
patients.
P394: INSIGHTS INTO PATIENT-DERIVED XENOGRAFT MODELS FOR DEK-NUP214 LEUKEMIA
F. Charles Cano1,*, A. Kloos1, T. Fangmann1, N. Kattre1, R. Schottmann1, K. Döhner2,
A. Ganser1, M. Heuser1
1Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Medizinische
Hochschule Hannover, Hannover; 2Department of Internal Medicine III, University Hospital
of Ulm, Ulm, Germany
Background: Acute myeloid leukaemia (AML) with the translocation DEK-NUP214 (t(6;9)(p23;q34))
is a rare subtype present in 1% of adult and 2% of paediatric patients and is recognized
by the world health organization (WHO) as a unique subgroup of AML. These patients,
typically, are diagnosed with de novo AML and have poor clinical outcome and resistance
to chemotherapy. DEK-NUP214 is the sole cytogenetic abnormality in the majority of
these patients at the time of diagnosis, suggesting its central role in disease development.
This subgroup of leukaemia remains poorly characterized. There is only one cell line
(FKH-1) and only very few reported DEK-NUP214 patient derived xenograft (PDX) models.
The major limitation is the lack of serially transplantable PDX models, which are
required for pharmacologic studies. The development of DEK-NUP214 PDX models will
aid in the understanding of the molecular pathogenesis of this disease.
Aims: To establish patient-derived xenograft models that recapitulate DEK-NUP214 leukaemia.
Methods: Five patient samples were selected based on their cytogenetic profile and
1x106 cells were injected intravenously into irradiated NSG-SGM3 (NSGS) mice (n=3)
per sample. Engraftment of human cells in peripheral blood by flow cytometry and blood
counts was monitored monthly. Engrafted cells were used for serial transplantations.
Cytospin preparations of peripheral blood, spleen and bone marrow of the PDX samples
as well as the patient samples were stained with Wright-Giemsa stain. The DEK-NUP214
fusion transcript was confirmed by Sanger sequencing. Mutational analysis was performed
with a custom myeloid sequencing panel.
Results: Three patient samples showed a mean of 12.5±6.0 (PDX1), 0.68±0.23 (PDX2)
and 4.9±2.5% (PDX3) engraftment of human CD45+CD33+ cells at 20-24 weeks in peripheral
blood and were transplanted into secondary mice. The characteristics of the patients
at diagnosis, whose samples engrafted, were 19, 53 and 51 years old, respectively,
with a median of 52.8 x109/L (range 34.1-116 x109/L) white blood cells (WBC), 7.4 g/dl
(range 6-9.4 g/dl) of haemoglobin (Hb), and 38 x109/L (range 26-61 x109/L) platelets
(Plt). The average circulating blasts were 56.7±18% in peripheral blood and blasts
in bone marrow were 84.7±4.7%. PDX models at the 1st transplantation (Tx) had an increased
WBC and decreased Hb at the time of sacrifice. Average spleen weights were 196±24.1,
59.7±3.18, and 103±35.4 g, respectively. The morphological analysis revealed large
blasts with partially granulated cytoplasm and few vacuoles similar to the patients’
cells. The engrafted cells showed a myeloid phenotype with expression of CD38, CD33
and CD14, with higher engraftment in bone marrow than spleen. Secondary transplants
from PDX1 showed 12.5% of hCD45+ cells at 16 weeks in peripheral blood, with average
spleen weight of 168 g, with a similar immunephenotype as primary mice, and were transplanted
into tertiary mice. Mutational analysis showed that the PDX samples retained the same
mutations as observed in the patient sample (FLT3-ITD for PDX1 and PDX2, and WT1 for
PDX1) and the fusion transcript as well.
Summary/Conclusion: We have successfully established AML PDX models for DEK-NUP214
leukaemia that reflect the morphological, immunophenotypic and molecular genetics
of this disease subtype. This will enable pharmacologic studies in this rare disease
and will allow a better understanding of the involved pathways.
P395: PHARMACEUTICAL TARGETING OF RAS IN ACUTE MYELOID LEUKAEMIA
D. Coleman1,*, L. Strate1, J. Griffin1, T. Rabbitts2, P. Cockerill1, C. Bonifer1
1University of Birmingham, Birmingham; 2The Institute of Cancer Research, London,
United Kingdom
Background: Acute Myeloid Leukaemia is characterised by a small number of co-occurring
driver mutations which combine to make a diverse collection of disease subtypes. Mutations
in genes associated with cell signalling are a key group of driver mutations and lead
to the constitutive activation of these proteins and their associated signalling cascades.
RAS mutations occur in 6% of AML and confer a proliferative advantage. However, 36%
of AMLs include mutations which lead to activated RAS such as FLT3 mutations and are
associated with a worse outcome after induction chemotherapy1.
Inhibitors which bind to mutated and activated members of the RAS protein family to
block the interaction between the protein and its downstream targets have been developed2
which allow us to investigate the effects of pan-RAS inhibition in Acute Myeloid Leukaemia.
These Pan-RAS inhibitors inhibit RAS-RAF and RAS-PI3K signalling leading to reduced
phosphorylation of ERK and AKT.
Aims: 1. We have used novel specific and panRAS RAS inhibitors designed in the Rabbitts
lab followed by system-wide analyses to identify the specific gene regulatory network
of mutant RAS AML and identify RAS-responsive genes
2. We have interrorgated key transcription factor nodes with RNAi
Methods: Primary cells were co-cultured on primary hMSCs for perturbation experiments.
Fresh blood or bone marrow samples from patients with AML were purified and sorted
for CD34 or CD117, these samples were profiled by RNA-seq an ATAC-seq. Data from multiple
samples with RAS mutations was integrated and compared to data from healthy CD34+
PBSCs to establish the gene regulagtory network.
Results: In vitro panRAS inhibitors cause cytotoxicity in primary AML samples with
RAS activating mutations whilst cells with WT RAS exhibit reduced sensitivity. An
exception to this finding are cells with FLT3-ITD mutations, which were also sensitive
to RAS inhibition. This is of particular interest as patients treated with FLT3 inhibitors
can relapse with a RAS mutant subclone.
To understand the role of RAS mutations in AML we have constructed a gene regulatory
network from the genomic and transcriptomic profiling of primary cell samples from
patients which has allowed us to identify a core network of transcription factors.
The direct association of aberrant RAS signalling with these transcription factors
has been determined through perturbation experiments using RAS inhibitors, and by
targeting key transcription factor nodes with RNAi. These include transcription factors
ETV5 and KANK1, both of which have been linked to RAS signalling in solid tumour models3,4.
Summary/Conclusion: These experiments will identify targets of the RAS pathway essential
for the survival of AML cells.
1. Ball, B.J., et al. (2019), “RAS Mutations Are Independently Associated with Decreased
Overall Survival and Event-Free Survival in Patients with AML Receiving Induction
Chemotherapy.”
Blood, 134(Supplement_1): 18-18.
2. Cruz-Migoni, A., et al. (2019), “Structure-based development of new RAS-effector
inhibitors from a combination of active and inactive RAS-binding compounds.” Proceedings
of the National Academy of Sciences
116(7): 2545-2550.
3. Mus, L.M., et al. (2020), “The ETS transcription factor ETV5 is a target of activated
ALK in neuroblastoma contributing to increased tumour aggressiveness.” Sci Rep
10(218)
4. Jonathan R. Dry, et al. (2010), “Transcriptional Pathway Signatures Predict MEK
Addiction and Response to Selumetinib (AZD6244)”. Cancer Res; 70(6): 2264–2273
P396: DYSREGULATED DNAM-1 AND KIR2DL1 EXPRESSION IN NATURAL KILLER CELLS CORRELATES
WITH POOR OUTCOME IN ACUTE MYELOID LEUKEMIA
A. F O Costa1,2,*, L. O Marani1, I. A Lopes1, L. S Binelli1, P. S Scheucher1, J. L
S Schiavinato1, M. I. A Madeira1, K. B B Pagnano3, B. K Duarte3, A. B. F Glória4,
E. M Rego5, F. Traina1, R. Welner2, L. L Figueiredo-Pontes1
1Hematology Division, Department of Medical Images, Hematology, and Clinical Oncology,
Ribeirao Preto Medical School at University of Sao Paulo, Ribeirao Preto, Brazil;
2Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer
Center, University of Alabama at Birmingham, Birmingham, United States of America;
3Hemocentro, University of Campinas, Campinas; 4Hematology Division, Hospital das
Clinicas da Universidade Federal de Minas Gerais, Belo Horizonte; 5Internal Medicine,
Medical School of the University of Sao Paulo, Sao Paulo, Brazil
Background: Although improvements have been made in understanding the physiopathology
of Acute Myeloid Leukemia (AML), most patients still relapse leading to poor long-term
prognosis and cure rates. Natural Killer (NK) cells are regulated by opposing signals
from receptors that activate and inhibit effector function and are known to mediate
anti-leukemic immunosurveillance in AML, but the mechanisms underlying the control
of hematopoietic neoplasms by these cells remain unclear.
Aims: We hypothesized that FLT3-ITD mutation, a molecular marker of adverse prognosis,
is associated with phenotypically and functionally impaired NK cells due to imbalanced
expression of inhibitory and activating receptors during AML.
Methods: We analyzed the expression of DNAM-1 and KNG2D activating and NKG2A, KIR2DL1
inhibitory receptors on CD56+, CD56bright CD16- and CD56dim CD16+ NK subsets and its
correlation with FLT3-ITD mutation in 70 AML patients and 12 healthy bone marrows
(NBM) by flow cytometry. Seven marrows from AML patients in complete remission after
induction chemotherapy (AML-CR) were used to evaluate NK cell and receptor recovery.
Mann Whitney or Kruskal-Wallis tests (P < 0.05) were performed using GraphPad Prism
(V8.0.2).
Results: A decreased frequency of all NK cell subsets was found in AML (CD56+, P=0.03;
CD56bright, P=0.04, CD56dim, P<0.01). Decreased expression of NKG2D (P<0.01) and a
tendency of decreasing DNAM-1 (P=0.38) was found in CD56+ NK in AML, recovering to
values closer to normal in AML-CR. Expression of NKG2D was decreased in both CD56bright
and CD56dim NK subsets (P=0.03 and P<0.01), while DNAM-1 was found to be considerably
decreased in CD56dim cytotoxic NK (P=0.02). FLT3 mutated patients showed even lower
expression of DNAM-1 (P=0.02) and NKG2D (P=0.13) in CD56+ NK. Even though the latter
was not statistically significant, we observed a decreasing pattern in these patients,
that was confirmed by an important decrease of both activating receptors uniquely
in the CD56dim subset (DNAM-1, P=0.03; NKG2D, P=0.05), which are known to contain
the most cytotoxic activity. As for the inhibitory receptors, our cohort demonstrated
a trending increase in NKG2A expression on all NK subsets, although not significant,
and no difference was found in FLT3 mutated patients. Meanwhile, KIR2DL1 expression
was higher in CD56+, CD56bright and CD56dim NK cells in AML (P<0.01; P=0.06 and P<0.01),
while AML-CR showed frequencies comparable to healthy In FLT3-ITD mutated patients,
CD56dim NK had increased expression of KIR2DL1 (P=0.03), strongly correlating with
an imbalance between activation and inhibition of NK cell activity.
Summary/Conclusion: Our data indicate that CD56dim NK show an exhaustion immunophenotype
in FLT3-ITD mutated AML, with high expression of KIR2DL1 inhibitory receptor and low
expression of both NKG2D and DNAM-1 activating receptors and that this imbalance likely
leads to impaired cytotoxicity, compromised anti-leukemic activity and could be contributing
to worst disease outcome, which will be confirmed through functional assays. Closing
these gaps in knowledge informs the fundamental characteristics of NK dysfunction
during leukemia and is of significant interest for targeting, therapeutic interventions
and NK cell-mediated immunotherapy.
P397: PROLACTIN RECEPTOR CONFERS CHEMORESISTANCE DUE TO SENESCENCE ENTRANCE IN ACUTE
MYELOID LEUKEMIA
L. Cuesta-Casanovas1,2,*, J. M. Cornet-Masana1, J. M. Carbó1, J. Delgado-Martínez1,3,
L. Clément-Demange4, A. Banús-Mulet5, F. Guijarro6,7, J. Esteve5,6,7,8, R. M. Risueño1
1Leukemia Stem Cell, Josep Carreras Leukemia Research Institute, Badalona; 2Faculty
of Bioscience, Autonomous University of Barcelona; 3Faculty of Pharmacy, University
of Barcelona; 4Leukos Biotech, Barcelona; 5Josep Carreras Leukemia Research Institute,
Badalona; 6Faculty of Medicine, University of Barcelona; 7Department of Hematology,
Hospital Clínic; 8Institut d’Investigacions Biomèdiques August Pi I Sunyer, Barcelona,
Spain
Background: Acute myeloid leukaemia (AML) is a hematopoietic neoplasm characterized
by frequent relapse because of chemotherapy resistance, which confers poor outcome.
For this reason, the study of new therapeutic approaches that completely eradicate
the disease is an urgent need. A previous in silico analysis performed in our laboratory
demonstrated that PRLR signalling pathway is overactivated in leukemic stem cells
(LSCs) in comparison to hematopoietic stem cells (HSCs), suggesting a relationship
between PRLR and AML resistance and progression.
Aims: The main objective was to decipher the role of PRLR and its signalling pathway
in the transformation events associated with the initiation and maintenance of AML.
Methods: mRNA expression of PRLR and Stat5a was analysed from public repositories.
Surface PRLR expression was assessed by flow cytometry and total protein expression
by western blot. Stat5 activation (phosphorylation status) and induced gene regulation
were determined in AML cells ectopically expressing the wildtype PRLR (PRLRwt) and
the dominant negative isoform (PRLRsh) and treated with its natural ligand (PRL) or
an antagonist (G129R-PRL), by western blot and luciferase reporters. Senescence status
was analysed by SA-β-galactosidase staining by microscopy and quantified using ImageJ
software. Cytarabine resistant cells (AraC-R) were obtained by treating them during
several weeks with increasing doses of cytarabine. PRLR expression was silenced using
the CRISPR-Cas9 technology. Clonogenicity was studied in AML samples and healthy linage-depleted
cord blood cells treated with PRL and hPRL-G129R and seeded in a semisolid medium
in presence of instructive cytokines.
Results: AML patient cells express higher levels of PRLR in terms of mRNA, protein
surface expression and total protein in comparison to healthy donors. Stat5 mRNA was
overexpressed in AML, and PRL induced its phosphorylation and activation in presence
of PRLR wt isoform, while the dominant negative did not respond to PRL. The antagonist
of the receptor did not induce any effect. Stat5 mRNA expression was increased at
relapse, and so it was the PRLR surface and total protein when AML cells acquire chemoresistance.
Moreover, PRLR wildtype cells showed greater levels of SA-β-galactosidase, an indicative
of the entrance in senescence. This effect was reverted when PRLR was knocked-down
using CRISPR-Cas9 technology. The inhibition of PRLR with an antagonist or its downregulation
with the CRISPR-Cas9 technology decreased clonogenicity capacity of AML patient cells,
without affecting healthy donor cells.
Summary/Conclusion: PRLR expression is higher in AML patient samples, and correlates
with chemoresistance and refractoriness, making the receptor an adequate biomarker
for AML diagnosis and follow-up. PRLR-PRL signalling promotes the activation of Stat5,
which is increased in AML samples, especially at relapse, suggesting an important
role in AML progression and chemoresistance, a process related with increasing levels
of PRLR surface expression. Moreover, the suppression of PRLR expression sensitizes
the cells to chemotherapy.
Those results demonstrate for the first time that PRLR suppression increases the sensitivity
to chemotherapy and decreases the clonogenicity capacity of AML cells, suggesting
the use of antagonists of the receptor as new therapeutic strategies against AML.
P398: DEVELOPMENT AND CHARACTERIZATION OF PRECLINICAL IN VIVO MODELS FOR THE IDENTIFICATION
AND TESTING OF NEW THERAPEUTIC APPROACHES FOR PEDIATRIC ACUTE MYELOID LEUKEMIA
A. Da Ros1,*, V. Indio2, E. Porcù1, G. Borella1, M. Benetton1, C. Tregnago1, G. Longo1,
S. Cairo3, B. Michielotto1, S. Bresolin1, B. Buldini1, A. Pession4, F. Locatelli5,
M. Pigazzi1
1Department of Women’s and Children’s Health, Haematology-Oncology Clinic and Laboratory,
University of Padova, Padova; 2Department of veterinary medical science, University
of Bolgna, Bologna, Italy; 3XenTech, Evry, France; 4Pediatric Unit, IRCCS Azienda
Ospedaliera-University of Bologna, Bologna; 5Department of Pediatric Hematology and
Oncology, IRCCS Bambino Gesù Children’s Hospital, Sapienza University of Rome, Rome,
Italy
Background: In pediatric acute myeloid leukemia (AML) chemotherapy is the standard
of care, but >25% of patients still relapse and after a disease recurrence the survival
probability is extremely low (<50%). To ameliorate patients’ outcome there is an urgent
need to discover new treatments. Nevertheless, pediatric drug development is extremely
reduced by the need of a better understanding of the adverse event profile of adult
cancer indications in children, by the lack of pediatric-specific formulations, and
by the reduced number of pediatric AML patients that can be included in clinical trials.
Thus, robust preclinical AML models to faithfully predict new drug efficacy is urgently
needed to advance new drugs in clinical setting.
Aims: This study aims to generate and characterize AML patient derived xenografts
(PDXs) and accelerate the evaluation of innovative medicines for AML.
Methods: We generated PDXs from primary AML samples by inoculating blasts in NSG mice
and, when engrafted, in 3 consequent mice recipients (namely P0, P1 and P2-PDX). We
characterized AML by immunophenotipic profile and by RNA and whole-exome sequencing
(WES). According to somatic mutations and copy number alterations we determined AML
clonal compositions. We selected drugs and performed drug testing in vivo, after the
expansion of P3-PDXs.
Results: We generated 22 AML-PDXs representing high-risk AML subtypes for genetic
characterization harboring NUP98-NSD1, or NPM1-MLF1, or CBFA2T3-GLIS2, or FUS-ERG
or KMT2A somatic translocations, or FLT3-ITD mutation. We monitored the AML associated
immunophenotype in PDXs finding it was similar to that of the original AML. By WES
we detected a consistent number of variants in each patients’ AML (ranging from 28
to 69), confirming an high AML intra-tumoral heterogeneity. Furthermore, we did not
find any mutation recurrence among models, underlining an high inter-tumoral heterogeneity.
In all models we tracked clonal evolution from patients’ AML to P2-PDX highlighting
that most of the variants were maintained, with very few variants acquired during
model development. Monitoring clonal dynamics we recognize a specific “founder” clone
characterized by an average of 30 variants which are maintained up to P2 at the same
allelic frequency, other small clones with average of 10 variants increasing the allelic
frequencies in P2 and, in a restricted number of models, we observed that some clones
were lost. By WES and transcriptome analysis we highlighted druggable mutations and
pathways allowing the selection of novel targeted drugs. We screened their efficacy
in vitro alone or combined with chemotherapic (Arabinoside) and biological agents
(Venetoclax) by using AML ex vivo cells and mesenchymal stromal cells in a 3D co-culture
system for exploring their synergy in reducing AML proliferation. Four selected drugs
are under evaluation in AML-PDX models.
Summary/Conclusion: We have created a series of paired AML and xenograft models for
advancing pediatric AML therapeutics. Our models represent a concrete perspective
for both, the identification of new variants and pathways involved in AML progression,
and the possibility to perform novel drug screenings useful to increase AML drug portfolio.
P399: UNRAVELING THE TUMOR SUPPRESSIVE ROLE OF STAT3Β IN ACUTE MYELOID LEUKEMIA
S. Edtmayer1,*, A. Witalisz-Siepracka1, B. Zdársky1, T. Eder2, V. Poli3, F. Grebien2,
D. Stoiber1
1Department of Pharmacology, Physiology and Microbiology, Karl Landsteiner University
of Health Sciences, Krems an der Donau; 2Institute of Medical Biochemistry, University
of Veterinary Medicine Vienna, Vienna, Austria; 3Department of Molecular Biotechnology
and Health Sciences, University of Torino, Turin, Italy
Background: Signal transducer and activator of transcription 3 (STAT3) is a mediator
of cytokine signaling existing in two alternatively spliced isoforms known as the
full-length isoform STAT3α (770aa) and the C-terminally truncated isoform STAT3β (722aa).
Initially STAT3β was described as the dominant negative form of STAT3α. It gained
more attention as it turned out to initiate transcription independent of STAT3α and
being capable to rescue embryonic lethality in absence of STAT3α. In acute myeloid
leukemia (AML) patients, STAT3 has been reported to be constitutively activated thereby
promoting proliferation and blast survival. However, most of those studies did not
consider the two distinct isoforms of STAT3. Recently, our group identified STAT3β
as a novel tumor suppressor and favorable prognostic marker in AML, however the underlying
mechanism in leukemic cells remained elusive.
Aims: With this study we aim to provide in-depth knowledge on the role of STAT3 isoforms
in AML development and disease outcome.
Methods: By using murine AML cell lines lacking either STAT3α or STAT3β, we aim to
elucidate the STAT3 isoform-specific impact on cellular processes shaping AML development
and progression. To gain better understanding we use AML in vivo models and in vitro
assays in combination with next-generation sequencing (NGS) approaches to identify
molecular mechanisms explaining the tumor suppressive function of STAT3β in AML. In
brief, fetal liver-derived stem cells, isolated from STAT3α and STAT3β deficient mice
on C57BL/6J background, were transduced with a retrovirus introducing the human fusion-oncogene
MLL-AF9, and intravenously transplanted into immunocompromised mice.
Results: Animals transplanted with leukemic cells lacking STAT3β showed accelerated
disease progression and poorer overall survival confirming its tumor suppressive properties.
Furthermore, absence of STAT3β favored leukemic infiltration and migration. Additionally,
RNA sequencing revealed that lack of STAT3β in leukemic cells leads to a dysregulation
of crucial genes driving leukemogenesis.
Summary/Conclusion: Understanding the mechanisms behind the tumor suppressive property
of STAT3β has potential translational impact and is crucial to identify novel therapies
and preventative strategies. Further validation and ChIP sequencing will provide novel
direct target genes of STAT3β which could serve as prognostic or therapeutic molecules.
P400: PROTEOGENOMIC CHARACTERIZATION OF 5-AZACYTIDINE EFFECTS ON ACUTE MYELOID LEUKEMIA
IMMUNOPEPTIDOME
G. Ehx1,*, N. Nandita2, C. Durette2, J. Courtois1, M.-P. Hardy2, K. Vincent2, J.-P.
Laverdure2, J. Lanoix2, F. Baron1, P. Thibault2, C. Perreault2
1GIGA-I3: Hematology, University of Liege, Liege, Belgium; 2IRIC, University of Montreal,
Montreal, Canada
Background: Hypomethylating agents like 5-azacytidine (AZA) are licensed for the treatment
of acute myeloid leukemia (AML) patients ineligible for allogeneic hematopoietic cell
transplantation. While previous reports suggested that AZA promotes the recognition
of AML blasts by cytotoxic T cells, the mechanism behind this improved recognition
is not fully understood. Specifically, AZA is assumed to promote the expression of
transcripts repressed by genomic methylation, namely cancer-testis antigens (CTA)
and endogenous retroelements (ERE), resulting in the presentation of immunogenic MHC-I-associated
peptides (MAPs, collectively referred to as the immunopeptidome) derived from the
translation of these transcripts. However, the presentation of such MAPs after AZA
treatment has not been firmly demonstrated thus far.
Aims: Our study aims to characterize how AZA treatment shapes the identity of MAPs
presented by AML cells.
Methods: Four different AML cell lines, THP-1, OCI-AML3, SKM-1, and MOLM-13 were treated
with a non-cytotoxic dose of AZA for 3 days. Their transcriptome has been characterized
by high-coverage RNA sequencing (RNA-seq) and their immunopeptidome by shotgun mass
spectrometry (MS). To identify MAPs deriving from unannotated genomic regions, we
have designed a cutting-edge proteogenomic pipeline using the RNA-seq data to build
personalized MS databases that enabled the identification of non-canonical MAPs such
as ERE-derived MAPs.
Results: Paired transcriptomic comparisons between treated and untreated cells showed
that AZA induces a large-scale gene upregulation (87% differentially-expressed transcripts
were upregulated). Among them, 38% were EREs and 6% were CTAs, suggesting that AZA-induced
MAPs have greater chances of deriving from EREs than from CTAs. However, we could
not identify a single ERE-derived upregulated MAP among the immunopeptidome of AZA-treated
AML cells while multiple CTA-derived MAPs (0.4 - 0.7% of the upregulated MAPs) were
presented at greater levels by AZA. Because a GO-term analysis of the upregulated
protein-coding genes evidenced a robust innate immune response in AZA-treated cells,
we conclude that AZA-induced enhanced CTL recognition is more dependent on CTA- than
on ERE-derived MAP presentation and that ERE enhanced expression could rather trigger
the observed innate immune response. An in-depth analysis of the immunopeptidome showed
that MAPs having an altered presentation following AZA treatment derived only partially
from transcripts whose expression was affected by AZA. The analysis of the other AZA-induced
MAPs showed that they resulted preferentially from the activity of the constitutive
proteasome (vs the immunoproteasome) and derived preferentially from proteins having
less aromatic residues (and therefore more prone to misfolding). Because the degradation
of misfolded proteins by the constitutive proteasome can indicate that AZA induces
an unfolded protein response, we have examined whether AZA stimulates autophagy, a
process frequently triggered in association with such stress. Accordingly, flow cytometry
assays on AML cells treated with AZA for 24h evidenced a robust autophagy induction.
Summary/Conclusion: Altogether our results show that AZA promotes the presentation
of CTA-derived rather than ERE-derived MAPs and that autophagy induction could enable
the survival of AML cells to AZA-induced proteotoxic stress. Our findings suggest
that autophagy inhibitors could synergize with AZA in AML therapy.
P401: NEXT-GENERATION FLOW CYTOMETRY TO DETECT MEASURABLE RESIDUAL DISEASE IN ACUTE
MYELOID LEUKEMIA USING A 19-COLOR FULL SPECTRUM FLOW CYTOMETRY PANEL
H. Fokken1,*, N. Kattre1, A. Kloos1, M. Heuser1, T. Kacprowski2,3, M. Tobias4, A.
Schwarzer1,5
1Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover
Medical School, Hannover; 2Division Data Science in Biomedicine, Peter L. Reichertz
Institute for Medical Informatics of TU Braunschweig and Hannover Medical School;
3Braunschweig Integrated Centre of Systems Biology (BRICS), TU Braunschweig, Braunschweig;
4Department of Pediatric Hematology; 5Institute of Experimental Hematology, Hannover
Medical School, Hannover, Germany
Background: The detection of residual leukemic cells (“measurable residual disease”/MRD)
has dramatic prognostic impact on relapse incidence and patient survival in AML and
has become key to guide therapeutic decision making. MRD can be measured by PCR, next-generation
sequencing, and conventional multicolor flow cytometry (Flow-MRD, 8-10 colors), respectively.
Flow-MRD relies on the aberrant expression of surface marker combinations on leukemic
blasts (leukemia-associated immunophenotypes - LAIPs), which are not found on normal
hematopoietic cells. Hence, the challenge of Flow-MRD is to detect and track small
populations of leukemic cells against the complex and heterogenous background of human
bone marrow cells. With the development of next-generation Spectral Flow Cytometers,
it is now possible to stain cells with 20-30 colors at once which would greatly increase
the number of LAIPs that can be tracked and increase sensitivity of Flow-MRD when
the number of available cells is limited. We hypothesized that spectral Flow-MRD allows
for monitoring leukemic disease burden with unprecedented resolution.
Aims: To establish a one-tube full spectrum MRD detection assay and a semi-automated
computational workflow for improved detection of minimal residual disease in AML.
Methods: We established a 19-color full spectrum flow cytometry panel with one tube
containing CD45, CD34, CD117, CD13, CD33, HLA-DR, CD2, CD4, CD5, CD7, CD11b, CD14,
CD15, CD19, CD22, CD38, CD56, CD64 and CD133. Limit of detection and quantification,
linearity, intra-/inter-assay precision, and robustness were determined via spike-in
assays. We measured normal bone marrow of healthy donors and MRD samples from AML
patients. Data were analyzed with GemStone using its unique highly automated approach
to characterizing normal cells in bone marrow based on a template model designed specifically
for data generated with this panel. Results were then explored with the Cen-Se dimensionality
reduction algorithm (Bagwell et al. J Biom Biostat (2019)) and subsequently compared
to expert gating of data generated on the same samples using a well-established 5-Tube
conventional flow cytometry panel.
Results: We defined reference values for known leukemia-associated immunophenotypes
using normal bone marrow. Expert analysis of data simultaneously analyzed with the
full-spectrum panel and the 5-tube conventional flow cytometry panel yielded highly
correlating results for the proportions of granulocytes, lymphocytes, monocytes, and
immature blasts in measured bone marrow with r2 values greater than 0.90. In addition,
quantitative and qualitative MRD results correlated very well when comparing semi-automated
analysis of our full-spectrum panel using GemStone and expert analysis of the 5-tube
conventional flow cytometry panel.
Summary/Conclusion: We have developed an integrated pipeline for full-spectrum flow
cytometry that improves the applicability and performance of flow-cytometry-based
MRD detection in AML.
P402: MUTATIONS IN BONA FIDE ONCOGENES KRAS AND DNMT3A ARE NOT RELATED TO DEPENDENCY
IN ESTABLISHED TUMORS, IN PDX MODELS OF ACUTE LEUKEMIA IN VIVO
Y. Gao1,*, M. Ghalandary1, M. Becker1, D. Amend1, M. Rothenberg-Thurley2, K. Metzeler2,3,
I. Jeremias1,4,5
1Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, German
Research Center for Environmental Health (HMGU); 2Laboratory for Leukemia Diagnostics,
Department of Medicine III, University Hospital, Ludwig Maximilians University (LMU),
Munich; 3Department of Hematology and Cell Therapy, University Hospital Leipzig, Leipzig;
4German Cancer Consortium (DKTK), Partner Site; 5Department of Pediatrics, University
Hospital, Ludwig Maximilians University (LMU), Munich, Germany
Background: The role of oncogenic mutations during the process of tumor formation
was intensively studied in genetically engineered mouse models, while much less is
known about their function in established tumors of patients.
Aims: To bridge the gap, we studied frequently mutated genes for their dependency
in established tumors in vivo, using genetic engineering in patient-derived xenograft
(PDX) models.
Methods: Primary tumor cells from seven patients with acute myeloid leukemia (AML)
were transplanted into immunocompromised NSG mice. PDX cells were lentivirally transduced
to express the Cas9 protein and a sgRNA. The customized sgRNA library was designed
and cloned using our CLUE platform (www.crispr-clue.de; Becker et al., Nucleic Acids
Res. 2020). PDX cells transgenic for Cas9 and the CRISPR/Cas9 sgRNA library were transplanted
into NSG mice, grown until advanced AML disease and sgRNA distribution measured by
next-generation sequencing. From dropout hits in PDX in vivo screens, single molecules
were validated by fluorochrome-guided competitive in vivo experiments, comparing cells
with and without knockout in the same mouse and determining cell distributions by
flow cytometry gating on the different recombinant fluorochromes. Human AML cell lines
were studied in vitro for comparison.
Results: We cloned a small customized sgRNA library targeting 34 genes recurrently
mutated in AML and tested the library in two PDX AML models in vivo. In hit validation
experiments, knockout of NPM1 abrogated in vivo growth in all PDX AML models tested,
reproducing the known common essential function of NPM1. KRAS proved an essential
function in PDX AML models both with and without an oncogenic mutation in KRAS, suggesting
that patients suffering tumors both with and without KRAS mutation might benefit from
treatment inhibiting KRAS. DNMT3A belongs to the genes most frequently mutated in
AML, but most AML cell lines tested in vitro did not depend on DNMT3A and knockout
of DNMT3A did not reduce, but rather increased AML cell line growth in vitro. In contrast,
in PDX models in vivo, we found a clear essential function for DNMT3A in certain AML
samples and PDX in vivo results were discordant to cell line in vitro data. These
data highlight the complementary use of PDX models to study gene dependencies. To
our surprise, the dependency of AML cells on DNMT3A was unrelated to the presence
or absence of a hot spot mutation in DNMT3A.
Summary/Conclusion: We conclude that both KRAS and DNMT3A harbor an essential function
in certain patients established AML tumors in vivo, although independently from the
presence of absence of a mutation in the gene. Warranting verification in additional
patient samples, oncogenes and tumor entities, our data might indicate re-consider
basic principles of decision making in Molecular Tumor Boards.
P403: PROGNOSTIC SIGNIFICANCE OF LONG NON-CODING RNA GAS5 AND MICRORNA-222 EXPRESSION
PROFILES IN YOUNGER AML PATIENTS
Ð. Pavlović1, N. Tošić1, B. Zukić1, I. Marjanović1, Z. Pravdić2, N. Suvajdžić Vuković2,3,
S. Pavlović1, V. Gašić1,*
1Institute of Molecular Genetics and Genetic Engineering; 2Clinic of Hematology; 3Faculty
of Medicine, Belgrade, Serbia
Background: Acute myeloid leukemia (AML) is a heterogeneous malignant disease, that
accounts for 80% of all acute leukemias in adults. The treatment protocol and prognosis
for AML are based on the classification of the patients into risk groups based on
the pretreatment karyotype analysis. However, nearly half of the patients don’t have
any detectable cytogenetic changes. Therefore, there is a constant need to find new
prognostic markers, as well as markers that can be used as targets for therapeutics
that could improve the treatment. Recently, using high-throughput technologies, the
search for new biomarkers has pointed towards non-coding RNAs, especially long non-coding
RNAs (lncRNAs) and micro RNAs (miRNAs). Numerous studies have shown lncRNA GAS5 expression
level dysregulation in multiple cancers, but it was poorly investigated in AML. Additionally,
miR-222 expression levels have shown significant influence on cancers in general,
including AML. Since GAS5 acts like a molecular sponge for miR-222, co-expression
profiles of these non-coding RNAs could be novel prognostic markers in AML.
Aims: We have investigated the expression level pattern of lncRNA GAS5 in younger
AML patients and examined its potential influence on disease outcome. GAS5 expression
levels were also investigated in AML patients with normal karyotype, while taking
into account miR-222 expression levels, in order to examine their potential role as
dual biomarkers.
Methods:: 94 bone marrow samples of younger (<65 years) AML patients have been collected.
Diagnostics and stratification were based on cytomorphology, immunophenotyping and
cytogenetic analysis. All patients received the same induction chemotherapy. Along
with them, 14 healthy controls were analysed. The AML patient group with normal karyotype
(AML-NK) had 39 patients. GAS5 and miR-222 expression levels were analysed using Real-Time
PCR. Relative quantification was performed using the ddCt method, respective healthy
controls were used as the calibrator for each non-coding RNA. The ROC curve analyses
found the optimal cut-off value to discriminate between Gas5high and GAS5low. While
the cut-off value for dividing miR-222 expression levels into miR-222high and miR-222low
was performed using the value of the median level of expression among healthy controls.
Results: Our results showed that GAS5 expression level in AML patients was lower compared
to healthy controls (p < 0.01). Lower GAS5 expression on diagnosis was related to
an adverse prognosis, since higher levels of lactate dehydrogenase was detected in
the GAS5low group compared to the GAS5high group (p < 0.01). The disease-free survival
(DFS) and the overall survival (OS) were significantly lower in the GAS5low group,
compared to the GAS5high group, but survival analysis failed to confirm this finding.
In the AML-NK group patients had higher expression of miR-222 compared to healthy
controls (p < 0.01). A synergistic effect of GAS5low/miR-222high status on disease
prognosis was not established.
Summary/Conclusion: This is the first study focused on examining the lncRNA GAS5 and
miR-222 expression pattern in AML patients. Its initial findings indicate potential
prognostic significance of GAS5 expression and the need for further investigation
of these two non-coding RNAs, their potential roles in leukemogenesis, and the prognosis
of AML patients.
P404: PROGNOSTIC IMPACT OF CEBPA-MUTATIONAL SUBGROUPS IN ADULT AML – RESULTS OF A
LARGE METAANALYSIS IN MORE THAN 1000 CEBPA-MUTANT PATIENTS
J.-A. Georgi1,*, S. Stasik1, M. Kramer2, C. Röllig1, T. Haferlach3, P. Valk4, D. Linch5,
T. Herold6, N. Duployez7, F. Taube1, U. Platzbecker8, H. Serve9, C. Baldus10, C. Müller-Tidow11,
C. Haferlach3, M. Meggendorfer3, S. Koch3, E. L. Boertjes4, R. K. Hills12, A. Burnett13,
G. Ehninger2, K. Metzeler8, M. Rothenberg-Thurley6, A. Dufour6, H. Dombret14, C. Pautas15,
L. Fenwarth7, M. Bornhäuser1, R. Gale5, C. Thiede1,16
1Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden; 2AvenCell Europe
GmbH, Dresden; 3MLL Münchner Leukämielabor GmbH, München, Germany; 4Erasmus University
Medical Center, Rotterdam, Netherlands; 5Department of Haematology, UCL Cancer Institute,
London, United Kingdom; 6Labor für Leukämiediagnostik, Medizinische Klinik und Poliklinik
III, Universitätsklinikum München, München, Germany; 7Institut de Recherche contre
le Cancer de Lille, Centre Hospitalier Universitaire de Lille, Lille, France; 8Klinik
und Poliklinik für Hämatologie, Zelltherapie und Hämostaseologie, Universitätsklinikum
Leipzig, Leipzig; 9Medizinische Klinik 2, Universitätsklinikum Frankfurt, Frankfurt
am Main; 10Klinik für Innere Medizin II, Universitätsklinikum Schleswig-Holstein,
Kiel; 11Klinik für Hämatologie, Onkologie und Rheumatologie, Universitätsklinikum
Heidelberg, Heidelberg, Germany; 12Nuffield Department of Population Health, Oxford
University, Oxford; 13Department of Haematology, Cardiff University, University Hospital
of Wales, Cardiff, United Kingdom; 14Institut de Recherche Saint-Louis, Université
de Paris, Hôpital Saint-Louis, Paris; 15Service d’Hématologie Clinique, Hôpital Henri
Mondor, Créteil, France; 16AgenDix GmbH, Dresden, Germany
Background:
CEBPA double mutations (dm) are included in the current AML WHO-classification as
a favorable-risk independent entity, but recent data suggest this might be limited
to those approximately 90% of patients (pts) with in-frame mutations in the basic
leucine zipper (bZIP) domain, and that bZIP single allele mutations (sm) might also
be favorable.
Aims: To investigate further, we performed a meta-analysis involving detailed sequencing
data from 1010 CEBPA-mutant pts.
Methods: Primary pt data was obtained from six study groups: 98 from ALFA, 104 AMLCG,
191 HOVON, 200 MLL, 240 SAL, 177 UK(MRC/NCRI). All pts were treated with curative
intention using age and risk-adapted strategies. CEBPA sequences were classified by
localization (bZIP vs transactivation domains [TAD] 1 + 2), allelic status (sm vs
dm), and type of mutation (in-frame [if] or frameshift [fs] insertions/deletions [InDel]
or missense [ms]). Data for GATA2, WT1, FLT3, NPM1 co-mutations were available for
most pts.
Results: Overall, 546 (54%) had dm and 464 (46%) sm CEBPA (289 TAD1/2; 175 bZIP).
To study the impact of the different mutations, we generated 8 groups: dmCEBPA pts
with 1/more bZIPInDel-if (Gr1), bZIPInDel-fs (Gr2) or bZIPms (Gr3) mutation or 2 TAD
mutations (Gr4), as well as the corresponding sm groups (Gr5-8). Of note, dm/sm pts
with bZIPInDel-if mutations (Gr1 + 5) were significantly younger and predominantly
had de novo AML (Tab.1) whereas Gr2-4 and Gr6-8 pts were older with higher incidence
of sAML. GATA2 and WT1 mutations were more common in Gr1 + 5 pts; mutations in NPM1,
spliceosome (SF3B1, U2AF1, SRSF2 and ZRSR2) and DNA-methylation (ASXL1, DNMT3A, TET2,
IDH1/2) associated genes were confined to the other groups. Pts in Gr1 showed the
highest rate of CR1 (Tab.1), better RFS and OS (Fig. 1), whereas dmCEBPA pts in Gr2-4
did significantly worse. Outcome of Gr5 pts was less favorable than Gr1, but still
better than for the other subgroups (Gr2-4 and 6-8). In a combined analysis of Gr2-4
and Gr6-8, prognostic factors according to ELN2017 identified three clearly separated
subgroups, with the favorable subgroup predominantly associated with mut. NPM1; Fig.
1B). The prognostic impact of Gr1 for CR, OS and RFS was confirmed in multivariable
analyses taking into account the individual study groups.
dmCEB
P
AbZIPInDel-fs
dmCEBPAbZIPInDel-if n=425Gr1
dmCEBPAbZIPInDel-fs n=26Gr2
dmCEBPAXbZIPmsnn=35Gr3
dmCEBPANNTADNNNn=60Gr4
smCEBPAbZIPInDel-if n=66Gr5
smCEBPAbZIPInDel-fs n=55Gr6
smCEBPAXbZIPmsnn=54Gr7
smCEBPANNTADN.Nn=289Gr8
p.value BPANDN289
Median age, years
42
60
57
64
47
59
52
58
<.001
AML type, %
<.001
de novo
98
81
94
88
92
96
90
90
sAML/tAML
2
20
6
13
8
4
10
10
FLT3-ITD
mut
,.%
11
8
9
7
8
29
9
32
<.001
NPM1
mut
, %
1
12
9
13
3
29
15
44
<.001
GATA2
mut
, %
39
8
12
9
33
10
12
4
<.001
WT1
mut
, %
20
4
15
3
13
4
6
8
<.001
Spliceosome mut., %
2
29
6
40
5
39
25
25
<.001
Methylation mut., %
25
83
69
71
35
74
46
63
<.001
CR1, %
94.3
73.9
78.1
73.1
92.1
77.6
79.6
73.5
<.001
Median RFS, months
152
9
15
16
64
9
16
22
<.001
Median OS, months
215
16
40
22
126
70
30
41
<.001
Image:
Summary/Conclusion: The results of this large metaanalysis confirm our previous results
that only dmCEBPA with InDel-if mutations show a specific biology and very favorable
prognostic implications, and further supports the notion that the subgroups with other
dm pts differ substantially and have a significantly worse outcome. SmCEBPA bZIPInDel-if
mutant pts and those with other CEBPA mutations
and
favorable ELN2017 genotype also show an improved outcome.
P405: INHIBITION OF CKS1-DEPENDENT PROTEOSTASIS REVEALS VULNERABILITIES IN LEUKAEMIC
STEM CELLS WITH CONCOMITANT PROTECTION OF HEALTHY HAEMATOPOIETIC STEM CELLS
W. Grey1,2,*, A. Rio-Machin3, P. Casado-Izquierdo3, E. Gronroos2, S. Ali2, J. Miettinen4,
F. Bewicke-Copley3, A. Parsons4, C. Heckman4, C. Swanton2, P. Cutillas3, J. Gribben3,
J. Fitzgibbon3, D. Bonnet2
1York Biomedical Research Institute, The University of York, York; 2The Francis Crick
Institute; 3Bart’s Cancer Institute, London, United Kingdom; 4HiLIFE, Helsinki, Finland
Background: Acute myeloid leukemia (AML) is an aggressive hematological disorder comprising
a hierarchy of quiescent leukemic stem cells (LSCs) and proliferating blasts with
limited self-renewal ability. AML has a dismal prognosis, with extremely low two-year
survival rates in the poorest cytogenetic risk patients, primarily due to the failure
of intensive chemotherapy protocols to deplete LSCs, and the significant toxicity
towards healthy hematopoietic cells. Whilst much work has been done to identify genetic
and epigenetic vulnerabilities in AML LSCs, little is known about protein homeostasis
– so called “Proteostasis” – in drug resistance and relapse.
Aims:
- Characterisation of proteostatic targeting in AML and haematopoiesis.
- Identify proteostatic vulnerabilities in LSCs.
Methods: This study used a range of primary patient AML samples, highthroughput drug
screening, in vivo transplantation models and proteomics to demonstrate mechanism
of action and functionality of a small molecule inhibitor of CKS1-dependent protein
degradation.
Results: We tested a range of proteostatic targeting agents against primary, poor
risk, AML patient samples and demonstrate a selective vulnerability of both AML blasts
and LSCs to inhibition of CKS1-dependent protein degradation in vitro and in vivo
using a small molecule inhibitor. Mechanistically, inhibition of CKS1 leads to hyperactivation
of RAC1, increased NADPH production and critical levels of intracellular ROS, resulting
in death of LSCs.
Conversely, CKS1 inhibition has the opposite effect on healthy haematopoiesis. Healthy
haematopoietic stem and progenitor cells (HSPCs) display increased quiescence upon
inhibition of CKS1, a phenotype which confers chemoprotection of healthy HSPCs during
clinical chemotherapy protocols (Cytarabine + Doxorubicin, 5 + 3). A phenotype conserved
by intestinal stem and progenitor cells.
Summary/Conclusion: Together these findings demonstrate a key vulnerability of AML-LSCs
to CKS1-inhibition, while it preserves healthy stem cells. This offers the prospect
of both depleting LSCs in vivo and bringing back clinically unfit patients into the
pool for intensive chemotherapeutic treatment.
P406: ENHANCED SIGNIFICANCE OF FLT3-ITD RESIDUAL DISEASE DETECTION ON TREATMENT OUTCOME
IN ACUTE MYELOID LEUKEMIA
T. Grob1,*, M. Sanders1, C. Vonk1, F. Kavelaars1, M. Rijken1, D. Hanekamp1, P. Gradowska1,
J. Cloos2, Y. Fløisand3, M. V. Marwijk Kooy4, M. Manz5, G. Ossenkoppele2, L. Tick6,
V. Havelange7, B. Löwenberg1, M. Jongen-Lavrencic1, P. Valk1
1Hematology, Erasmus Medical Center, Rotterdam; 2Hematology, Amsterdam University
Medical Center, Amsterdam, Netherlands; 3Hematology, Oslo University Hospital, Oslo,
Norway; 4Hematology, Isala Hospital, Zwolle, Netherlands; 5Hematology, University
Hospital Zurich, Zurich, Switzerland; 6Hematology, Maxima Medisch Centrum, Eindhoven,
Netherlands; 7Hematology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
Background: The prognostic significance of FLT3-internal tandem duplications (FLT3-ITD)
in acute myeloid leukemia (AML) in relation to the allelic mutational burden and other
concurrent gene mutations at diagnosis remains subject of scientific controversy.
Increasing evidence indicates that treatment outcome prediction can be improved by
assessing the kinetics and depth of response during therapy by detection of minimal
residual disease (MRD). Until now, FLT3-ITD MRD detection by RQ-PCR has been hampered
by the variety in patient specific FLT3-ITD (i.e. sequence, position and length) and
is currently not recommended because of potential instability of FLT3-ITD clones during
relapse. However, systematic studies evaluating the applicability of FLT3-ITD MRD
detection with next-generation sequencing (NGS) in AML are currently lacking.
Aims: Here, we set out to compressively investigate the impact of NGS-based FLT3-ITD
MRD detection on treatment outcome in a cohort of newly diagnosed patients with AML
in the context of current prognostic factors at diagnosis and other MRD measurements
during therapy, including mutant NPM1 and multiparameter flow cytometry (MFC).
Methods: In 176 de novo FLT3-ITD AML patients who were treated in HOVON-SAKK multicenter
prospective phase III clinical trials, NGS was performed at diagnosis and in complete
remission (CR) following two cycles of standard induction chemotherapy. The NGS libraries
were paired-end sequenced (2×221-bp) with custom primers on an Illumina MiSeq according
to manufacturer’s recommendation (Illumina, San Diego, CA). We used our in-house data
analysis pipeline for variant calling. The primary endpoints of the study were relapse
and overall survival. The Cumulative Incidence of Relapse (CIR) was estimated with
competing-risks regression analyses according to the method of Fine & Gray. The Cox
proportional hazard model was used to calculate overall survival estimates.
Results: NGS-based FLT3-ITD MRD was present in 43 of 176 (24%) AML patients with a
median variant allele frequency of 0.01% (range 3.1x10-4% to 3.10%). Presence of FLT3-ITD
MRD was associated with increased risk of relapse (4-year risk of relapse, 79% FLT3-ITD
MRD vs. 34% no FLT3-ITD MRD; P<0.001) and inferior overall survival (4-year rate of
overall survival, 27% FLT3-ITD MRD vs. 57% no FLT3-ITD MRD; P<0.001). In multivariate
analysis, detection of FLT3-ITD MRD in CR confers independent prognostic significance
for relapse (hazard ratio, 4.00; P<0.001) and overall survival (hazard ratio 2.78;
P<0.001). Strikingly, FLT3-ITD MRD exceeds the prognostic value of most generally
accepted clinical and molecular prognostic factors, including FLT3-ITD allelic ratio
at diagnosis and MRD assessment by NGS-based mutant NPM1 detection or MFC.
Image:
Summary/Conclusion: NGS-based detection of FLT3-ITD MRD in CR identifies AML patients
with a profound risk of relapse and death that outcompetes the significance of most
accepted prognostic factors at diagnosis and during therapy, and furnishes support
for FLT3-ITD as a clinically relevant biomarker for dynamic disease risk assessment
in AML.
P407: SYNERGISTIC EFFECT OF A NOVEL COMBINATION OF HDACI CHIDAMIDE WITH CLADRIBINE
ON CELL PROLIFERATION ARREST AND APOPTOSIS IN ACUTE MYELOID LEUKEMIA BY TARGETING
MYC/HDAC2 SIGNALING
S. Gu1, C. Song2,3, Z. Ge1,*
1Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University,
Institute of Hematology Southeast University, Nanjing, China; 2Hershey Medical Center,
Pennsylvania State University Medical College, Hershey; 3Division of Hematology, The
Ohio State University Wexner Medical Center, the James Cancer Hospital, Columbus,
American Samoa
Background: Chidamide (CHI), a novel Histone deacetylase inhibitor (HDACi), which
is now involved in the salvage treatment of relapsed/refractory acute myeloid leukemia
in a clinical trial. Cladribine (2-chloro-2’-deoxyadenosine (2-CdA)), is a purine
nucleoside antimetabolite analog, which resists degradation by adenosine deaminase
and therefore accumulates to cytotoxic levels. The anti-leukemia effects of either
single drug have been reported by inducing cell apoptosis through suppressing multi-oncogenic
pathways in human tumor cells. However, whether and how the combination of the two
drugs exerts the synergistic anti-tumor effects are still undetermined in AML.
Aims: This study is to investigate the synergistic therapeutic effects of 2-CdA in
combination with CHI in AML.
Methods: Cell Counting Kit-8 (CCK-8) cell proliferation assay, Annexin V apoptosis
assay, Propidium Iodide cell cycle assay, and Western Blot (WB) were performed in
U937 AML cells in presence of CHI only, 2-CdA only, CHI+ 2-CdA, and vehicle control
for 48h. RNA-seq was performed in U937 cells treated with CHI and 2-CdA; the differentially
expressed genes (DEGs) were identified by R studio, and pathway enrichment of the
DEGs was analyzed with Metascape online tool. Co-Immunoprecipitation (co-IP) was used
to examine the interaction of cMYC with HDAC1/2. Data were expressed as the mean ±
standard deviation. T-tests statistical analysis was employed for determining the
significant difference between groups. A synergistic effect was determined by the
combination indices (CIs) with CompuSyn software.
Results: The 50% inhibiting concentration (IC50) of CHI and 2-CdA on U937 cell proliferation
is 9.358±1.448μmol/L and 0.024±3.071μmol/L, respectively. The combination of various
doses of CHI with either IC50 or 1/2 IC50 of 2-CdA significantly decreases the survival
rate of U937 cells compared to single drug control (Fig.1a). The CI analysis showed
the high synergistic therapeutic effects of CHI with ether dose of 2-CdA (Fig. 1b).
The combination of (8mM) CHI with (0.02mM) 2-CdA ignificantly elevated the apoptosis
rate and induced the G0/G1 cell phase arrest of AML cells compared with either single
drug alone (p<0.0001, Fig. 1c, d). Moreover, the expression of apoptotic markers,
caspase-3, PARP-1, and caspase-9 is elevated, and the cell cycle marker p21 proteins
were significantly upregulated, but Cyclin-dependent kinase CDK2 and Cyclin E2 down-regulated
in the combination group compared to either single drug control (p<0.05, Fig. 1e).
To further understand the underlying mechanism of the synergy, RNA-seq analysis was
performed in U937 cells treated with either CHI or 2-CdA, respectively. The overlapped
altered DEGs were identified, and cMYC is dramatically down-regulated. The pathway
analysis of the altered DEGs showed the enrichment of MYC pathways further indicated
the critical role of MYC (Fig. 1f). It is reported that cMYC is associated with the
histone deacetylase complex. Indeed, the co-IP assay showed an interaction of MYC
with HDAC2, not HDAC1 in U937 (Fig. 1g). These data suggested the role of cMYC/HDAC2
signaling in the synergistic effect of the combination of CHI with 2-CdA in the disease.
Image:
Summary/Conclusion: The combination of Chidamide and Cladribine has a synergistic
effect on cell proliferation arrest, apoptosis, and cell cycle arrest in AML cells
through MYC/HDAC2 pathway. Our data provide experimental evidence for the novel potential
combination of AML therapy. More in vivo pre-clinical studies will be explored for
future clinical trials.
P408: NRAS MUTATIONS ARE INVOLVED IN ACUTE MYELOID LEUKAEMIA DRUG RESISTANCE
F. Healy1,*, V. Marensi1, J. Woolley1, D. MacEwan1
1Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and
Integrative Biology, University of Liverpool, Liverpool, United Kingdom
Background: Acute Myeloid Leukaemia is the most common adult leukaemia, typically
associated with poor prognosis. While rates of response to initial cytarabine-based
induction chemotherapy are high, the majority of patients will relapse even with haematopoietic
stem cell transplant. Thus, the addition of targeted therapies to induction regimens
are becoming more widespread, particularly the use of midostaurin and gilteritinib.
Even so, resistance is emerging to FLT3 inhibitors, and can often be attributed to
NRAS mutations.
Aims: Here, we use over-expression and CRISPR-Cas9 to assess the involvement of NRAS
mutations in FLT3-inhibitor resistance cell line models (MOLM-13, FLT3-ITD heterozygous
and MV4-11, FLT3-ITD homozygous) that had been previously generated in our lab.
Methods: Drug sensitivity was determined by quantification of apoptosis 72 hours post-treatment
(Annexin-V/Propidium Iodide staining). NRAS mutational hotspots associated with increased
activation and drug resistance were analysed by Sanger Sequencing. NRAS over-expression
and knock-out cell lines were generated using either transfection (HEK293T) or lentiviral
transduction (MOLM-13 and MV4-11). Signalling dynamics driving resistance were determined
by immunoblotting.
Results: Mutations arose in the MOLM-13-resistant and MV4-11 resistant cell lines
at common NRAS mutational hotspots (Q61 and G12, respectively). NRAS Q61K over-expression
in the parental MOLM-13 cell line decreased sensitivity to the FLT3 inhibitors gilteritinib,
quizartinib, as well as cytarabine, relative to the parental cell line. However, they
remained more sensitive to FLT3 inhibition and cytarabine than the resistant cells.
In contrast, NRAS-Q61K over-expression caused ~3-fold decrease in sensitivity to trametinib,
the MEK inhibitor that acts downstream of NRAS, relative to both the MOLM-13 and MOLM-13-resistant
cell lines (Table 1). MOLM-13-NRAS-Q61K over-expression cells showed a growth advantage
compared to parental cells in normal culture. In MV4-11-NRAS-Q61K cells, preliminary
mechanistic analysis has shown a decrease in ERK phosphorylation. Conversely, CRISPR-Cas9-mediated
NRAS knockout in HEK293T appeared to increase ERK activity.
Image:
Summary/Conclusion:
NRAS mutations contribute to a FLT3-inhibitor-resistant phenotype, confer a growth
advantage in FLT3-ITD+ AML and may act independently of ERK. We have engineered the
MOLM-13-resistant and MV4-11-resistant cell lines to express Cas9 under a doxycycline-inducible
promoter, to permit stable reversion of the respective mutations. Future work will
use these cells to create a CRISPR-mediated reversion of the NRAS mutations which
have arisen in the resistant cell lines, to better understand the role of NRAS mutations
in FLT3-ITD+ AML. We will better characterise the impacts of a host of NRAS mutations
within AML using over-expression and knockout cell lines, and assess drug sensitivity
and subsequent pathway re-wiring.
P409: NPM1 MUTATED SAMPLES ARE ESPECIALLY VULNERABLE WHEN TARGETING RAC1 IN PATIENT-DERIVED
AML CELLS IN VITRO
A. Hemsing1,2,*, K. P. Rye2, K. J. Hatfield3, H. Reikvam1,2
1Department of Medicine, Haukeland University Hospital; 2Department of Clinical Science,
University of Bergen; 3Department of Immunology and Transfusion Medicine, Haukeland
University Hospital, Bergen, Norway
Background: Ras-related C3 botulinum toxin substrate 1 (Rac1) is a GTPase signaling
molecule known to be overexpressed in many solid cancers and leukemia. Rac1 plays
a part in several pathways regulating, e.g., cell migration, adhesion, and cell proliferation.
Recent studies have revealed activation and widespread implication of the Rac1 molecule
in acute myeloid leukemia (AML), and in vitro Rac1 inhibition in AML cell lines induces
apoptosis and improves chemosensitivity.
Aims: We studied the in vitro effect on proliferation, apoptosis, and cytokine release
of five RAC1 inhibitors in a cohort of heterogeneous, consecutive patient-derived
AML cells at diagnosis. We aimed to map the antileukemic effect in a relatively large
AML cohort to identify potential subgroups of AML patients that could be particularly
vulnerable to RAC1 inhibition.
Methods: 79 patient-derived AML cell samples from diagnosis were thawed after preservation
in liquid nitrogen and cultured at standard in vitro methods with the Rac1-inhibitors
ZINC69391, ITX3, EHOP-16, 1A-116, and NSC23766. Cell proliferation was measured by
the 3H-thymidine incorporation assay, and apoptosis was measured by the Annexin V/Propidium
Iodine apoptosis assay for flow cytometry. Assessment of cytokine profiles in culture
media was done with Luminex multiplex immunoassay.
Results: Measured IC50 values for each compound were as follows; ZINC69391 23 µM (95%
CI 15 -29 µM), ITX3 24 µM (15 – 40µM), EHOP-16 3 µM (1,8 – 3,9 µM), 1A-116 10 µM.
(8-11 µM) and NSC23766 12 µM (9-15 µM). All five Rac1 inhibitors resulted in an overall
antiproliferative effect, tested at dosage around IC80. Using bioinformatical classification,
we observed that high versus intermediate/low antiproliferative effect was more common
in NPM1 mutated (Fischer’s exact test, p=0.002) and CD34 negative (p=0.008) patient
samples. The differences were insignificant regarding the presence of FLT3 mutation
or according to cytogenetics, FAB classification, gender, and age.
All five compounds did induce significant apoptosis and necrosis compared to untreated
control by Wilcoxon signed-rank test (p< 0.0001) at dosages around IC40 and IC80.
For NSC23766 only, the presence of NPM1 mutation was associated with more severe apoptosis
and necrosis (p=0.01). The presence of FLT3 mutation or CD34 negativity or positivity
did not influence the depth of apoptosis and necrosis.
Presence of NPM1 mutation was associated with reduced viability after treatment with
EHOP-16 (p=0.025), ITX3 (p=0.047) and NSC23766 (p=0.005). CD34 negativity was significantly
associated with reduced viability after treatment with NSC23766 (p=0.006). Finally,
Rac1 inhibition significantly reduced the cytokine release of several cytokines crucial
for leukemogenesis, with the strongest effects observed for Rac1 inhibitors 1A-116
and NSC23766 (Figure 1, Euclidean clustering).
Image:
Summary/Conclusion: Our results suggest potent and significant antiproliferative and
antiapoptotic effects of Rac1 inhibition in primary AML samples. Interestingly, patients
harboring the NPM1 mutation seem significantly more vulnerable to such inhibition.
Our findings support further research regarding the role of Rac1 and its pharmacological
inhibition in AML.
P410: LPIN1 REGULATES LIPID HOMEOSTASIS AND MAINTAINS STEM CELL ENRICHED POPULATIONS
IN AML
K. Huber1,2,*, S. Garg3, L. He1,4, A. Pouya1, C. Rohde1,4, C. Lüchtenborg5, C. Arnold4,6,
J. Zaugg4,6, S. Raffel1, C. Müller-Tidow1,4, B. Brügger5, C. Pabst1,4
1Medizinische Klinik V für Hämatologie, Onkologie und Rheumatologie, Universitätsklinikum
Heidelberg, Heidelberg; 2Medizinische Klinik II für Hämatologie und Onkologie, Universitätsklinikum
Schleswig-Holstein, Kiel, Germany; 3Dana-Farber Cancer Institute, Boston, United States
of America; 4Molecular Medicine Partnership Unit (MMPU), University of Heidelberg
and European Molecular Biology Laboratory (EMBL); 5Heidelberg University Biochemistry
Center (BZH); 6European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
Background: Leukemic stem cells (LSCs) often escape chemotherapy treatment due to
the acquisition of stem cell like properties such as slow cell cycle progression,
which makes them main drivers of cytologic relapse in acute myeloid leukemia (AML).
Identification of novel therapeutic targets to specifically eradicate residual chemoresistant
AML cells therefore represents a key challenge in AML therapy. Since LSCs show altered
metabolic features, which include dependency on low ROS levels and oxidative phosphorylation
of amino acids, targeting LSC metabolism has evolved as a promising approach. Recently
the phospholipid phosphatidylserine was shown to be required for LSC stemness and
differentiation. However, the role of the main phospholipid synthesis pathway in which
the phosphatidic acid phosphatase (PAP) lipin1 catalyzes a rate limiting step remains
largely unexplored.
Aims: We investigated the role of the phosphatidic acid phosphatase LPIN1 in AML and
healthy hematopoietic cells.
Methods: We applied a gene depletion approach using shRNAs and CRISPR/Cas9 on AML
cell lines, primary AML samples and cord blood derived CD34+ hematopoietic stem and
progenitor cells (HSPCs) to monitor the effects of LPIN1 depletion on proliferation,
differentiation, colony forming capacity and bone marrow engraftment in mice. To gain
insights into the mechanisms of action, we combined RNA-sequencing (RNA-seq) with
lipidomics assays.
Results: Here we show that regulation of lipid homeostasis by lipin1 is required for
HSPC and LSC function. RNA-seq data analysis of healthy hematopoietic cells revealed
highest LPIN1 expression in the immature CD34 positive compartment, which steadily
decreases during myeloid differentiation. In normal karyotype (NK) AML LPIN1 was utmost
expressed in an AML subpopulation characterized by high LSC frequency and co-positivity
for the LSC markers CD34 and GPR56. Knock down (KD) of LPIN1 in CD34 positive HSPCs
and patient derived xenograft (PDX) leukemic cells hampered proliferation and reduced
the CD34 positive LSC compartment in vitro. In xenotransplantation assays LPIN1 KD
reduced bone marrow engraftment of healthy HSPC and human AML cells. Furthermore,
in vivo differentiation of CD34 positive cells into B-cells was almost completely
abrogated upon LPIN1 KD. In the leukemia setting, LPIN1 KD particularly reduced the
LSC-enriched CD34+GPR56+ co-positive fraction. While RNA-seq of the AML cell line
OCI-AML3 and CD34 positive cells showed only subtle gene expression changes in phospholipid
metabolism upon LPIN1 KD, lipidomics analyses revealed significant differences in
the composition of phospholipids upon LPIN1 KD.
Summary/Conclusion: Our data establish LPIN1 as an essential enzyme maintaining lipid
homeostasis and stem cell functions in the hematopoietic system. Identification of
phospholipid entities with differential impact on healthy versus leukemic cells might
help refine LSC targeted therapies.
P411: SF3B1 MUTATIONS IN AML ARE STRONGLY ASSOCIATED WITH MECOM REARRANGEMENTS AND
MAY BE INDICATIVE OF AN MDS PRE-PHASE
S. Huber1,*, S. Hutter1, M. Meggendorfer1, G. Hoermann1, W. Walter1, C. Baer1, W.
Kern1, T. Haferlach1, C. Haferlach1
1MLL Münchner Leukämie Labor GmbH, München, Germany
Background: Mutations in the splicing factor gene SF3B1 are most frequent in myelodysplastic
syndrome (MDS) or myelodysplastic/myeloproliferative neoplasm (MDS/MPN) but are also
recurrently found in acute myeloid leukemia (AML). Splicing factor mutations have
been suggested to have additive value if incorporated into the current risk classification
of AML (van der Werf et al., Blood Advances 2021).
Aims: Explore the genetic landscape of SF3B1 mutated AML and determine the prognostic
impact of SF3B1 mutations.
Methods: We analyzed 735 patients diagnosed with AML based on cytomorphology, cytogenetics,
and molecular genetics according to WHO classification 2017. Reads from amplification-free
whole genome sequencing (WGS, median coverage 103x, Illumina, San Diego, CA) were
aligned to the human reference genome (GRCh37, Ensembl annotation, Isaac aligner)
and variants were called using Strelka Somatic Variant Caller v2.4.7.
Results: We identified SF3B1 mutations in 41 of 735 (6%) AML patients. SF3B1 mutations
were found in AML with recurrent genetic abnormalities (24/471; 5%), AML with myelodysplasia-related
changes (MRC; 11/158; 7%) and AML not otherwise specified (NOS; 6/106; 6%). When focussing
on SF3B1
mut cases, AML-MRC (11/41; 27%) and AML with GATA2::MECOM (10/41; 24%) were most frequent.
9 additional cases showed another MECOM rearrangement (-rear) resulting in 46% (19/41)
of patients with SF3B1 mutations accompanied by a MECOM-rear. A prior history of MDS
or MDS/MPN was documented in 20% (8/41) of SF3B1
mut patients. Thereof, 63% (5/8) harbored a MECOM rearrangement and 25% RUNX1
mut. On average, SF3B1
mut patients harbored 2.85 mutations (including SF3B1). The most frequent additional
mutations of SF3B1
mut patients were RUNX1 (9/41; 22%) and NRAS (8/41; 20%), and NPM1, TET2, or DNMT3A
mutations or FLT3-ITD were detected in 15% (6/41) each. SF3B1 showed an average variant
allelic frequency (VAF) of 41%. Notably, the VAF of SF3B1 in AML with NPM1 or RUNX1
was higher than the VAFs of NPM1 or RUNX1 in 9/11 (82%) cases, suggesting that SF3B1
preceded these mutations. In 16/41 (39%) cases molecular follow-up data was available.
The SF3B1 mutation persisted during the entire disease course in 8 cases (including
1 with relapse). In particular, in two of these AML patients with mutated NPM1, the
SF3B1 mutation remained detectable by NGS, despite complete hematologic remission
and undetectable NPM1 mutation by high-sensitive qPCR. In all 5 cases with relapse
the SF3B1 mutation re-occurred at relapse. In 4 patients, the VAF of SF3B1 decreased,
similar to accompanying mutations. In contrast to MDS, SF3B1 mutations did not affect
the overall survival (median OS: 16.9 months in SF3B1
mut; 5.7 months in unmutated group; p=0.822) in the total AML cohort. When stratified
for AML sub-entities, the prognosis of the SF3B1
mut AML seems to be dominated by the sub-entity.
Image:
Summary/Conclusion: Within AML, SF3B1 mutations are enriched in AML patients with
GATA2::MECOM and AML-MRC. This association with poor risk subtypes dominates the prognosis
of SF3B1
mut AML. The high VAF of SF3B1 mutations and the persistence of SF3B1 mutations in
AML patients in complete remission suggest the early acquisition of SF3B1 mutations
in a pre-leukemic clone in a number of patients. While an antecedent MDS or MDS/MPN
has been documented in some cases of SF3B1
mut AML, it might be unidentifiable in others. Alternatively, SF3B1
mut clonal hematopoiesis of indeterminate potential (CHIP) may represent a relevant
pre-phase of SF3B1
mut AML.
P412: LOW EXPOSURE, EXTENDED DOSING MIMICKING CLINICAL EXPOSURES OF ORAL AZACITIDINE
SYNERGIZES WITH VENETOCLAX IN PRECLINICAL AML MODELS
D. Jeyaraju1,*, M. Alapa1, A. Polonskaia1, A. Risueno Perez2, R. Hurren3, X. Wang3,
M. Gronda3, A. Ahsan1, C. Wang1, P. Subramanyam4, J. Sriganesh4, A. Anand4, M. Bysani
Reddy4, K. Ghosh5, C. Kyriakopoulos2, N. Lailler6, C. Hartl6, D. Lopes de Menezes7,
A. Schimmer3, P. Hagner1, A. Gandhi1, A. Thakurta1
1Translational Medicine, Bristol Myers Squibb, Summit, United States of America; 2BMS
Center for Innovation and Translational Research Europe, Bristol Myers Squibb, Seville,
Spain; 3Princess Margaret Cancer Center, Toronto, Canada; 4BBRC; 5Bristol Myers Squibb,
Bangalore, India; 6Rancho Biosciences, San Diego; 7Bristol Myers Squibb, San Francisco,
United States of America
Background: Acute myeloid leukemia (AML) has a high incidence of relapse despite the
advancement of novel therapies. AML patients that are ineligible for intensive chemotherapy
are typically administered injectable azacitidine (AZA) at a dose of 75 mg/m2 for
7 days in combination with BCL-2 inhibitor venetoclax (VEN) as induction therapy.
Injectable-AZA dose was determined primarily through safety, tolerability, and bioavailability
studies based on the CALGB and AZA-001 trials and not through a dose escalation study.
Thus, it is not known if similar efficacy and combinability of AZA with VEN could
be achieved at lower exposures and extended duration with oral-AZA dosing. The approval
of oral-AZA in AML maintenance therapy, albeit at a lower cumulative exposure conferred
at doses of 300 mg for an extended duration of 14 days of a 28-day dosing cycle, creates
opportunities to answer these questions.
Aims: To investigate the mechanism of action and combinability of oral-AZA with VEN
in pre-clinical models.
Methods: For in vitro cell-line based work, 1 day x 1mM AZA and 5days x 0.2mM AZA
were used to mimic injectable-AZA like and oral-AZA like regimens respectively. For
in vivo studies we modeled injectable-AZA and oral-AZA doses with 3mg/kg, qd x 5 days
and 1 mg/kg, qd x 15 days respectively (intraperitonially in both cases, modified
from Vu et al., Nat. Commun. 2020). VEN was administered orally at 100 mg/kg, qd x21
days. CRISPR screen was conducted with OCI-AML2-Cas9 cells using the Cellecta sgRNA
library in the presence of AZA (121nm (IC50) x 5 days).
Results: To identify synthetic lethal targets of oral-AZA, we conducted a genome-wide
CRISPR screen. Among others, BCL-2 emerged as a synergistic hit. During in vitro validation
of the CRISPR screen data, VEN synergized (anti-proliferative effect) with oral-AZA
like exposure only when added after 5 days (0.2mM /day) of repeated AZA dosing. On
the other hand, injectable-AZA like exposure synergized with VEN in 24 hours suggesting
that the Oral-AZA/VEN synergy was driven more through epigenetic changes and not by
stress response as has been reported for injectable-AZA. To further validate the CRISPR
screen findings, we utilized in vivo AML xenograft mouse (injected with human AML
cell lines) treated with an oral-AZA like or injectable-AZA like regimen with/without
VEN. Mouse survival data demonstrated that both AZA regimens combined with VEN with
equivalent efficacy.
The persistence of leukemic stem cells (LSCs) is one of the reasons for relapse in
AML. In vitro, oral-AZA like exposure resulted in decreased numbers of CD34+ CD38-
LSCs. In an in vivo primary AML xenograft model using intrafemoral engraftment, oral-AZA
like exposure was effective in reducing engraftment of leukemic cells from the right
to the left femur (10-fold reduction). Analysis of cells at the site of injection
(right femur) revealed induction of differentiation from a granulocyte-monocyte progenitor
(GMP) to a megakaryocyte-erythrocyte progenitor (MEP) or common myeloid progenitor
(CMP) state in response to oral-AZA.
Summary/Conclusion: Collectively, our pre-clinical in vitro and in vivo data in AML
models indicates that oral-AZA can combine effectively with VEN. In addition, oral-AZA
can effectively eliminate LSCs and induce differentiation of immature cells. Through
the CRISPR screen, our work identifies potential novel combination partners for oral-AZA.
P413: THE POTENTIAL USE OF AN ORAL HYPOMETHYLATING AGENT, OR-2100, AS A COMBINATION
THERAPY FOR MYELOID MALIGNANCIES
K. Kamachi1,2,*, H. Ureshino1,2, K. Kawasoe1,2, N. Yoshida-Sakai1,2, Y. Yamamoto1,
Y. Fukuda-Kurahashi2,3, Y. Kurahashi1,2,3, T. Watanabe2, S. Kimura1,2
1Division of Hematology, Respiratory Medicine and Oncology, Department of Internal
Medicine, Faculty of Medicine; 2Department of Drug Discovery and Biomedical Sciences,
Saga university, Saga; 3OHARA Pharmaceutical Co.,Ltd., Shiga, Japan
Background: The combination therapy of hypomethylating agents (HMAs), azacytidine
(AZA) and decitabine (DAC), with venetoclax (VEN), a selective BCL-2 inhibitor, have
revolutionized the outcome of elderly patients with acute myeloid leukemia (AML),
which have demonstrated the potential effect of HMAs as a combination therapy with
molecular targeting agents for myeloid malignancies. However, there are a few studies
on the combined effect of HMAs with tyrosine kinase inhibitors (TKIs) in chronic myeloid
leukemia (CML) and the mechanism by which HMAs enhance the efficacy of molecular targeting
agents remain poorly understood. We have developed OR-2100 (OR21), an oral HMA, a
5’-O-trialkylsilylated DAC. OR21 is as effective as DAC, but less myelosuppressive,
in xenograft mouse models of solid and hematological malignancies (Hattori et al.
Clin Epigenetics 2019, Watanabe et al. Blood 2020, Ureshino et al. Mol Cancer Ther
2021, Kamachi et al. Cancer Letters 2022). We investigated the efficacy and mechanism
of OR21 in combination with VEN or TKIs for myeloid malignancies, AML, and CML.
Aims: To investigate the efficacy and mechanism of the combination therapy of OR21
for myeloid malignancies.
Methods: We investigated the efficacy and mechanism of OR21 in combination with VEN
against AML and with TKIs against CML, in vitro and in vivo.
Results: In AML, OR21 plus VEN synergistically inhibited cell growth, increased apoptosis
in AML cell lines as similar to AZA and DAC, and significantly prolonged survival
in a xenograft mouse model using HL60 cell line (p<0.05). Gene expression analysis
using AML cell lines (HL60, KG1a) showed that OR21 plus VEN significantly downregulated
VAMP7 which regulates mitophagy/autophagy to maintain mitochondrial homeostasis compared
to VEN monotherapy. Consistently, OR21 plus VEN enhanced apoptosis by increasing ROS
accumulation and attenuated mitophagy/autophagy response triggered by decreased mitochondrial
membrane potential. The addition of mitophagy inducers, such as rapamycin and p62-mediated
mitophagy inducer, to OR21 plus VEN decreased ROS accumulation and apoptosis, indicating
OR21 enhanced ROS accumulation via attenuating VEN-induced mitophagy/autophagy pathway.
In CML, OR21 plus TKIs (imatinib, dasatinib, ponatinib) synergistically inhibited
cell growth, increased apoptosis in CML cell lines as similar to AZA and DAC, and
significantly suppressed the colony-forming capacity of bone marrow CD34+ stem/progenitor
cells in CML patients. OR21 plus imatinib significantly suppressed tumor growth compared
to imatinib monotherapy in a xenograft mouse model using K562 cell line (p<0.05).
In phosphoproteomic, transcriptome and methylome analysis (K562, KBM5), OR21 augmented
imatinib-induced apoptosis by dephosphorylating of STAT and Src family proteins by
upregulating several tumor suppressor genes, including PTPN6, through demethylation
of their promoter CpG regions.
Image:
Summary/Conclusion: OR21 enhanced the anti-tumor effect in combination with the molecular
agents for myeloid malignancies with different mechanisms: OR21 increased ROS accumulation
in combination with VEN, and dephosphorylated BCR-ABL1 downstream protein kinases
by demethylating effect in combination with TKIs. With its advantage as an oral HMA,
OR21 is expected to have future clinical trials as a combination therapy for myeloid
malignancies.
P414: RNA MODIFICATION-FOCUSED CRISPR-CAS9 SCREEN REVEALS POTENTIAL THERAPEUTIC TARGETS
MEDIATING CHEMORESISTANCE IN AML
M. Kienhöfer1,2,3,*, C. Pauli1,2, S. Delaunay2, C. Rohde1,3,4, M. F. Blank1,2,4, D.
Heid1,4, M. Frye2, C. Müller-Tidow1,3,4,5
1Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital
Heidelberg; 2German Cancer Research Center (DKFZ); 3University of Heidelberg Medical
Faculty; 4Molecular Medicine Partnership Unit, European Molecular Biology Laboratory–Heidelberg
University Hospital; 5National Center for Tumor Diseases (NCT), Heidelberg, Germany
Background: Acquired therapy resistance is one of the main causes of death in Acute
Myeloid Leukemia (AML). RNA-modifying proteins (RMP) are a diverse group of enzymes
responsible for the deposition and the removal of over 170 chemical modifications,
impacting key features of RNA biology. The RMPs represent a new focus for target therapies.
Aims: The objective of the current study is to uncover how RNA-modifying proteins
affects resistance to treatment in AML.
Methods: A CRISPR-Cas9-based screening approach was utilized to study RNA-modifying
proteins in an AML cell line and a derived cell line resistant to cytarabine. The
library pool targeted 150 RNA modifying enzymes, the 250 most abundant expressed snoRNAs
in AML, as well as a group of control genes and non-targeting sgRNAs. A lentiviral
system was used to introduce the CRISPR system. Multiplicity of infection (MOI) was
held under 0.3 to guarantee one gene knockout per cell. Post-infection, the cell lines
were divided into a control and a treatment group. The treatment group was stimulated
with increasing doses of cytarabine over 22 days. Samples were analyzed via NGS.
Results: In the CRISPR screen, we observed that many of the gene knockouts led to
cell depletion. Overall, 37 genes knockouts were depleted in the control group of
both cell lines without additional drug treatment, suggesting that the related genes
are required for survival and/ or proliferation. Further, 28 RNA-modifying enzymes
and 15 snoRNAs were altered upon drug treatment in at least the parental or the resistant
cell line. Interestingly, 15 of those 28 enzymes show additional effects on proliferation.
Based on this data, we chose a methyltransferase responsible for specific modification
on tRNAs for further analysis. Single knockout of the enzyme sensitized different
AML cell lines for cytarabine treatment and showed impairment of proliferation as
well as the ability to form colonies.
Summary/Conclusion: RNA modifications promote several hallmarks of cancer. This study
reports the involvement of specific RNA-modifying enzymes for therapy resistance and
proliferation in AML. However, it remains unclear whether these enzymes are dysregulated
in leukemic cells and therefore could display a targetable vulnerability. In addition,
further studies of the affected cellular pathway could contribute to a better understanding
of the underlying resistance mechanisms.
P415: TARGETED 3RD-GENERATION (NANOPORE) SEQUENCING REVEALS MANY NOVEL CIRCULAR RNAS
OF THE APOPTOSIS-RELATED BCL2L12 GENE, EXPRESSED IN HUMAN CELLS FROM ACUTE MYELOID
LEUKEMIA AND MYELODYSPLASTIC SYNDROMES
K. Xenou1, C. Sotiropoulou1, P. Karousi1, N. Machairas1, A. Scorilas1, V. Pappa2,
S. Papageorgiou2, C. Kontos1,*
1Department of Biochemistry and Molecular Biology, National and Kapodistrian University
of Athens; 2Second Department of Internal Medicine and Research Unit, University General
Hospital “Attikon”, Athens, Greece
Background: Circular RNAs (circRNAs) are non-coding RNAs generated via back-splicing.
They are indirect – yet pivotal – regulators of gene expression, as many of them bind
microRNAs (miRNAs) and render them unavailable to exert their direct regulatory functions.
They also interact with chromatin, altering its structure and affecting transcription
of several genes. Lastly, though considered as non-coding, few of them have also been
shown to encode short, functional polypeptides. Due to their multifaceted role and
the deregulation of their expression in cancer, circRNAs are related to the development
and progression of malignancies, including myelodysplastic syndromes (MDS) and acute
myeloid leukemia (AML). BCL2L12 is a prominent member of the apoptosis-related BCL2
family, with an already established role in AML. Moreover, BCL2L12 mRNA expression
has been associated with chemoresistance of malignant bone marrow blasts. However,
the identity and role of the circRNAs produced by alternative back-splicing of BCL2L12
primary transcripts in MDS and AML has not been investigated, so far.
Aims: We sought to determine the identity of BCL2L12 circRNAs expressed in MDS and
AML cell lines and to explore their putative interactions with miRNAs having an established
role in these disease entities.
Methods: MDS-L cells [MDS with del(5q) and complex karyotype], NB-4 and HL-60 cells
(acute promyelocytic leukemia with and without PML-RARA, respectively), U-937 and
THP-1 cells (acute monocytic leukemia), KASUMI-1 cells (AML with t(8;21)(q22;q22.1);
RUNX1-RUNX1T1) and MOLM-13 cells (AML with t(9;11)(p21.3;q23.3); KMT2A-MLLT3) were
propagated. Total RNA was extracted from these seven cell lines; 2μg of each RNA extract
were reverse-transcribed using random hexamers. Next, nested PCR with divergent primers
was performed, starting from each exon of the BCL2L12 gene; thus, only cDNAs from
BCL2L12 circRNAs were amplified. After having mixed and purified all nested-PCR products
of each cell line cDNA, nanopore sequencing libraries were built. Following nanopore
sequencing and basecalling in a MinION Mk1C 3rd-generation sequencer, sequencing reads
were aligned and corrected using Minimap2 and TranscriptClean, respectively. circRNA
identification was performed using an in-house–built, PERL-based algorithm; miRDB
was used for prediction of their interactions with miRNAs.
Results: We discovered 83 novel BCL2L12 circRNAs, 9 of which were common between MDS-L
cells and at least one AML cell line. The exon structure of these novel circRNAs showed
a remarkable diversity, comprising exons also encountered in messenger RNAs (mRNAs)
of BCL2L12 as intronic regions. This phenomenon was mostly observed in MDS-L cells.
Several BCL2L12 circRNAs in NB-4 and HL-60 cells exhibited complete intron retention;
additionally, a novel exon was present at a remarkable frequency in NB-4 cells. Moreover,
several BCL2L12 circRNAs are predicted to sponge miRNAs with a key role in AML (miR-7-5p,
miR-125b-5p and miR-182-5p) and/or MDS (miR-150-5p).
Summary/Conclusion: Numerous circRNAs are produced through alternative back-splicing
of the primary BCL2L12 transcripts. The circRNA expression profile of this gene differs
among the MDS and AML cell lines, as well as among the six AML cell lines. Intronic
regions of this apoptosis-related gene are included in several circRNAs, augmenting
the repertoire of miRNAs predicted to be bound by BCL2L12 transcripts. This phenomenon
was more common in the MDS cell line, followed by both acute promyelocytic leukemia
cell lines. Our data suggest an important regulatory role for this gene in MDS and
AML subtypes.
P416: CLONAL HEMATOPOIESIS IS COMMON IN AML LONG-TERM SURVIVORS AND MAY ASSOCIATE
WITH DIABETES AND SECONDARY NEOPLASIAS, BUT NOT OTHER HEALTH OUTCOMES
S. M. Krauß1,*, E. Telzerow2, A. S. Moret2, D. Richter3, M. Rothenberg-Thurley2, D.
Görlich4, M. C. Sauerland4, W. E. Berdel5, B. Wörmann6, U. Krug7, J. Braess8, P. Heussner9,
W. Enard3, W. Hiddemann2, K. Spiekermann2, U. Platzbecker1, K. H. Metzeler1
1Dept. Hematology and Cell Therapy, University Hospital Leipzig, Leipzig; 2Dept. of
Medicine III, University Hospital, Ludwig-Maximilians University; 3Dept. Biology II,
Ludwig-Maximilians University, Munich; 4Institute of Biostatistics and Clinical Research;
5Dept. of Medicine, Hematology and Oncology, University of Münster, Münster; 6Deutsche
Gesellschaft für Hämatologie und medizinische Onkologie, Berlin; 7Dept. of Medicine
3, University Hospital Leverkusen, Leverkusen; 8Dept. of Oncology and Hematology,
Hospital Barmherzige Brüder, Regensburg; 9Psycho-Oncology, Hospital Garmisch-Partenkirchen,
Garmisch-Partenkirchen, Germany
Background: Clonal hematopoiesis (CH), the outgrowth of hematopoietic stem cells and
their progeny with somatically acquired gene mutations, is a common phenomenon in
elderly individuals without known hematological disorders. CH has been linked to an
increased risk of developing myeloid neoplasia and cardiovascular diseases, resulting
in higher all-cause mortality. More specifically, clonal hematopoiesis of indeterminate
potential (CHIP) has been defined by hematopoietic clones with a variant allele frequency
(VAF) ≥2% in the absence of hematologic disease. CH is also known to be common in
patients with acute myeloid leukemia (AML) briefly after therapy, but prevalence and
relevance of CH in AML long-term survivors (AML-LTS) has not been investigated so
far.
Aims: We aimed to study CH in a cohort of 380 AML-LTS using a targeted next generation
sequencing (NGS) assay utilizing single-molecule Molecular Inversion Probes (smMIPs),
and to analyze determinants of CH and its effects on somatic health outcomes.
Methods: We employed smMIP-technology, which follows a hybridization-capture protocol
for NGS library preparation to selectively enrich and amplify hundreds of genomic
target loci covering 82 regions in 24 CH and AML-related genes in a single reaction.
Unique molecular identifiers (UMIs) within probes enabled computational correction
of sequencing errors to significantly increase precision of variant calls.
Results: We successfully established a UMI-based smMIP NGS assay as well as a custom
computational analysis pipeline to enable the reliable detection of variants ≥0.5%
VAF. In whole blood specimens from 380 AML-LTS we detected CH in 61.4% of individuals,
including CHIP (VAF ≥2%) in 35.8%. Stratification by treatment group, i.e. chemotherapy/autologous
hematopoietic stem cell transplantation only (chemo) vs. allogeneic hematopoietic
stem cell transplantation (alloSCT), revealed a significantly lower prevalence of
CHIP in the alloSCT group (26.0% vs. 52.1%; p<.001). Concordantly, median VAF of CH
mutations after alloSCT was significantly lower than in the chemo group (1.2% vs.
1.8%; p=.005). CHIP prevalence increased with age in the chemo group, from 21.7% (<50
years) to 70.0% (≥70 years), whereas it was independent of patient age in the alloSCT
group (Figure A). Conversely, CHIP prevalence was independent from time since start
of therapy in the chemo group, but increased with time after transplantation in the
alloSCT group, from 18.4% (0-5 years) to 46.2% (16-18 years; Figure B). In both treatment
groups, the known CHIP driver genes DNMT3A and TET2 were most frequently mutated.
Notably, PPM1D (p<.001) and TP53 (p<.001) were more frequently mutated in the chemo
group (Figure C). We did not detect significant differences in complete blood counts
of AML survivors according to CH status. While most somatic comorbidities showed no
clear association with CH status, prevalence of diabetes and development of secondary
cancer after leukemia therapy were increased in patients with CH (VAF <2%), and more
so in those with CHIP (Figure D).
Image:
Summary/Conclusion: We report a high prevalence of CH in AML long-term survivors,
particular in those treated with chemotherapy without alloSCT. CHIP prevalence increases
with age in patients treated with chemotherapy, and with time elapsed after transplantation
in patients who underwent alloSCT. We reveal distinct mutation spectra for both groups.
Although associations between CH and somatic morbidities seem to be weak overall,
we found associations with diabetes and the development of secondary cancer after
leukemia treatment, which warrant further exploration.
P417: A SINGLE AMINO ACID AT THE PNT DOMAIN OF ERG MEDIATES ITS LEUKEMOGENIC ACTIVITY
THROUGH INTERACTION WITH THE NCOR-HDAC3 CO-REPRESSOR COMPLEX.
E. Kugler1,2,*, S. Madiwale2, D. Yong3, Y. Birger2,4, J. A. I. Thoms5,6, D. B. Sykes7,8,
M. Yassin9, N. Aqaqe9, A. Rein2,4, H. Fishman2, I. Geron2, C.-W. Chen10,11,12, B.
Raught13, Q. Liu10, M. Milyavsky9, J. Pimanda5,6, G. G. Privé3,13, S. Izraeli2,4
1Institute of Hematology, Davidoff Cancer Center, Rabin medical center, Petah Tikva;
2Department of Human Molecular Genetics and Biochemistry, Sackler Medical School,
Tel Aviv University, Tel Aviv, Israel; 3Department of Biochemistry, University of
Toronto, Toronto, Canada; 4The Rina Zaizov Pediatric Hematology and Oncology Division,
Schneider Children’s Medical Center, Petach Tikva, Israel; 5Adult Cancer Program,
Lowy Cancer Research Centre; 6Faculty of Medicine, UNSW Sydney, Sydney, Australia;
7Center for Regenerative Medicine, Massachusetts General Hospital, Boston; 8Harvard
Stem Cell Institute, Cambridge, United States of America; 9Department of Pathology,
Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; 10Department of
Systems Biology, Beckman Research Institute, City of Hope, Duarte; 11Department of
Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston;
12City of Hope Comprehensive Cancer Center, Duarte, United States of America; 13Princess
Margaret Cancer Centre, University Health Network, Toronto, Canada
Background: The ETS transcription factor ERG is essential for the maintenance of hematopoietic
stem cells. However, it has also been implicated as an oncogene in the development
of acute leukemia. Our studies and those of others have demonstrated that ERG directly
contributes to the initiation and maintenance of lymphoid and myeloid acute leukemia
subtypes. Nevertheless, ERG co-factors critically involved in leukemogenesis remain
largely uncharacterized.
Aims: Here we report a critical role for the conserved amino-acid proline at position
199, at the 3’ end of the PNT domain, for ERG’s leukemogenic activity.
Methods: A genomic and proteomic analysis has been used to study the role of P199
in the leukemogenic pathway of ERG.
Results: We specifically demonstrate in functional and gene expression assays that
P199 is required for ERG-induced myeloid differentiation restriction and for self-renewal
and maintenance of murine hematopoietic stem and progenitor cells (HSPC).
As P199 is located within the PNT domain (protein interaction domain), we attempted
to identify key protein interactions associated with leukemia progression induced
by ERG. To this end, we used proximity ligation-mass spectrometry (BioID). In HEK293
cells, proximity maps of WT-ERG and a mutated form of ERG at position 199 (P199L-ERG)
were compared. A total of 240 putative protein interactors with a significance analysis
of interactome were identified. The hits were highly enriched with chromatin modifiers.
Most significantly, the mutation severely impaired ERG’s interaction with components
of the NCoR-HDAC3 complex, resulting in a 40% reduction in the number of spectral
counts in comparison with wild-type ERG.
ChIP sequencing for histone markers conducted on ER-Hoxb8 cells (transformed murine
GMPs) demonstrated a decrease in H3K27ac signature at over 1500 unique sites in cells
transduced with WT-ERG compared to those transduced with the mutant. Interestingly,
these sites were predominantly associated with enhancers of genes expressed in mature
myeloid cells.
Next, we addressed the role of HDAC3 in ERG’s transcriptional activity. RNA sequencing
of human ER-Hoxb8 cells treated with BRD3308 (an HDAC3 inhibitor) for 48 hours was
performed. Interestingly, leukemia-associated pathways (Hoxa9-Meis1, CBFA2T3 and repression
of myeloid differentiation genes) were ranked at the leading edge of gene sets that
were significantly perturbed after HDAC3 inhibition.
Moreover, we tested the significance of the ERG/NCoR-HDAC3 interaction in models of
human leukemia. We demonstrated that ERG-dependent human AML cells as SKNO1 (AML1-ETO)
and TF1 (CBFA2T3-ABDH12) were more sensitive to HDAC3 inhibition (RGFP966) as compared
to ERG independent cells.
Furthermore, RGFP966 exhibited a significant in-vivo antileukemic effect as measured
by the burden of disease in the bone marrow of NSG mice transplanted with SKNO1 cells.
By using the CRISPR-dCAS9 system we were able to show that leukemia development is
abrogated in mice transplanted with SKNO1 cells expressing a reduced level of HDAC3.
Remarkably, this effect was translated into a significant survival advantage.
Summary/Conclusion: Taken together, our findings indicate that the aberrant overexpression
of ERG maintains HSPCs in an undifferentiated state and promotes AML development.
We suggest that the interaction between ERG and the NCoR-HDAC3 complex has an important
role in the leukemogenic process, and that HDAC3 inhibition could be beneficial in
AML characterized by high ERG expression.
P418: MODELLING AND TARGETING ACUTE MYELOID LEUKAEMIA CELLS IN THE BONE MARROW PROTECTIVE
NICHE
E. E. Ladikou1,2,*, K. Sharp1, T. A. Burley1, E. Kennedy1, T. Chevassut2, C. Pepper1,
A. G. Pepper1
1Clinical and Experimental Medicine, Brighton and Sussex Medical School; 2Department
of Haematology, Brighton and Sussex University Hospital Trust, Brighton, United Kingdom
Background: AML is a therapeutic challenge due to its aggressiveness and biological
heterogeneity. Although 80% of patients initially achieve a complete remission, the
long-term disease-free survival is poor. One issue contributing to disease relapse,
is persistence of disease in the protective niche of the Bone Marrow microenvironment
(BMME). Here, AML cells are surrounded by other cell types that promote their survival,
which enables them to evade therapeutic destruction and promotes the emergence of
drug resistance. Clinical trials of single agent mobilising drugs, like Plerixafor,
have yielded promising results but they are not curative. So, the development of combinatorial
therapies that simultaneously target different components of AML cell adhesion may
release more AML cells into the circulation, where they can be targeted by standard
therapies.
Aims: a) To develop a multi-cellular, co-culture system to recapitulate the adhesive
BMME, b) To test rational drug combinations and identify targets with the potential
to mobilise anchored AML cells and c) To perform paired transcriptomic and phenotypic
analysis of persistently adhered versus mobilised AML cells to detect novel targets.
Methods: We have developed a novel 96-well plate multi-cellular, co-culture system
using stromal cells (HS5), endothelial cells (HUVEC), osteoblasts (hFOB 1.19) and
AML cells (OCI-AML3 and KG1a). Multiple mobilisation drugs, alone and in combination,
were tested and AML cell release was quantified by flow cytometry. Drugs tested to
date include: Plerixafor (CXCR4 inhibitor), Natalizumab (anti-α4-integrin antibody),
ONO-7161 (novel CXCR4 inhibitor), purified anti-CD44 and anti-E-Selectin. Phenotypic
and transcriptional comparisons were made between the persistently adherent and mobilised
AML cells.
Results: The most adhesive physiological “BMME mix” was identified as HS5, HUVEC and
hFOB 1.19 in equal 1/3 proportions (p=0.0001). Co-culture with AML cells resulted
in 74% and 68% adherence of KG1a and OCI-AML3, respectively. Phenotypic analysis of
adhered vs non-adhered AML cells identified CD44, CXCR4, CD49d and CD38 as important
markers of optimal BMME adhesion. Subsequently, our novel co-culture system was used
as a drug testing platform to assess clinically available agents. Plerixafor and Natalizumab
increased detachment by 1.3-fold (p=0.0132) and 1.2-fold (p=0.015) respectively. In
contrast, ONO7161 and anti-E-selectin had little effect. The most promising candidate,
anti-CD44, resulted in a 2-fold and 1.6-fold detachment of KG1a (p=0.004) and OCI-AML3
(p=0.027), respectively. However, even in the presence of the maximum dose of anti-CD44
(5µg/mL)), 46% of KG1a and 48% of OCI-AML3 cells remained persistently adhered. Synergy
experiments with anti-CD44 in combination with either Plerixafor or Natalizumab yielded
no better release than anti-CD44 alone.
Summary/Conclusion: We have developed a novel, multi-cellular co-culture system that
recapitulates the adhesive BMME. We are using this as a drug testing platform for
AML to identify the most potent release agents and test the effects of cytotoxic drugs
on those most persistently adhered. To-date, anti-CD44 is the most effective release
agent in our model, but it still failed to release all AML cells. Transcriptomic analysis
of the persistently adherent AML cells should enable us to delineate the critical
biological interactions required for BMME adhesion. This will enable us to identify
rational new targets to effectively remove chemo-resistant AML cells from the protective
BM niche.
P419: IDENTIFICATION OF NOVEL THERAPEUTIC OPTIONS FOR VENETOCLAX-RESISTANT AML CELLS
THROUGH DRUG REPURPOSING
A. Ladungova1,2,*, D. Busa3, Y. Lodhi1,4, J. Hyl3, M. Culen3,5, M. Smida1,3,5
1Central European Institute of Technology, Masaryk University; 2National Centre for
Biomolecular Research, Faculty of Science, Masaryk University; 3Department of Internal
Medicine - Hematology and Oncology, Faculty of Medicine, Masaryk University; 4Department
of Biology, Faculty of Medicine, Masaryk University; 5Department of Internal Medicine
- Hematology and Oncology, University Hospital Brno, Brno, Czechia
Background: Acute myeloid leukemia (AML) is a malignant disease derived from the bone
marrow precursors of myeloid lineage. Treatment options are rather limited, primarily
based on chemotherapy, and often result in disease progression. Recently, venetoclax-based
therapies have transformed the frontline regimens of elderly patients and patients
unfit for intensive chemotherapy. Despite its promising outcomes in clinical studies,
multiple resistant subclones evolved during the treatment acting as a barrier in disease
regression. Understanding the venetoclax-resistance mechanisms and detecting the major
determinants could reveal previously unrecognized novel perspectives for therapeutic
strategies to improve patients’ outcomes. Moreover, performing high-throughput screenings
with clinically approved drugs could reveal novel treatment options for resistant
subclones of AML.
Aims: Our project aims 1) to identify novel FDA-approved drugs effective for treating
venetoclax-resistant AML cells through a high throughput screening, 2) to validate
the most effective compounds, prove their specificity, and 3) to investigate the molecular
background mechanisms underlying the observed sensitivity.
Methods: Two different models mimicking the microenvironment of venetoclax-resistance
formation have been established: 1.) Patient-derived mouse xenograft (PDX) model.
AML PDX model was generated by using AML patient sample transplanted into an immunocompromised
murine host and treated with different regimens of venetoclax; 2.) Venetoclax-resistant
cell lines (MOLM-13, OCI-AML3, HL-60). Generation of the venetoclax-resistant AML
cell lines was achieved through the chronic administration of the compound, inhibiting
the cell viability to 80-90% in multiple rounds with gradually increasing concentrations
until the cells become fully resistant. Both models are next used for the high-throughput
screening of 859 approved drugs, covering broad chemical space. The standard concentration
(1 μM) of drugs in the library is automatically added to cells on 384-well-plates
with a programmed liquid handling system epMotion (Eppendorf). Cell viability is determined
after 72 hours by a Cell-titer Glo assay. Compounds with the strongest effect upon
resistant AML cell viability are validated through 10-point dose-response curves with
2-fold-dilutions. Moreover, the investigation of molecular mechanisms responsible
for the sensitization of resistant AML cells is planned by using various cell biology
and molecular biology techniques.
Results: Treating the PDX mouse model with venetoclax for 6 weeks resulted in the
generation of resistant cells. Importantly, these cells also acquired additional resistance
to topoisomerase inhibitors, DNA-damaging agents and many other compounds. On the
contrary, both 6-week as well as 3-week venetoclax treatments generated cells exquisitely
sensitive to the iron chelator deferoxamine mesylate and a purine nucleoside analogue
clofarabine. In addition, we generated a venetoclax-resistant MOLM-13 cell line with
1500-fold decreased sensitivity and have subjected it to the drug screening to correlate
its response with the PDX model.
Summary/Conclusion: The outcomes of this research project point out the importance
of novel treatment options to overcome venetoclax-resistance in AML cells. Further
validations and investigation of the molecular mechanisms are planned for the top-performing
drugs to strengthen the translational potential for AML therapy.
This project was supported by the grants number MUNI/A/1330/2021, MUNI/A/1419/2021
and by OPRDE (No.CZ.02.2.69/0.0/0.0/19_073/0016943).
P420: FORCING LEUKAEMIC STEM CELL TO LEAVE QUIESCENT STATE BY INHIBITING HEDGEHOG,
NOTCH AND WNT/BETA-CATENIN SIGNALLING PATHWAYS
D. Láinez-González1,*, J. Serrano-López1, P. Llamas-Sillero2, J. M. Alonso-Dominguez2
1Experimental Hematology Lab, Instituto de Investigación Sanitaria Fundación Jiménez
Díaz; 2Department of Hematology, Hospital Universitario Fundación Jiménez Díaz, Madrid,
Spain
Background: Acute myeloid leukaemia (AML) is a clonal neoplasm with a high relapse
rate. It is thought that this relapse is caused by chemoresistance of leukaemic stem
cells (LSC) due to their quiescent state. Numerous signalling pathways have been described
that regulate quiescence. Key players in this setting are the Hedgehog (Hh), Notch
and WNT/β-Catenin pathways. Current chemotherapeutic agents target the cell population
in the proliferative phase of the cell cycle. Therefore, cells in quiescence are not
affected by drugs such as cytarabine (AraC). Theoretically, forcing quiescent cells
into the proliferative phase would help to eradicate the LSC population and avoid
relapses.
Aims: To force quiescent LSC to enter cell cycle by inhibiting Hedgehog, Notch or/and
Wnt/Beta-Catenin.
Methods: Three acute myeloid leukemia cell lines, HL60, OCI-AML3 and KASUMI1, and
13 primary samples were used to study cell cycle and cytotoxicity. They were treated
during 24-48 hours with Glasdegib, Nirogacestat and PRI-724 which target Smoothened
(Hedgehog), Gamma-Secretase (Notch) and Beta-Catenin (Wnt) proteins respectively.
Studies of drug combination were also performed to inhibit different signaling pathways
at the same time. Cytarabine was employed during 48-72 hours in cell lines or during
24-48 hours in primary samples. Cytotoxicity was analyzed by WST8 assay in cell lines
and flow cytometry in primary samples by using KI67 and Annexin V. Cell cycle was
study by flow cytometry using KI67 and 7AAD, and in primary samples CD45, CD34 and
CD38 antibodies were used to detect LSC. Cell lines experiments were triplicated.
Results: In HL60, double inhibition during 48h of Hedgehog and Notch with 15 nM Glasdegib
+ 5 nM Nirogacestat reduced quiescent state in 67.27% compared with DMSO. Furthermore,
this proposed treatment in combination with 109.1 nM AraC decreased 27-70% viability
compared with AraC monotherapy at 72h. Similar results were obtained in OCI-AML3,
in which the proposed treatments reduced quiescent phase compared with control, but
no changes were observed in viability. On the other hand, in KASUMI1 there were no
differences either in cell cycle or viability assay. In primary CD34+ samples, when
Hedgehog and Notch were inhibited during 48h, quiescent state decreased in 33.06%,
but no effects in viability were observed.
Image:
Summary/Conclusion: The number of primary samples should be increased to shed light
on the efficacy of this interesting therapeutic strategy. Nevertheless, inhibition
of Hh, Notch and WNT pathways reduce the quiescent population either measuring the
disease as a whole or looking at the LSC population. Differences observed between
different AML cell lines regarding increase of the citotoxicity might be explained
by AML genetic heterogeneity.
P421: LOWER VΓ9VΔ2 T CELLS RELATIVE FREQUENCIES PREDICT POORER SURVIVAL IN NEWLY DIAGNOSED
ACUTE MYELOID LEUKEMIA
A.-C. Le Floch1,2,*, F. Orlanducci1,2, A. Le Roy1,2, J.-F. Hamel3, N. Ifrah4, P. Cornillet-Lefebvre5,
J. Delaunay6, C. Récher7, E. Delabesse8, A. Pigneux9, M.-C. Béné10, N. Vey11, A.-S.
Chretien1,2, D. Olive1,2
1Equipe Immunité et Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM),
INSERM U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, UM105;
2Plateforme d’immunomonitoring, Institut Paoli-Calmettes, Marseille; 3Département
de Biostatistiques, CHU d’Angers, Université d’Angers; 4Département d’Hématologie,
CHU d’Angers, Université d’Angers, Inserm, CRCINA, Angers; 5Laboratoire d’Hématologie,
CHU de Reims, Reims; 6Département d’Hématologie, CHU de Nantes, Nantes; 7Département
d’Hématologie; 8Laboratoire d’Hématologie, CHU de Toulouse, Institut Universitaire
du Cancer de Toulouse Oncopôle, Université Toulouse III Paul Sabatier, Toulouse; 9Département
d’Hématologie et Thérapie Cellulaire, CHU de Bordeaux, Bordeaux; 10Laboratoire d’Hématologie,
CHU de Nantes, Nantes; 11Département d’hématologie, CRCM, INSERM U1068, CNRS UMR7258,
Institut Paoli-Calmettes, Aix-Marseille Université UM105, Marseille, France
Background: Major role of γδ T cells has recently emerged in anti-tumor immunity.
Across several tumor subtypes, increased relative frequencies of γδ T cells had the
best prognostic value in comparison to other immune subsets. In acute leukemias, patients
with increased γδ T cells reconstitution after allogenic stem cell transplantation
had better outcomes. The less studied putative prognostic impact of Vγ9Vδ2 T cells
abundance in newly diagnosed (ND) AML relies almost on inference of transcriptomic
data or post induction frequencies and has not been assessed with respect to other
confounding factors.
Aims: The aim of this study was to determine the predictive and prognostic impact
of blood Vγ9Vδ2 T cells frequencies at diagnosis, as measured using phenotypic analysis,
taking into account biological and clinical parameters in patients treated for AML
with induction.
Methods: Proportion of Vγ9Vδ2 T cells among lymphocytes has been assessed by flow
or mass cytometry on PBMCS from AML patients at diagnosis, obtained from the Department
of Hematology of Institut Paoli Calmettes or from the tumor bank of the FILO group.
Correlations between clinical outcome, patient or disease characteristics, and Vγ9Vδ2
T cells frequencies have been analyzed. For some of the samples, we characterized
immune alterations displayed by Vγ9Vδ2 T cells, or performed Vγ9Vδ2 T cell expansion
and degranulation assays of autologous Vγ9Vδ2 T cells.
Results: 198 patients were included in this study. Mean age was 56 years [18.9-81.5],
79 (39.9%) and 119 (60.1%) patients had respectively a favorable or an intermediate
ELN classification. Mean CR rate was 81.3%. Analysis of overall survival (OS) and
relapse free survival (RFS) based on Vγ9Vδ2 T cell frequencies among lymphocytes,
highlighted that the 0.8% threshold was the most significant. Vγ9Vδ2 T cells relative
frequencies were considered low for values below this threshold.
By univariate analysis, patients with lower Vγ9Vδ2 T cells at diagnosis had significantly
lower 5-year OS and RFS (HR = 1.54, 95%CI = 1.03-2.31, p = 0.028 and HR = 1.69, 95%CI
= 1.09-2.62, p = 0.015). These results were confirmed in multivariate analysis (HR
= 1.55 [1.04-2.30], p = 0.030 and HR = 1.64 [1.06, 2.53], p = 0.025). Phenotypic alterations
found in patients with lower Vδ2 included a loss of some cytotoxic Vγ9Vδ2 T cell subsets
and a decrease in BTN3A expression on the surface of blasts. Finally, in vitro, samples
irrespective of their Vγ9Vδ2 T cells numbers did expand and displayed similar functional
effects against target cells after stimulation with zoledronate or anti-BTN3A 20.1
agonist mAb.
Summary/Conclusion: This study confirmed the prognostic value of Vγ9Vδ2 T cells predominance
among lymphocytes, in ND AML patients treated with induction. Vγ9Vδ2 T cells frequencies
could be easily included in algorithm of treatment decision. Our findings contribute
also to a strong rationale to elaborate consolidation protocols targeting enhancement
of Vγ9Vδ2 T cells response, such as anti-BTN3A agonist antibodies that are under development
(Marabelle et al. ESMO 2021).
P422: OMIPALISIB, A DUAL PI3K/MTOR INHIBITOR, TARGETS MITOCHONDRIA AND IMPAIRS OXIDATIVE
PHOSPHORYLATION IN ACUTE MYELOID LEUKEMIA
C.-Y. Tseng1,*, C.-Y. Kuo1, Y.-H. Fu1,2, H.-A. Hou3, H.-F. Tien3, L.-I. Lin1
1Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University,
Taipei, Taiwan; 2Irell & Manella Graduate School of Biological Sciences, Beckman Research
Institute of City of Hope, Duarte, United States of America; 3Internal Medicine, National
Taiwan University Hospital, Taipei, Taiwan
Background: Mitochondria play a central role in cell metabolism. Recent studies explored
mitochondrial alterations contributing to metabolic vulnerability in myeloid leukemia
cells may considered as therapeutic targets. We previously reported that omipalisib,
a PI3K/mTOR dual inhibitor, could significantly inhibit the growth of myeloid leukemia
cells and impair their mitochondrial respiration. However, the precise mechanism of
the mitochondria dysfunction in response to omipalisib is still not fully studied.
Aims: To explore the mechanism of the mitochondria dysfunction in response to omipalisib.
Methods: OCI-AML3 and THP-1 myeloid leukemia cell lines were used in this study. Omipalisib
(GSK2126458) and another PI3K/mTOR dual inhibitor gedatolisib (PF-05212384) were used.
Flow cytometry was used for mitochondrial analysis. RNA-seq was performed by using
an Illumina NovaSeq 6000 platform. Differentially expressed genes (DEGs) between control
and omipalisib (or gedatolisib) were identified by EBseq with a threshold of p ≤ 0.05.
The mRNA, nuclear and mitochondrial DNA quantification were measured by using QuantStudio 3
Real-Time PCR Systems. The parameters of mitochondrial respiration were analyzed by
using the XFe 24 extracellular flux analyzer.
Results: We demonstrated the anti-proliferative effect of omipalisib and gedatolisib
on a panel of AML cell lines with different genetic backgrounds. OCI-AML3 cell lines
had significant response to omipalisib and gedatolisib with IC50 of 17.45 nM and 153.38 nM,
respectively. Subsequently, the transcriptome analysis of 50 nM omipalisib or 200 nM
gedatolisib-treated OCI-AML3 cells were analyzed. Compared to DMSO-treated, a total
of 1199 (595 upregulated and 604 downregulated) and 326 (218 upregulated and 108 downregulated)
DEGs were identified in the omipalisib-treated cells and the gedatolisib-treated cells,
respectively. Gene set enrichment analysis (GSEA) indicated that both omipalisib and
gedatolisib could suppress “E2F targets”, “Myc targets”, and “G2M checkpoint”, and
trigger “TNFα signaling via NFκB”, and “Inflammatory response”. Of them, we found
that omipalisib had more significant effect to inhibit “Oxidative phosphorylation”
(NES: -2.01) than gedatolisib did (NES: -1.40), especially mitochondrial biogenesis
and amino acid metabolism-related pathways. Omipalisib had an inhibitory effect on
the mitochondrial basal respiration rate and maximal respiration. Further analysis
revealed quantitative reverse-transcription PCR analysis revealed that both omipalisib
and gedatolisib could potently suppress serine synthesis (PHGDH, PSAT1, PSPH), glycine
synthesis (SHMT1/2), and tetrahydrofolate cycle-related genes (MTHFD1/2) at the transcriptional
level. On the other hand, rather than gedatolisib, omipalisib could significantly
inhibit expression of glycolysis-related genes (GLUT1, HK2, PKM2, and LDHA) as well
as mitochondrial biogenesis-related genes (PPARGC1B, TFAM, and NDUFS6). In addition,
omipalisib could strongly decrease mtDNA and mitochondrial mass. These results suggest
that omipalisib could suppress the mitochondrial biogenesis through alternative mechanisms,
including but not limited to PI3K-AKT-mTOR signaling.
Summary/Conclusion: Rather than gedatolisib, omipalisib could significantly inhibit
mitochondrial biogenesis and suppress the respiration of mitochondria in myeloid leukemia
cells. Our results indicate that omipalisib could suppress cell proliferation not
only through PI3K-AKT-mTOR signaling but also via impairing mitochondrial biogenesis.
This information may be potentially suitable for future clinical applications.
P423: ADAPTING CRISPR CAS9 DROPOUT SCREENS TO IN VIVO PDX MODELS OF ACUTE LEUKEMIAS
R. Ludwig1,2,*, D. Amend1, E. Bahrami1, I. Jeremias1,2,3
1Apoptosis in Hematopoietic Stem Cells, German Research Center for Environmental Health
(HMGU), Munich; 2German Cancer Consortium (DKTK), partner site Munich; 3Department
of Pediatrics, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany
Background: Acute leukemias require better treatment and targeted therapies represent
interesting future therapeutic options. Such therapies precisely inhibit molecules
with essential function, also called vulnerabilities or dependencies, so that leukemia
cells die once the target is inhibited.
Aims: Here, we aimed at identifying therapeutic targets on a patient individual level
and in the surrounding of a living organism. Towards this aim, we established CRISPR
Cas9 dropout screens in PDX models of acute leukemias in vivo.
Methods: Primary patient acute lymphoblastic leukemia (ALL) and acute myeloid leukemia
(AML) samples were transplanted into immunocompromised NSG mice and re-passaged to
develop serially transplantable PDX models. To overcome limited transduction efficiency
inherent to PDX leukemia cells, lentiviruses were used.
Results: We first elaborated on the maximum library size to be used in PDX models
in vivo. As only a limited number of PDX cells home into mice, library size is limited
to ensure the coverage of the library. We used, screening sgRNAs as genetic barcodes
in Cas9 negative samples to determine the maximum library size upon next generation
sequencing (NGS); a regular distribution of all sgRNAs recovered from mice at the
end of the experiments indicated a suitable library size. We found that PDX ALL models
tolerated a larger library compared to PDX AML models that might reflect lower intra-sample
heterogeneity and higher leukemia stem cell frequency in ALL compared to AML.
To perform knockout screens, a split-construct for Cas9 was used to reduce plasmid
size and optimize lentiviral transduction efficiency into PDX cells; concomitantly
split-GFP was used to enrich cells expressing both Cas9 split plasmids by flow cytometry.
A customized library consisting of 146 target genes combined with positive and negative
control genes was designed at 5 sgRNAs per gene using the CLUE platform (www.crispr-clue.de)
(Becker et al., Nucleic Acids Res. 2020) and cloned into a lentiviral vector backbone.
The sgRNA library vectors include either a puromycin-resistance cassette or an H-2Kk
surface marker, each combined with a BFP fluorescent marker. This allows to determine
transduction efficiency by FACS-based detection of BFP and to enrich the transduced
PDX cells either by puromycin selection or by H-2Kk-MACS selection. Input samples
were collected after library transduction and enrichment to >90% CRISPR/Cas9 sgRNA
library positive cells, while remaining cells were injected into immunocompromised
NSG mice, grown in vivo and re-isolated at advanced disease stage. Analysis of input
and endpoint samples using NGS enabled to determine the frequency of sgRNAs and bioinformatically
compare the abundance of different sgRNAs between the control and the samples of interest
using MAGeCK (Model-based Analysis of Genome-wide CRISPR-Cas9 Knockout). A significant
sgRNA dropout was characterized by a p-value below 0.05 and a FDR below 0.1.
The screening experiments led to the top 10 depleted dropout genes for ALL and AML
PDX samples.
Besides individual dropouts of the different samples, also shared dropouts were detected.
Especially sample overlapping dropouts might represent interesting starting points
for new therapeutic options.
Summary/Conclusion: In summary, we have established a CRISPR Cas9 screening pipeline,
which allows investigating therapeutic targets on a patient-individual level in ALL
PDX models and, for the first time, in AML PDX models in vivo.
P424: INVESTIGATING GPR56 FUNCTION IN HUMAN IPSC-DERIVED HEMATOPOIETIC CELLS USING
A TET-ON MODEL OF CONSTITUTIVE ACTIVATION
L. Mackintosh1,*, A. Maglitto1, C. Rodriguez Seoane1, S. A. Mariani1, A. G. Rossi1,
E. Dzierzak1
1Centre For Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
Background: GPR56 (ADGRG1) is a widely expressed adhesion G-coupled protein receptor
in humans and mice which is implicated to play a role in both haematopoietic development
and Acute Myeloid Leukaemia (AML). In AML, there exists a low frequency subpopulation
of leukaemic stem cells (LSCs) which, similar to their benign Hematopoietic Stem Cell
(HSC) counterparts, exhibit stem cell behaviours. High expression of GPR56 forms part
of the molecular signature of these LSCs, and AML cases with high levels of GPR56
at diagnosis have poorer outcomes. Our hypothesis is that GPR56 activity underlies
some of the stem cell behaviours seen in both HSCs and LSCs.
Aims: To characterise the effects of GPR56 receptor activation so as to better understand
its role in AML and hematopoietic development.
Methods: We developed a Tet-On human inducible stem cell line (M1) transfected with
a construct encoding a truncated GPR56 protein. This constitutively active GPR56 receptor
form is overexpressed following doxycycline administration. Flow cytometry-based cell
cycle and apoptosis studies were performed on the M1 line and wild type Sci55 (WT)
iPSCs +/- doxycycline exposure. IncuCyte® transmigration studies utilising an SDF-α
gradient were performed to examine cell migration. Finally, the M1 cells were subjected
to a hematopoietic differentiation protocol and assessed for CD34+ CD38- compartment
size at day 12 of differentiation.
Results: GPR56 activation alters cell cycle progression with significant accumulation
of M1 cells in the G0/1 and G2/M phases of the cell cycle compared to WT (p <0.005).
GPR56 activation also causes a significant reduction in apoptotic cells in dox-treated
M1 cells (p <0.005), despite doxycycline being pro-apoptotic in WT cells (p <0.05).
GPR56 activation reduces the number of transmigrating M1 cells in response to an SDF-α
gradient (p <0.05). Importantly, GPR56 activation during differentiation increases
the CD34+ CD38- compartment size in M1 cells treated with doxycycline at day 12 (p<0.05)
as compared to untreated cells
Summary/Conclusion: GPR56 activation results in accumulation of undifferentiated M1
iPSCs in the G0/1 and G2/M phases, suggestive of cell cycle checkpoint arrest, and
exerts a powerful anti-apoptotic effect. GPR56 activation also reduces cell transmigration
in response to SDF-α, suggesting receptor involvement in cell adhesion and migration.
Finally, GPR56 activation during hematopoietic differentiation results in an increased
yield of CD34+ CD38- cells, suggesting that GPR56 activity may influence the proportion
of immature cells arising during haematopoietic differentiation. These results are
of interest in relation to normal HSC functions and may indicate the mechanisms through
which overexpression of GPR56 (with increased activation) in LSCs contributes to poorer
outcomes in AML. Future work will include identification and characterisation of the
signalling pathways downstream of the GPR56 receptor to better understand this receptor’s
role in the haematopoietic system in health and disease.
P425: ROLE OF LYSOSOMAL-ASSOCIATED GENE LAPMT4B IN LEUKEMIA
J. Delgado-Martínez1,2,*, J. M. Cornet-Masana1, L. Cuesta-Casanovas1,3, J. M. Carbó1,4,
L. Clément-Démange4, R. M. Risueño1
1Josep Carreras Leukaemia Research Institute; 2Faculty of Pharmacy, University of
Barcelona; 3Faculty of Bioscience, Autonomous University of Barcelona; 4Leukos Biotech,
Leukos Biotech, BARCELONA, Spain
Background: Despite the efforts to find druggable targets in acute myeloid leukemia
(AML), successful therapeutic approaches are needed in this disease. Due to the particular
characteristics of lysosomes in cancer, the clinical relevance of key lysosomal genes’
expression was searched in public repositories. LAPTM4B gene expression was found
to be directly correlated with a worse clinical outcome in AML.
Aims: To decipher the role of the lysosome-associated gene LAPTM4B in AML will be
the main objective of this study.
Methods: Generation of AML cells overexpressing both LAPTM4B isoforms (35kDa -long-
and 24kDa -short) were obtained by lentiviral transduction. Sensitivity to chemoresistance
was measured by flow cytometry. To determine the role of LAPTM4B in the leukemia regeneration
capacity, conditioned NSG mice were transplanted with both LAPTM4B isoforms-expressing
AML cells and the engraftment was analysed by flow cytometry. Activation of key secondary
messengers was measured by luciferase response elements and Western Blot.
Results: LAPTM4B overexpression directly correlated with a higher resistance to cytarabine.
LAPTM4B short isoform presented an increased clonogenic potential versus the long
isoform. Interestingly, a competitive bone marrow transplantation assay confirmed
this effect in terms of in vivo engraftment capacity. Moreover, LAPTM4B-mediated activation
was able to induce MAPK signalling in the leukemic cell context.
Summary/Conclusion: LAPTM4B overexpression not only increases the regeneration potential
but also confers resistance to cytarabine, suggesting an effect in the maintenance
of leukemia. In summary, this study will increase the knowledge of the role of lysosomes
in leukemogenesis, and additionally, it will help to identify new lysosome-related
biomarkers.
P426: A SINGLE-CENTER RETROSPECTIVE ANALYISIS OF THE MUTATIONAL PROFILE IN ACUTE MYELOID
LEUKEMIA PATIENTS
Y. Martínez Díez1,*, A. Franganillo Suárez1, J. Cornago1, L. Solan2, R. Ayala3, J.
Martinez3, P. Llamas1, J. L. Lopez Lorenzo1
1Hematology; 2Hospital Fundación Jiménez Díaz; 3Hematology, Hospital 12 de Octubre,
Madrid, Spain
Background: Acute myeloid leukemia (AML) is a clonal disorder originating from specific
genetic aberrations in myeloid precursor cells. Next generation sequencing (NGS) analyses
provide clinicians with a deeper knowledge of AML pathogenesis and is being implemented
as a routine test at diagnosis due to its relevance in management and prognosis, following
the characterization of molecular subgroups.
Here we provide a summary of our experience in a single centre (Fundación Jiménez
Díaz University Hospital (FJDH), Madrid, Spain), describing the molecular profile
of our AML patients, studied as part of a national Spanish project lead by the PETHEMA
group, and an analysis on which alterations could be significant to overall survival
(OS).
Aims: Analyse prognosis associated to molecular profile of new cases of AML
Methods: AML new diagnoses and relapses in FJDH from 2018 to 2020 were retrospectively
evaluated. The samples were referred to a central laboratory (12 de Octubre University
Hospital, Madrid, Spain) where NGS custom panels were run. Other significant variables
were analysed.
Results: 66 AML patients were evaluated with a mean age of 60yo (22-93). 191 mutations
were detected and most NGS were run at diagnosis 57 (86.4%). Most of the sample received
an intensive treatment 42 (63.6%), however only 25 (37.9%) patients achieved CR, 2
(3%) CRi, 6 (9.1%) +MRD, 4 (6.1%) PR and 26 (39.4%) were refractory. 11 (16.7%) patients
relapsed. Median OS was 13 months (CI 95% 7.9 – 18.1). Survival analysis confirmed
significant differences between the 3 ELN prognostic groups and some of the genetic
alteration (Figure 1).
In this series there was high incidence of secondary and adverse prognosis AML, as
expected from the high expression of ASXL1 (12.1%) and TP53 (15.2%) mutations. DNMT3A
mutations were most frequent (25.8%), followed by NPM1 (22.8%). We also found IDH
mutations (28.8%), which though less common could also be expected in adverse prognosis
AML. Our sample size was big enough that prognostic differences within each ELN classification
group were observed, as well as related to treatment. There was statistical significance
in OS between patients with wildtype vs mutated TP53 (8.9 vs 22.8; p=0.037) or JAK2
(3.8 vs 22.5, p=0.001), as supported by literature. Interestingly, we also found DNMT3A
mutated AML had a worse OS (24.1 vs 12.7; p=0.023). This may support recommending
HSCT in DNMT3A-mutated AML, as argued for TP53- and JAK2-mutated AML.
Although our OS graphs inclined towards favourable prognosis in the presence of NPM1
mutations (27.9 vs 19.9; p=0.12) and adverse prognosis with ASXL1 (11.1 vs 22.5; p=0.055),
the statistical analysis did not show significance in either case, probably due to
small sample size. RUNX1 did not seem to play a role in prognosis (19.1 vs 20.4; p=
0.741) which might be because most patients underwent HSCT, although current studies
seem to suggest RUNX1 mutations are independent to prognosis. Similarly, FLT3-ITD
mutations did not confer worse outcomes (25.5 vs 20.1; p=0.464). This may be due to
the mutation often being absent in disease relapse or progression, sometimes in relation
to targeted therapies. In our sample IDH mutations did not confer changes to OS (18.2
vs 21.9; p=0.707), though this could be explained by the limited availability of targeted
therapies in our country.
Image:
Summary/Conclusion: We found high incidence of high risk AML. Most of our data agree
with the published literature. Of note, RUNX1 doesn’t seem to play an important role
in prognosis. However, DNMT3A mutations alone and mainly as a group with TET2 and
ASXL1 were related to a bad outcome.
P427: THE SINGLE CELL T CELL LANDSCAPE OF AML PATIENTS POST ALLOGENEIC STEM CELL TRANSPLANTATION
A. Mathioudaki1,2,*, X. Wang3, R. Huth3, J. Zaugg4, C. Pabst3,4
1Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL);
2Faculty of Biosciences, Heidelberg University; 3Department of Medicine V, Hematology,
Oncology and Rheumatology, University Hospital Heidelberg; 4Molecular Medicine Partnership
Unit, EMBL Heidelberg, Heidelberg, Germany
Background: In Acute myeloid leukemia (AML), chemotherapy may lead to long-term remission,
but allogeneic stem cell transplantation (alloSCT) often remains the only curative
therapeutic approach, particularly in AML with high-risk genetics. However, not every
patient responds to alloSCT. Several hypotheses have been associated with therapy
failure, such as the incapability of T cells to recognize and eliminate residual leukemia
stem cells (LSCs), which thus escape the graft-versus-leukemia (GVL) effect. However,
the role of the T cells’ repertoire in therapy outcome still remains unclear.
Aims: Here, we investigated the impact of the bone marrow (BM) T cell composition
on the therapy outcome of AML patients after alloSCT.
Methods: We performed single-cell RNA sequencing (scRNA-seq) of T cells and hematopoietic
stem and progenitor cells (HSPCs) isolated from BM aspirates 100 days post alloSCT
of three patients in complete remission (CR) versus three suffering relapse (REL).
We investigated the prognostic impact of putative T-cell biomarkers in therapy outcome,
using flow cytometry analysis on an independent cohort.
Results: Using scRNA-seq, we identified 21 T cell and 9 HSPC populations and observed
differences in T cell population abundances between REL and CR. In particular, we
observed an enrichment of specific CD8+ T cell subsets in the CR group, while certain
CD4+ T cell subsets, such as regulatory T cells were enriched in REL patients. Furthermore,
we observed an increased T cell cytotoxicity signature in the CR group, that potentially
contributes to better clinical outcome. Within the genes upregulated in the CR condition,
we identified surface molecules linked to T cell cytotoxicity. Subsequent flow cytometry
analysis corroborated a link between these markers and better clinical outcome. Our
data suggest that the early bone marrow T cell signature post alloSCT might reflect
GVL-effects required to achieve long-term survival.
Image:
Summary/Conclusion: Understanding the role of T cell composition on therapy response
may offer novel approaches for both predicting prognosis as well as developing new
therapeutic strategies.
P428: SYK INHIBITION DRIVES DEEP RESPONSES IN A BIOMARKER GUIDED SUBSET OF AML ALONE
AND IN RATIONAL COMBINATIONS
M. McKeown1,*, L. A. Carvajal1, M. A. Day1, C. Noe1, P. Kumar1, J. DiMartino1, D.
Saffran1
1Kronos Bio, Inc., San Mateo, United States of America
Background: Spleen tyrosine kinase (SYK) acts as a key integrator of signals from
cell surface receptors containing an immunoreceptor tyrosine-based activation motif
to boost cellular proliferation. In acute myeloid leukemia (AML), SYK serves as a
relay to an oncogenic transcriptional regulatory network linked to NPM1, HOXA9 and
MEIS1.
The selective, orally bioavailable SYK inhibitor entospletinib (ENTO) has demonstrated
clinical activity and tolerability in HOXA9/MEIS1-driven AML. ENTO is currently being
investigated in a global Phase 3 trial, AGILITY (NCT05020665), in combination with
intensive induction/consolidation chemotherapy in patients with treatment-naive NPM1-mutated
(NPM1m) AML. Lanraplenib (LANRA) is a next-generation SYK inhibitor with similar potency
and selectivity to ENTO but with more favorable pharmacologic properties that is currently
being evaluated in combination with gilteritinib in patients with relapsed or refractory
(R/R) FLT3-mutated AML (NCT05028751).
Aims: To assess the potential of biomarker-guided responses to ENTO and LANRA in mutationally
defined subsets of AML and its activity in combination with other targeted agents
in translationally relevant models.
Methods: Full kinome profiling was run at fixed 1uM concentration followed by dose
response IC50 determination for top hits. AML and lymphoma cell lines were tested
for antiproliferative effects using CellTiter Glo (CTG) at 5 days. AML patient-derived
ex vivo models were tested in microtiter plate viability assays with CTG or in methylcellulose
colony viability assays for the NPM1m/FLT3-ITD model used in combination studies.
Results: Kinase selectivity profiling found >10-fold higher selectivity of ENTO and
LANRA for SYK vs other kinases with improved potency and selectivity compared with
the approved first-generation agent, fostamatinib. SYK inhibition with either agent
showed robust anti-proliferative activity in a panel of AML and lymphoma cell lines
with varying mutational backgrounds. In addition, synergy was observed when ENTO or
LANRA was combined with a Menin inhibitor in MLLr, FLT3-ITD cell lines. LANRA and
ENTO showed strong anti-proliferative activity in NPM1m AML patient samples validating
NPM1m as a patient selection biomarker for ENTO and LANRA. In an analysis of a large
public dataset of ex vivo patient samples, the presence of NPM1m and/or FLT3-ITD was
significantly predictive of response to ENTO. In an ex vivo model of NPM1m/FLT3-ITD
AML, consistent sub-micromolar response to LANRA was observed. Combinations with AML
standard-of-care and investigational agents, including azacytidine, showed at least
additive effects. Strong synergy with the JAK inhibitor ruxolitinib was consistent
mechanistically with a reporter model that demonstrated the ability of LANRA to block
activation of STAT5 in response to proleukemic paracrine signaling. Finally, robust
synergy across a range of LANRA concentrations was found when paired with venetoclax
and gilteritinib. These results prompted further testing with an in vivo, patient-derived
xenograft.
Summary/Conclusion: SYK is a promising therapeutic target for subsets of AML patients
defined by specific mutations. The highly predictive response for SYK inhibition with
concurrent NPM1m/FLT3-ITD mutations and synergy with gilteritinib observed in patient
derived models is supportive of its clinical evaluation in combination with a FLT3
inhibitor. Our work supports the rationale for an ongoing Phase 1b/2 study of LANRA
in combination with gilteritinib in patients with R/R FLT3-mutated AML.
P429: CLONALLY RESOLVED SINGLE-CELL MULTI-OMICS IDENTIFIES LEUKEMIA SURFACE ANTIGENS
A. K. Merbach1,*, S. Beneyto-Calabuig2, J.-A. Kniffka1, C. Szu-Tu2, C. Rohde1, M.
Antes1, M. Janssen1, A. Waclawiczek3, J. J. M. Landry4, V. Benes4, J. Anna5, M. Brough5,
B. Besenbeck1, J. Felden1, S. Bäumer6, M. Hundemer1, T. Sauer1, C. Pabst1, M. Scherer2,
S. Raffel1, L. Velten2, C. Müller-Tidow1
1Department of Medicine, Hematology, Oncology and Rheumatology, University Hospital
Heidelberg, Heidelberg, Germany; 2Centre for Genomic Regulation (CRG), The Barcelona
Institute of Science and Technology, Barcelona, Spain; 3Division of Stem Cells and
Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance; 4Genomics
Core Facility, European Molecular Biology Laboratory (EMBL); 5Institute of Human Genetics,
University Hospital Heidelberg, Heidelberg; 6Department of Medicine A, Hematology
and Oncology, University Hospital Muenster, Münster, Germany
Background: At the time of diagnosis, AML is a highly heterogeneous disease, usually
consisting of various epigenetically and genetically distinct pre-leukemic, as well
as leukemic subclones of various maturation stages that coexist in a competitive,
or even cooperative manner. This clonal heterogeneity and, in particular, the strong
resemblance of leukemic cells with their healthy counterparts presents a significant
challenge in the identification of cancer specific gene expression and surface markers,
both prerequisites for the development of targeted therapies. Existing approaches
to add clonal resolution to droplet-based single-cell RNAseq data use qualitative
information available from calls of individual mitochondrial or genomic single nucleotide
variants as well as copy number variants. These single cell mutational calls, however,
cannot be taken at face value, but need to be analysed through appropriate statistical
methods, which are currently missing.
Aims: Here we aimed at developing a novel method, CloneTracer, to add clonal resolution
to new or existing human single-cell transcriptomic or multiomic datasets to ultimately
allow identification of leukemia surface antigens through intra-sample comparison
of cancerous and healthy cells of matched differentiation state.
Methods: CloneTracer amplifies mitochondrial and selected nuclear SNVs, infers copy
number variation, and estimates clonal hierarchies and identities through a Bayesian
model that accounts for the noise properties of single-cell RNA-seq data.
Results: Through combination of clonal identities with information on surface markers
at single cell resolution, CloneTracer identified novel leukemia markers that enabled
quantification of the leukemic burden in clinical specimens and enrichment for leukemic
or healthy cells by FACS.
Image:
Summary/Conclusion: In summary, our data illustrate the benefits of clonally resolved
single cell multi-omics for identifying cancer specific surface antigens and potential
druggable targets by intra-patient comparisons.
P430: MOLECULAR CHARACTERIZATION OF CLINICAL RESPONSE IN NEWLY-DIAGNOSED ACUTE MYELOID
LEUKEMIA PATIENTS TREATED WITH IVOSIDENIB + AZACITIDINE COMPARED TO PLACEBO + AZACITIDINE
S. de Botton1,*, S. Choe2, D. Marchione2, P. Montesinos3, C. Recher4, S. Vives Polo5,
E. Zarzycka6, J. Wang7, G. Bertani8, M. Heuser9, R. Calado10, A. Schuh11, S.-P. Yeh12,
J. Hui2, S. Pandya2, D. Gianolio2, S. Daigle2, C. DiNardo13, H. Dohner14
1Institut Gustave Roussy, Villejuif, France; 2Servier Pharmaceuticals LLC, Boston,
United States of America; 3Hospital Universitari i Politècnic La Fe, València, Spain;
4Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France; 5Hospital
Universitario Germans Trias i Pujol-ICO Badalona, Josep Carreras Research Institute,
Badalona, Spain; 6Department of Hematology and Transplantology, Medical University
of Gdansk, Gdansk, Poland; 7Institute of Hematology & Hospital of Blood Disease, Tianjin,
China; 8ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; 9Hannover Medical
School, Hannover, Germany; 10Ribeirão Preto School of Medicine, University of São
Paulo, São Paulo, Brazil; 11Princess Margaret Cancer Centre, Toronto, Canada; 12China
Medical University, Taichung, Taiwan; 13The University of Texas M.D. Anderson Cancer
Center, Houston, United States of America; 14Ulm University Hospital, Ulm, Germany
Background: Acute myeloid leukemia (AML) is a disease with a dynamic mutational landscape;
6–10% of patients (pts) have somatic mutations in isocitrate dehydrogenase 1 (IDH1),
which can drive oncogenesis. Ivosidenib (IVO) is a potent oral targeted inhibitor
of mutant IDH1 (mIDH1). IVO 500 mg QD + azacitidine (AZA) 75 mg/m2 SC or IV for 7
days in 28-day cycles was shown to significantly improve event-free survival (HR=0.33
[95% CI 0.16, 0.69], p=0.0011), median overall survival (24.0 vs 7.9 months), and
complete remission + partial hematologic recovery rates (CR/CRh; 52.8% vs 17.6%) vs
placebo + AZA in the double-blind phase 3 AGILE study (NCT03173248) in pts with newly
diagnosed IDH1-mutated AML (ND-AML).
Aims: To assess the impact of IVO+AZA on IDH1-mutation clearance (IDH1-MC) and baseline
co-mutation analysis from AGILE.
Methods: Genomic DNA from bone marrow mononuclear cells (BMMCs) or peripheral blood
mononuclear cells (PBMCs), and/or bone marrow aspirate (BMA) were used for molecular
studies. IDH1-MC analysis on BMMCs was performed by BEAMing digital PCR (limit of
detection 0.02%-0.04%). BMA, BMMCs and PBMCs were utilized for co-mutational analysis
by next-generation sequencing, ACE Extended Cancer Panel (detection limit 2%). All
patients gave written informed consent.
Results: 146 pts were randomized: 72 to IVO+AZA; 74 to placebo+AZA. Median (range)
baseline mIDH1 variant allele frequency in BMMCs was 36.7% (3.1–50.5%) in the IVO+AZA
arm and 35.5% (3.0–48.6%) in the placebo+AZA arm. Updated IDH1-MC data (October 2021)
from 47 IVO+AZA and 32 placebo+AZA treated pts with at least 1 on-treatment sample
demonstrated IDH1-MC in 21/35 (60%) IVO+AZA pts achieving CR/CRh vs 4/11 (36%) placebo+AZA
pts. In CR/CRh pts with time points available after IDH1-MC, suppression of the mIDH1
was durable and IDH1-MC maintained in all subsequent samples in 17/17 (100%) IVO+AZA
treated pts and 1/3 (33%) placebo+AZA pts. Further analysis of baseline co-mutations
on 120 pts (IVO+AZA: n=58; placebo+AZA: n=62) showed that DNMT3A, SRSF2, and RUNX1
were the most frequent in both treatment arms. Importantly, comparison of CR/CRh and
non CR/CRh responses by cohort did not identify any single gene or pathway associated
with an inferior outcome in IVO+AZA pts compared to placebo+AZA pts (p<0.05, Fisher’s
Exact test). Several genes (DNMT3A, RUNX1, SRSF2, STAG2) and pathways (Differentiation,
Epigenetics, Splicing) were associated with improved outcomes with IVO+AZA, including
the RTK pathway, which was previously reported to be associated with primary resistance
to IVO monotherapy. Further analysis of patient subgroups, including R132 variants
(i.e., R132C vs R132S), will be presented.
Summary/Conclusion: These data suggest that improved clinical outcomes with IVO+AZA
are associated with sustained clearance of the mIDH1 clone including pts with disease
that harbor mutations implicated in resistance to IVO monotherapy (e.g., with RTK
pathway mutations).
P431: THE ROLE OF SMARCA4 IN HEMATOPOIESIS AND ACUTE MYELOID LEUKEMIA
F. Modemann1,2,*, L. Ramke1, J. Muschhammer1, L. Behrmann1, V. Thaden3, M. Schoof3,4,
C. Göbel3, S. Neyazi3,4, C. Bokemeyer1, J. Wellbrock1, U. Schüller3,4,5, W. Fiedler1
1Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf;
2Mildred Scheel Cancer Career Center, University Medical Center Hamburg-Eppendorf;
3Research Institute Children’s Cancer Center Hamburg; 4Department of Pediatric Hematology
and Oncology, University Medical Center Hamburg-Eppendorf; 5Institute of Neuropathology,
University Medical Center Hamburg-Eppendorf, Hamburg, Germany
Background:
SMARCA4 (SWI/SNF related, matrix associated, actin dependent regulator of chromatin,
subfamily a) is the central ATPase containing enzyme in the SWI/SNF- (switch/sucrose
non-fermentable) complex, which influences gene transcription by regulating chromatin
accessibility. In solid tumors, SMARCA4 mainly possesses tumor suppressive features.
However, in the setting of acute myeloid leukemia (AML), SMARCA4seems to play a different
role, acting as a proliferation stimulus for myeloid blasts.
Aims: The aim was to investigate the role of SMARCA4 in normal hematopoiesis and in
pathogenesis and maintenance of AML to evaluate the prognostic and potential therapeutic
relevance of this protein.
Methods: We screened 152 bone marrow samples of newly diagnosed adult AML patients
compared to 29 healthy bone marrow samples for SMARCA4 expression by using RT-qPCR.
SMARCA4 expression was correlated to overall and relapse free survival. We further
analyzed proliferation and colony-formation capacity of AML cell lines with lentivirally
transduced shRNA-mediated SMARCA4 knockdown or SMARCA4 overexpression induced by lentiviral
transduction. Two mouse models were generated: Firstly, to evaluate the impact of
SMARCA4 knockout on normal hematopoiesis, a transgenic mouse model with Cre-recombinase
mediated conditional knockout of SMARCA4 under the control of an Mx-1 promoter was
developed. Blood counts, bone marrow smear, colony formation assays, and RNA expression
profiles were analyzed. Secondly, a xenograft mouse model was generated by transplantating
the AML cell line MV4-11 with SMARCA4 knockdown or overexpression in NSG mice to analyze
overall survival.
Results: Expression of SMARCA4 was significantly higher in adult AML samples compared
to healthy CD34+ cells (p<0.0001). High SMARCA4 expression was associated with worse
overall (p=0.001) and relapse free survival (p=0.0073). SMARCA4 knockdown in AML cell
lines resulted in significantly lower proliferation rate in three different AML cell
lines compared to controls, whereas SMARCA4 overexpression led to higher proliferation
rates. Overall survival of mice transplanted with SMARCA4 overexpressing MV4-11 cells
was significantly worse than in the control group, whereas mice transplanted with
SMARCA4 knockdown MV4-11 cells lived significantly longer.
Transgenic SMARCA4 knockout mice showed significantly lower leukocyte, reticulocyte,
and thrombocyte counts and showed lower colony forming capacity compared to Cre-negative
control mice. Bone marrow smears showed aplasia of all cell lines in SMARCA4 knockout
mice. RNA-sequencing revealed 749 upregulated and 520 downregulated genes with a log
fold change ≥ +/- 1 in the bone marrow of SMARCA4knockout mice compared to Cre-negative
control mice. Relevant pathways according to REACTOME data base performing over representation
analysis included genes of generation of second messenger molecules, cell surface
interactions at the vascular wall, and immunoregulatory interactions between lymphoid
and non-lymphoid cells.
Image:
Summary/Conclusion:
SMARCA4 is upregulated in primary AML cells and is an unfavorable prognostic marker
in AML patients. SMARCA4 overexpression in AML cell lines leads to higher proliferation
rate and to a worse overall survival in transplanted mice. In normal hematopoiesis,
SMARCA4 knockout leads to decreased cell growth in bone marrow, highlighting the dependence
on SMARCA4 for proliferation of hematopoietic (stem) cells.
P432: RISK STRATIFICATION IN ACUTE MYELOID LEUKEMIA: EXTERNAL VALIDATION OF THE STELLAE-123
GENE EXPRESSION MODEL AND COMPARISON WITH CURRENT PROGNOSTIC SCORES
A. Mosquera Orgueira1, A. Peleteiro Raindo1,*, J. A. Diaz Arias1, M. Cid Lopez1, R.
Abal Garcia1, B. Antelo Rodriguez1, M. S. Gonzalez Perez1, J. C. Vallejo Llamas1,
M. M. Perez Encinas1
1Hematology, CHUS, Santiago de Compostela, Spain
Background: The identification of recurrently mutated genes and cytogenetic anomalies
has provided high prognostic and therapeutic significance in patients diagnosed with
AML. Currently, some of this information has been incorporated into risk stratification
guidelines, such as those of the European Leukemia Net (ELN). However, these guidelines
are based on cytogenetic analyses and a limited number of mutations, and they don’t
consider the genomic complexity of AML. Recently, we presented a new prognostic score
based on gene expression analysis (Stellae-123) which achieved high discriminative
power in survival prediction of AML patients, particularly among those bearing high-risk
mutations. It is necessary to evaluate this signature in new AML cohorts and to test
its performance compared with standard risk stratification scores.
Aims: To validate the prognostic precision and discriminative power of the Stellae-123
gene expression signature in external cohorts and to compare its performance with
standard prognostic scores: ELN-2017 in adult patients and clinical risk score in
pediatric patients.
Methods: RNA-seq data from 2 adult cohorts and one pediatric AML cohort was retrieved.
The BeatAML (N=334) and the AMLCG-2008 (N=199) cohorts consisted of adult AML patients,
whereas the TARGET AML cohort (N=144) was composed solely of pediatric patients. Gene
expression estimates (FPKM) were normalized between cohorts using ComBat. Common genes
were retrieved, and we selected those included in the Stellae-123 predictor, reaching
a total of 69 genes. Random forests were built to predict survival in the largest
cohort (BeatAML). The precision of the predictors was evaluated using cross-validated
time-dependent AUCs derived from cox models. In the particular case of BeatAML, gene
expression survival models were based on out-of-bag predictions in order to reduce
the risk of overfitting during the training phase of the model.
Results: We initially built a survival predictor using random forests based on the
expression of the 69 genes incorporated in Stellae-123. This predictor was trained
in the BeatAML cohort, and external validation was performed on the remaining two
cohorts. The model achieved c-indexes of 0.635, 0.645 and 0.598 in the BeatAML, AMLCG-2008
and TARGEL AML cohorts, respectively. We then evaluated the precision of this signature
to predict survival at 6 months, 1 year and 2 years after diagnosis using cross-validated
cox models. The results indicated that this model achieved greater precision than
the ELN-2017 and the pediatric clinical risk score in the majority of evaluated time
points (Figure 1).
Since age is a variable deeply associated with survival in AML, we evaluated the performance
of the models including this covariate. We observed clear improvements in AUCs for
the BeatAML and TargetAML, but not for the AMLCG-2008 cohort, a finding which is probably
related to the fact that these were fit adult patients recruited in a clinical trial.
Notably, we also observed an improved performance of the gene expression signature
plus age model compared with the ELN-2017 and pediatric clinical risk scores plus
age models, particularly in the prediction of survival within the first year after
diagnosis.
Image:
Summary/Conclusion: The Stellae-123 gene expression signature can predict overall
survival in AML with greater precision than the ELN-2017 and the pediatric clinical
risk score. Therefore, gene expression profiling emerges as a powerful tool to optimize
patient risk stratification. There is a growing need to standardize these tests for
clinical use.
P433: ADAPTER HOLIDAYS MAINTAIN IN-VITRO ADCAR-T CELL FUNCTIONALITY AGAINST AML
D. Nixdorf1,2,*, M. Sponheimer1,2, D. Berghammer2, N. Zieger1,2, L. Rohrbacher1,2,
S. Dapa3, C. M. Seitz4, K. Brandstetter5, M. von Bergwelt-Baildon1, H. Leonhardt5,
J. Mittelstaet6, A. Kaiser3, V. Bücklein1,2, M. Subklewe1,2
1Department of Medicine III; 2Laboratory for Translational Cancer Immunology, LMU,
Munich; 3R&D Department, Miltenyi Biotec GmbH, Bergisch Gladbach; 4Department of General
Pediatrics, University Children’s Hospital Tuebingen, Tuebingen; 5Department of Biology
II, LMU, Munich; 6R&D Department, Miltenyi Biotech GmbH, Bergisch Gladbach, Germany
Background: The rise of immunotherapies has sparked hope for improving the treatment
options of acute myeloid leukemia (AML). However, bispecific antibodies (AB) and CAR-T
cells demonstrated only moderate efficacy in clinical trials. The heterogeneous nature
of the disease requires more flexible, potent and safer approaches to make T-cell
based treatments available for AML patients. We have recently developed a novel, highly
modular adapter CAR-T cell (AdCAR) platform capable of targeting multiple antigens
via biotin-labeled adapter molecules (AMs) in the context of a specific linker (Seitz
et al., 2021).
Aims: Utilizing an AdCAR-T cell approach to overcome well described obstacles in AML
we evaluated several key parameters of our platform, including cytotoxicity, AM internalization
and serial targeting capability. Furthermore, we highlight the possibility of intermittent
(int.) AM dosing regimens (AM holidays) to delay AdCAR-T cell dysfunction due to continuous
(cont.) stimulation.
Methods: The cytotoxic capacity of AdCAR-T against AML cell lines and primary AML
(pAML) samples was evaluated in co-culture assays utilizing Fab-based AMs. Specific
lysis was assessed by FC after 48-72 h. Internalization kinetics of AMs were assessed
via surface staining of AML cells with AMs and secondary staining with anti-Biotin-PE
AB. The % surface bound AMs over time at 37 °C vs 4 °C was analyzed. For serial targeting
experiments the AMs were either added once, replenished every 3 days, or changed to
AMs with different target specificity. To mimic AM holidays
in vitro, cont. vs int. AM-exposed AdCAR-T were co-cultured with OCI-AML-3 (21 d vs
7 d AM exposure, followed by 7 d AM absence). Cytotoxic capacity, T cell proliferation
and cytokine secretion were assessed.
Results: AdCAR-T cells elicited high cytotoxicity against several AML cell lines utilizing
AMs of different specificity (AMs for CD33, CD123, CLL-1; IC50 = 2.1, 0.6, 1.9 ng/ml).
Against pAML similar data were obtained in an E:T and AM-concentration dependent manner.
We demonstrated feasibility of serial AM usage by performing long-term pAML/AdCAR-T
co-cultures. In comparison to one-time or low-dose cont. CD33 AM exposure, which led
to an outgrowth of pAML cells after 12 d, increasing AM concentrations and/or switching
the target antigen improved AdCAR-T cell efficacy. We further observed receptor-mediated
endocytosis of AMs (reduction of MV4-11 surface bound CD33 AM after 6 h = 81%) and
postulate that AM internalization is a common form of antigen sink in AM-based T-cell
recruiting therapies. Internalization of AMs reduces AM half-life and decreases cytotoxic
capacity of AdCAR-T cells. While limited AM levels can be overcome by repetitive dosing,
AM internalization potentially also contributes to faster off-switch dynamics. Importantly,
we observed that cont. AdCAR-T cell stimulation leads to T cell dysfunction (Fig 1)
in in vitro long-term efficacy studies. Strikingly, these effects can be mitigated
by implementing int. AM dosing regimens, shown by significantly increased T cell proliferation
(fold change d14: 1.9 vs 3.5), cytotoxic capacity (% specific lysis d14: 43 vs 64)
and IL-2 production (202 vs 823 pg/ml; all mean+/-SEM).
Image:
Summary/Conclusion: Taken together, AdCAR-T cells provide flexibility in target antigen
choice, safety through short half-life of the AMs and long-term functionality by mitigating
AdCAR-T cell dysfunction through AM holidays. Future trials need to consider the high
risk of CAR-T cell exhaustion upon cont. stimulation and AdCAR-T cells might pave
the way for rational design of more sophisticated treatment schedules.
P434: MITOCONDRIAL MCL1 REGULATES LEUKEMIC CELLS METABOLISM VIA DIRECT INTERACTION
WITH HEXOKINASE II. METABOLIC SIGNATURE AT ONSET PREDICTS OVERALL SURVIVAL IN AMLS’
PATIENTS
N. I. Noguera1,2,*, G. Catalano1,2, A. Zaza1,2, C. Banella3, T. Ottone2,4, E. Pelosi5,
S. Travaglini1,2, M. Divona6, M. I. Del Principe1, F. Buccisano1, L. Maurillo1, E.
Amatuna7, U. Testa5, A. Venditti1, M. T. Voso1,2
1Tor Vergata University; 2Fondazione Santa Lucia, Rome; 3Meyer Children’s University
Hospital, Florence; 4Tor Vergata University, Roma; 5Istituto Superiore di Sanità;
6Policlinico Tor Vergata, Rome, Italy; 7University Medical Center Groningen, Groningen,
Netherlands
Background: Metabolic reprogramming is a basic feature in cancer. Leukemia associated
cellular reprogramming, metabolic heterogeneity at onset and metabolic clonal evolution,
driven by therapy, are essential insights that still remain unclear
Aims: We compared Acute Myeloid Leukemias’ (AMLs) cells metabolism to hematopoietic
progenitors’’ and normal maturing bone marrow cells’, to acquire useful prognostic
information and to uncover actionable therapeutic targets.
Methods: Methods We analyzed fresh primary blast from 19 AML patients, hematopoietic
progenitors obtained from normal CD34+ cells differentiated to promyelocytes and granulocyte
and the MV4-11, OCI-AML2 and OCI-AML3 cell lines, using a Seahorse Bioscience XFe96
analyzer. We silence MCL1 by siRNA and evaluated the interaction between HK2, MCL-1
and VDAC by co-immunoprecipitation and confocal microscopy. We assessed expression
of MCL-1 and HK2 by western blot; HK2 by q-RTPCR.
Results: Primary AML blast cells feature a lower spare respiratory capacity (SRC)
(p=0.02) and lower glycolytic capacity (p=0.02) as compared to early progenitors/precursors
(EP/P) from cultured CB CD34+ cells at day 7 of culture (N7, mostly promyelocytes)
(Figure 1 a and b). Primary AML blast depend principally on fatty acids (p<0.05);
they display a great flexibility, switching to glucose or glutamine to meet their
energetic needs (Figure 1c). Consistent with the high adaptability of AML cells, and
the emergence of resistance to therapy. We could define two populations (cut off value
10 pmol/min/x105 cells): one with higher (22±12 pmol/min/x105 cells) and on with lower
(3±2 pmol/min/x105 cells) p<0.0001 levels of proton leak. The cases with higher proton
leaks levels presented a reduced, extremely short, overall survival (p=0.048). We
defined two SRC populations (cut off value 80 pmol/min/x105 cells): higher (124±47
pmol/min/x105 cells) and lower (52±25 pmol/min/x105cells) levels (p=0.0001). Interestingly
the cases with higher SRC showed a trend of reduced overall survival. Associating
high proton leak levels with high SRC the significance increases to p= 0.007. Considering
the Basal OXPHOS of the AML patient’s cells, we observed two populations higher (54±12
pmol/min/x105 cells) and lower (16±6 pmol/min/x105cells) levels (p=0.0001). In patients
with both high SRC plus high basal OXPHOS, the overall survival shortage is significant
(p=0.002), indicating that an high SRC associated to higher basal respiration in AML
blast cells confers greater aggressiveness and resistance to the therapy (Figure 1d).
Patients with high mitochondrial respiration had a significantly higher myeloid cell
leukemia 1 protein (MCL1) expression. We ascertained that MCL1 directly binds to Hexokinase
II (HK2) on the outer mitochondrial membrane (OMM) affecting its stability.
Image:
Summary/Conclusion: We demonstrate that high proton leak and high mitochondrial respiration
at onset, arguably with the concourse of MCL1/HK2 action, is significantly linked
with a shorter overall survival in AMLs’ patients. Our data describe a new function
of MCL1 protein in AMLs’ cells, forming a complex with HK2 co-localized to the voltage
dependent anion channel (VDAC) on the OMM, thus promoting glycolysis and OXPHOS, ultimately
conferring metabolic plasticity and promoting resistance to therapy. The lower spare
respiratory capacity and lower glycolytic capacity of AML patients’ blast respect
to normal early hematopoietic precursors suggest a therapeutic window to use glycolytic
and mitochondrial inhibitors in resistant AML patients.
P435: ANALYSIS OF MYELOBLAST IMMUNOPHENOTYPE PROFILE IN ACUTE MYELOID LEUKAEMIA AS
PREDICTOR FOR CLINICAL OUTCOME
M. Sobas1, M. Nowak1,*, D. Szymczak1, I. Andrasiak2, T. Wróbel1
1Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw
Medical Univeristy; 2Independent Researcher, -, Wrocław, Poland
Background: The European LeukemiaNet (ELN) classification, is currently widely accepted
risk stratification of acute myeloid leukaemia (AML). ELN stratification is based
on genetic abnormalities and divides AML in low, intermediate and high risk groups.
However, the outcome of AML patients, even within one classified group may be different.
Flow cytometry analysis of blast cells play an essential role in diagnosis of AML,
but the prognostic value of particular antigen expression is still unknown.
Aims: The aim of this study was to investigate the prognostic impact of the immunophenotype
of blast cells.
Methods: We performed a retrospective analysis on 146 AML patients (aged 21-65) who
were diagnosed and treated with intensive chemotherapy between 2017-2020. Allo-HSCT
recipients were labelled TPL1, the others - TPL0. Patients with acute promyelocytic
leukaemia were excluded from the study. Quantitative value (%) of particular antigen
(CD2, CD4, CD7, CD9, CD11b, CD11c, CD13, CD14, CD15, CD16, CD19, CD25, CD33, CD34,
CD36, CD38, CD41, CD45, CD56, CD61, CD64, CD65, CD71, CD73, CD96 CD117, CD123, HLA-DR)
expression was collected. ELN classification at diagnosis, cytogenetic, mutational
status (FLT3-ITD, FLT3-TKD, NPM1, CEBPA, CBFB-MYH11, RUNX1-RUNX1T1) and outcome (complete
remission (CR), primary resistance, relapse after achieving CR were recorded). Categorical
variables were presented as frequencies with percentages, median and interquartile
range (IQR) were used to describe continuous variables. Non-normally distributed variables
were analysed using Mann Whitney test and Kruskal-Wallis test. The Kaplan-Meier method
and Cox proportional-hazards model were used to estimate time-to-event variables and
determine hazard ratio (HR). All statistical tests were two-tailed with the significance
level set at α=0,05. The study was performed in accordance with the Declaration of
Helsinki.
Results: The baseline characteristics of the patients are presented in a table. The
median age at diagnosis was 54 (aged 21-65). CR rate was 73%. The median follow up
of 146 AML was 443 days. The median value of HLA-DR of relapsed (n=31, 21%) vs non-relapsed
AML (n=115, 79%) was higher: 100 (90-100) vs 90 (67-99); p=0.006. There was no relationship
between the HLA-DR expression (%) and ELN risk (p=0.705) nor mutational status (p>0.05).
The primary resistance was more common in patients with higher CD34 and lower CD38
levels. The median value of CD34 in resistant AML vs those who achieved CR was 93%
(22-99) vs 47% (0-91); p=0.017 and the median value of CD38 was 61% (31-93) vs 93%
(83-98); p=0.011. The median CD34 value was significantly higher in the adverse vs
favourable ELN-group: 79% (18-96) vs 2% (0-69), respectively (p=0.001) while the median
CD38 value was significantly higher in the favourable vs adverse ELN-group, 96% (88-99)
vs 86% (58-96), respectively, (p=0.005). Improved EFS was reported for decrease of
HLA-DR (HR=1.01; p=0.049) and increase of CD38 (HR=0.99; p=0.046) among TPL0 patients.
These variables are borderline during OS prediction for TPL0 - HLA-DR (HR=1.01; p=0.081)
and CD38 (HR=0.99; p=0.051).
Image:
Summary/Conclusion: Together with genetic mutations, immunophenotyping could be an
equivalent component of risk assessment. The levels of CD34 and CD38 turned out to
modify the first line treatment response and correlate with the ELN stratification.
Higher percentage of CD34 and lower of CD38 may characterise refractory to standard
intensive therapy AML, while higher HLA-DR may proceed relapse. Further, analyses,
on prospective trials, should be conducted to confirm these correlations.
P436: TARGETED DELIVERY OF T22-PE24-H6 TO CXCR4 POSITIVE CELLS FOR ACUTE MYELOID LEUKEMIA
TREATMENT
Y. Núñez Amela1,2,*, A. Garcia-León1,2, A. Falgàs1,2,3, N. Serna3,4,5, L. Sánchez-García3,4,5,
A. Garrido2,6, J. Sierra2,6, A. Gallardo1,7, U. Unzueta1,2,3,5, E. Vázquez3,4,5, A.
Villaverde3,4,5, R. Mangues1,2,3, I. Casanova1,2,3
1Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona; 2Josep Carreras
Leukaemia Research Institute (IJC), Badalona; 3CIBER de Bioingeniería, Biomateriales
y Nanomedicina (CIBER-BBN), Madrid; 4Institut de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona; 5Departament de Genètica i de Microbiologia, Universitat Autònoma
de Barcelona, Cerdanyola del Vallès; 6Department of Hematology, Hospital de la Santa
Creu i Sant Pau; 7Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona,
Spain
Background: Acute myeloid leukemia (AML) is a malignant heterogeneous group of hematological
diseases that originates from a hematopoietic progenitor with altered maturation capacity.
Recently, considerable advances have been made in the development of targeted therapies
for distinct molecularly defined subtypes. Although many AML patients initially respond
to current treatment, the majority of them relapse. Therefore, new targeted therapies
are needed to overcome drug resistance and improve treatment effectiveness. CXCR4
is a chemokine receptor that is expressed in over 20 cancer types, both hematologic
cancers and solid tumors. CXCR4 receptor and its ligand CXCL12 are key mediators of
the interactions between AML cells and the bone marrow (BM) microenvironment. Moreover,
CXCR4 is highly expressed in around 50% of AML patients. Thus, we have generated a
nanoparticle (T22-PE24-H6) that selectively delivers the exotoxin A from Pseudomonas
aeruginosa to CXCR4+ leukemic cells.
Aims: To assess the selectivity of T22-PE24-H6 cytotoxicity in CXCR4+ AML cell lines
as well as in BM samples from AML patients. In addition, we evaluated the antineoplastic
effect of the nanoparticle in a CXCR4+ AML disseminated mouse model.
Methods: OCI-AML-3, MONO-MAC-6 and HEL cell lines were characterized for CXCR4 expression
using flow cytometry and immunohistochemistry (IHC). The in vitro antineoplastic effect
of T22-PE24-H6 was determined by cell viability assays. Competition assays with the
CXCR4 antagonist AMD3100 were performed to demonstrate the CXCR4-dependent cytotoxicity
of the nanoparticle. In vivo effect was assessed in NSG mice intravenously injected
with 1x106 luminescent MONO-MAC-6 cells. Animals were treated with 5 μg T22-PE24-H6
or Buffer (166 mM NaCO3H pH 8) daily for 10 doses. Bioluminescence signal (BLI) was
monitored to quantitatively measure and evaluate in vivo disease progression. Animal
tissue samples were collected after mice euthanasia and used to analyze AML dissemination
and toxicity. CXCR4 expression levels and T22-PE24-H6 effect were also determined
in BM samples from 10 newly-diagnosed AML patients.
Results: T22-PE24-H6 nanoparticle demonstrated a potent in vitro anticancer effect
in MONO-MAC-6 cell line. The specific cytotoxic activity of the nanoparticle was CXCR4
dependent as demonstrated by performing competition assays with AMD3100. Furthermore,
T22-PE24-H6 intravenous administrations showed significant BLI reduction in a CXCR4+
AML disseminated mouse model compared to buffer-treated mice. In addition, no differences
between groups in mouse body weight, biochemical parameters or histopathological changes
in normal tissues were detected. Finally, T22-PE24-H6 causes a significant cell viability
reduction in AML patient samples with high CXCR4 expression. In contrast, the nanoparticle
has no effect in AML patient samples with low expression of CXCR4.
Summary/Conclusion: T22-PE24-H6 selectively kills AML cell lines and BM samples from
AML patients with CXCR4 overexpression. The nanoparticle also induces a high antineoplastic
effect in a CXCR4+ AML disseminated mouse model without detectable systemic toxicity.
These data strongly support that T22-PE24-H6 may obtain therapeutic benefit for AML
patients with high CXCR4 expression.
P437: SURVIVAL-BASED 4-GENE PROGNOSTIC INDEX IMPROVES THE ELN-RISK CLASSIFICATION
IN ACUTE MYELOID LEUKEMIA
C. A. Ortiz Rojas1,2,3,*, D. A. Pereira-Martins1,2,3,4, C. C. Bellido More5, J. R.
Hilberink4, J. L. Coelho-Silva1,3, I. Weinhäuser1,2,3,4, L. Yamamoto de Almeida1,3,
V. M. de Deus Wagatsuma1,3, C. Hassibe Thomé1, G. Aguiar Ferreira1, E. Ammatuna4,
G. Huls4, J. J. Schuringa4, E. Magalhães Rego1,2,3
1Center for Cell-Based Therapy, University of Sao Paulo, Ribeirão Preto; 2Hematology
Division, LIM31, Faculdade de Medicina, University of Sao Paulo, Sao Paulo; 3Department
of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University
of Sao Paulo, Ribeirão Preto, Brazil; 4Department of Hematology, Cancer Research Centre
Groningen, University Medical Centre Groningen, University of Groningen, Groningen,
Netherlands; 5Department of Pediatrics, Ribeirao Preto Medical School, University
of Sao Paulo, Ribeirão Preto, Brazil
Background: The European Leukemia-Net (ELN) criteria have been routinely used for
risk categorization in acute myeloid leukemia (AML), classifying patients into favorable,
intermediate and adverse-risk groups based on the presence of specific chromosomal
and molecular alterations (Döhner et al., 2017). Despite this classification, high
heterogeneity in the disease course is still a clinical barrier since leukemia can
progress differently than expected. In this sense, the identification of markers that
could improve the risk prediction is an unmet medical need.
Aims: We decide to perform a multicohort analysis of transcriptomic data to identify
a gene signature consistently associated with prognosis in AML patients.
Methods: Publicly available data of five AML cohorts treated with intensive chemotherapy
were included in this study: TCGA, n=122; BeatAML, n=230; HOVON (GSE6891), n=392;
NTUH (GSE71014), n=104; German (GSE37642), n=136. Also, we retrospectively included
38 patients diagnosed with AML, treated with intensive chemotherapy at the University
Medical Center Groningen, in order to validate our gene signature by qRT-PCR. Univariate
and multivariate Cox regression and area under the ROC curve (AUC) was applied separately
to each gene in each cohort, in order to filter genes associated with prognosis. Prognostic
indexes (PIs) models were generated by weighted sum using the pooled β coefficient.
Then, the best PI was evaluated in its ability of improving ELN-risk classification.
Results: We found seven genes whose expression levels were associated with overall
survival. After modeling possible PIs, we selected the PI that generates a better
model in combination with ELN classification by using the Akaike’s Criterion Information
(AIC) estimator. Thus, we selected a 4-gene prognostic index (4-PI) composed of CYP2E1,
DHCR7, IL2RA, and SQLE genes. Survival stratification by 4-PI resulted in 5-year survival
rates of 44.7 vs. 7.8% in the TCGA cohort, 28.5 vs. 0% in the BeatAML cohort, 47.7
vs. 26.6% in the HOVON cohort, 41.5 vs. 9.1% in the German cohort, and 88.7 vs. 44.1%
in the NTUH cohort. Next, after correction for confounders variables, the 4-PI showed
to be independently associated with prognosis, with an HR=3.94, 2.1, and 1.78 for
TCGA, BeatAML, and HOVON cohorts, respectively. Among the patient molecular features,
we found that inv(16) and dmCEBPA cases were more frequent in the low 4-PI, while
TP53 alterations were frequent in patients with a higher 4-PI. Next, we evaluated
the utility of the 4-PI in recategorizing patients from the ELN-risk subgroups in
a best-fitted risk category. In the BeatAML cohort, ELN2017-adverse patients represented
36.9% (n=85) of patients, with a median OS of 12.2 months, but in the integrated risk,
the adverse group increased to 38.7% (n=89) with a median OS of 10.3 months. We found
similar results after integration of the ELN2010 with our 4-PI in the TCGA and HOVON
cohorts. This power of stratification was maintained after applying the revised ELN
proposed by Herold et al., 2020. Next, in our validation cohort, when we integrated
the ELN2017 with the 4-PI, 14 patients were recategorized from the intermediate to
the adverse group, improving the difference between them (median OS=33.8 vs. 7.6 months).
Finally, we found a consistent enrichment of cholesterol metabolism related-pathways
in cases with high 4- PI in all the cohorts.
Summary/Conclusion: We conclude that our 4-PI outperforms currently used prognosis
predictors in AML and its incorporation into ELN categorization can improve risk stratification.
P438: ARYL HYDROCARBON RECEPTOR SIGNALLING IN NORMAL HAEMATOPOIETIC (HSC) AND LEUKEMIA
STEM CELLS (LSC): DIFFERENTIAL EFFECT OF AHR ANTAGONIST ON LSC COMPARED TO HSC
A. Oryshchuk1,*, R. Desai1, P. Kakadiya1, S. Bohlander1
1Molecular medicine and pathology, University of Auckland, Auckland, New Zealand
Background: Acute myeloid leukaemia (AML) is a devastating disease with poor prognosis.
It is driven by leukemia stem cells (LSCs), which are able to survive treatment and
drive relapse. This highlights the importance of characterizing LSCs. LSC-targeted
treatment strategies are limited because there are no known surface markers that are
specific for LSCs in every leukaemia. Targeting a signalling pathway that is altered
in all LSCs could be a novel, promising treatment strategy. The aryl hydrocarbon receptor
(AHR) pathway was shown to be critical for the self-renewal properties of normal haematopoietic
stem cells (HSCs).
Aims: We aimed to study the consequences of manipulating the AHR pathway in vivo in
a murine leukaemia model.
Methods: We used the following methods: murine bone marrow transplantation leukemia
model (MBMTLM) - CALM-AF10 with limiting dilution assay (LDA); competitive repopulation
assay (CRA) with LDA (CD45.2 donors into CD45.1 recipient mice), transcriptome analysis
(sequencing with the Illumina platform and analisys with the DESeq2). Treatment of
the cells was performed at various experiments with the AHR agonist, ITE, and an AHR
antagonist, CH-223191 (a vehicle control is DMSO).
Results: We used a murine retroviral transduction bone marrow transplantation leukemia
model (MBMTLM)) with the CALM-AF10 minimal fusion gene (CAMF) as the oncogenic driver,
to study the effect of AHR agonist (ITE) and an AHR antagonist (CH-223191) treatment
on LSCs. Interestingly, both AHR agonist and antagonist treatment affected LSC frequency
and survival of mice transplanted with treated cells. The LSC frequency as measured
by limiting dilution transplantation assays (LDA) was 3.7 times lower after agonist
treatment (1:163, blue survival curve in fig. 1, A) compared to a vehicle control
(1:44, DMSO, black survival curve)). Very interestingly, after antagonist treatment
the LSC frequency was so low that could not be measured as none of the mice transplanted
with antagonist-treated cells in the LDAs developed leukaemia (n=9, CH-223191 (red
curve in fig. 1, A)). Of note, our CAMF MBMTLM has a very high LSC frequency of about
1:4 in cells that were not cultured after thawing. We performed RNA-Seq on 90 samples
to determine changes in gene expression patterns after treatment of healthy bone marrow
cells and CAMF leukemia cells with the AHR agonist and antagonist. Preliminary differential
expression analysis with DESeq2 revealed differential expression of genes involved
in apoptosis and proliferation in leukaemia cells treated with CH-223191. As leukemia
did not develop in recipients of cells treated with the antagonist CH-223191, genes
differentially regulated in these cells might be critical and specific to LSC function
and will be candidate targets for LSC-specific treatments. To confirm that AHR antagonist
treatment affects LSCs differently than normal HCS, we performed competitive repopulation
assay (CRA). Our preliminary data (n = 12) show that treatment of HSCs with CH-223191
results in a steady increase in the engraftment over a 47-week period following transplantation
(coral bars, fig. 1, B), as opposed to the grafts from the ITE-treated arm (blue bars),
vehicle or untreated HSCs (black and green bars, respectively).
Image:
Summary/Conclusion: In summary, our data show that the LSCs react differently than
normal HSCs to changes in AHR signalling. These differences suggest it might be possible
to specifically inhibit LSCs while sparing normal HSCs, thus paving the way to develop
strategies to treat leukemia based on manipulating AHR signalling.
P439: ASSOCIATING EX VIVO DRUG SENSITIVITY WITH METABOLIC STATUS IDENTIFIES EFFECTIVE
COMBINATION STRATEGIES IN ACUTE MYELOID LEUKEMIA
D. A. Pereira-Martins1,2,3,*, E. Griessinger1, I. Weinhauser1,2, J. L. Coelho-Silva2,3,
D. R. Silveira4, L. Quek4, A. Erdem1, J. R. Hilberink1, E. V. de Paula5, S. T. Olalla
Saad5, E. Ammatuna1, G. Huls1, E. M. Rego2, A. R. Lucena-Araujo3, J. J. Schuringa1
1Department of Experimental Hematology, Cancer Research Centre Groningen, University
Medical Center Groningen, Groningen, Netherlands; 2Center for Cell Based Therapy,
University of Sao Paulo, Ribeirao Preto; 3Department of Genetics, Federal University
of Pernambuco, Recife, Brazil; 4Myeloid Leukaemia Genomics and Biology Group, School
of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom;
5Hematology and BloodTransfusion Center, University of Campinas, Campinas, Brazil
Background: Metabolic reprogramming is a hallmark of cancer, and acute myeloid leukemia
(AML) is no exception. Yet, we and others have shown that clear heterogeneity exists
in metabolic programming between genetically distinct AML subtypes and altered function
of mitochondria (mt) might be linked to chemoresistance in some patients.
Aims: Here, we evaluated the mitochondrial function of AML cells in more detail.
Methods: Retrospective analysis of the mtDNA content (mtDNAc) in a large cohort of
Brazilian patients with de novo AML (03 centers, n=482, age:18-65y) treated with the
3 + 7 scheme, and sex- and age-paired healthy donors (HD, n=308), revealed higher
mtDNAc in 34% of the AML samples when compared to the highest value of mtDNAc in the
HD group.
Results: High mtDNAc was independently associated with increased relapse risk. These
findings were confirmed in a validation cohort of AML patients treated with the 3 + 7
scheme at the UMCG in the Netherlands (n=92, age:18-65y). Clustering analysis using
label-free quantified proteome of sorted CD34+ cells from AML samples (n=30) associated
high mtDNAc with oxidative phosphorylation (OxPhos) metabolism and L-GMP leukemia.
Flow cytometry analysis for Hematopoietic progenitors (defined by CD34, CD38, CD123,
and CD45RA gated inside the CD45+ cells) and mitochondrial markers (Mitotracker Green/DeepRed
for mitochondrial mass/potential, respectively) in a set of primary AML samples (n=32),
showed an enhanced proportion of L-GMP cells and a positive correlation with the mitochondrial
potential (r=0.51) in AMLs with high mtDNAc. Consistently, oxygen consumption rate
(OCR) and lactate production measured by Seahorse was increased in primary AML blasts
(n=11) and AML cell lines (n=14) with high mtDNAc, suggesting increased energetic
metabolism. Multiomics analysis of AML cell lines (n=19) revealed a positive correlation
between mtDNAc and mitochondrial metabolism and apoptosis regulation. To evaluate
the role of mtDNAc during drug-induced apoptosis, we treated primary AML blasts (n=67)
with cytarabine (AraC, 100 nM) for 48h. We observed a negative correlation between
mtDNAc and apoptosis rate (r=-0.75). Moreover, the reduction of mtDNAc in AML blasts
(n=16) using shRNA targeting the POLG gene resulted in mixed phenotypes. POLG silencing
impaired cell proliferation and induced monocytic differentiation for AMLs with high
mtDNAc, while promoted cell proliferation in mtDNAclow AMLs, suggesting a completely
distinct dependency on OxPhos for these 2 groups of AMLs. Pharmacological treatment
of 9 AML cell lines with ETC complex I inhibitors (CIi; rotenone, 50 nM and metformin,
1 mM) in combination with AraC (10-500 nM) and Venetoclax (VEN, 10-1000 nM), resulted
in a reduction of mtDNAc and functional respiration in AML cells. Combination therapy
of CIi with AraC resulted in increased cell death in OxPhos - but not in glycolysis-driven
AML cells. In contrast, the combination of CIi+VEN resulted in enhanced cell death
in all AML cells, regardless of their metabolic state. Since metformin is an FDA-approved
drug, we assessed the metformin-induced apoptosis, in combination with AraC or VEN
in 64 primary AML samples ex vivo. Metformin treatment resulted in a reduction of
basal OCR of AML blasts (n=7). The metformin-induced apoptosis rate positively correlated
with mtDNAc (r=0.89), displaying additive effects with AraC- and VEN-induced apoptosis
in AML samples.
Summary/Conclusion: Overall, our findings suggest that mtDNA content can be used to
identify patients with a higher risk of chemoresistance and supports the idea of repurposing
metformin into AML therapy.
P440: THE COMBINATION VENETOCLAX PLUS MCL1 OVERCOMES DRUG RESISTANCE IN ACUTE PROMYELOCYTIC
LEUKEMIA
D. A. Pereira-Martins1,2,*, N. I. Noguera3, T. Ottone3,4, A. Diepstra5, C. Ortiz1,
I. Weinhauser1,2, G. Huls2, J. J. Schuringa2, E. M. Rego1, E. Ammatuna2
1Center for Cell Based Therapy, University of Sao Paulo, Ribeirao Preto, Brazil; 2Department
of Experimental Hematology, Cancer Research Centre Groningen, University Medical Center
Groningen, Groningen, Netherlands; 3Department of Biomedicine and Prevention, University
of Tor Vergata; 4Neuro-Oncohematology, Santa Lucia Foundation, I.R.C.C.S., Rome, Italy;
5Department of Pathology and Medical Biology, Cancer Research Centre Groningen, University
Medical Center Groningen, Groningen, Netherlands
Background: Although acute promyelocytic leukemia (APL) is the most favorable subtype
of acute myeloid leukemia, it remains a life-threating disease for high-risk patients.
Despite progress in APL with arsenic (ATO) plus retinoic acid (RA) as front-line treatment,
approximately 10-15% of patients will acquire resistance to ATO and/or RA and relapse.
Relapse after ATO/RA based therapy remains very challenging, and novel drug schemes
to overcome resistance are urgently needed. Analysis of bone marrow (BM) biopsies
of 15 consecutive APL patients at diagnosis, revealed a homogeneous staining for BCL2
in all cases. These results suggest BCL2 inhibition could be a therapeutic option
for resistant APL. In vitro treatment with the BCL2 inhibitor Venetoclax (VEN), resulted
in increased MCL1 expression, suggesting a resistance mechanism to this therapy.
Aims: Here, we investigated the effects of VEN+MCL1 inhibitor S64315 in APL cells
resistant to ATO/RA, and evaluated the effect of the combination therapy in vivo,
using a cell line derived xenograft (CLDX) model for APL with the ATO-resistant associated
mutation PML A216V.
Methods: Primary leukemic blasts from APL mice (n=4) and patients (age, 25-52y; n=10)
were treated with VEN (250-500nM), ATO, ATRA (1µM), and S64315 (10-50nM) monotherapies
and combinations and evaluated for cell survival. In addition, we evaluated the transcriptome
of 16 APL patients (age, 25-74y; 7 males) enrolled in the TCGA study regarding the
BCL2 expression. NB4/NB4R2 (RA-resistant) cell lines were transduced with PML A216V.
Additionally, U937 cells were transduced with the inducible PML-RARA system. After
induction of PML-RARA expression with Zinc for 0-24h, BCL2 protein levels were evaluated
by western blot. Additionally, RA/ATO treated (1µM) NB4 cells for 6-72h were evaluated
for BCL2/MCL1 levels. For the CLDX model, NB4 PML A216V cells were transplanted into
NSG mice, and after chimerism detection (hCD45+) in the peripheral blood, mice were
equally distributed into 4 groups of treatment: vehicle, VEN (25mg/kg/day), S64315
(20mg/Kg/day) and combination (n=5/group) for 21 consecutive days.
Results: Gene set enrichment analysis using the transcriptome of APL patients with
BCL2
high associated these patients with the terms “EPPERT_HSC_LSC” and “17_LSC”. These
results suggest that APL patients with high BCL2 expression differ in their metabolic
and proliferative state compared to patients with BCL2
low. Functionally, the induction of PML-RARA expression in U937 cells resulted in
increased BCL2 expression in a time-dependent manner (peak at 6h). Contrarily, treatment
with ATO/RA resulted in decreased BCL2 levels with upregulation of the MCL1 protein.
BH3 profile revealed that ATO/RA treated NB4 cells displayed an increase in the MCL1
anti-apoptotic dependency. Additionally, in vitro and ex vivo treatment of APL cells
(cell lines and primary blasts) revealed synergic activity of S64315, but not the
BCL2 inhibitor (VEN), in combination with ATO. Furthermore, treatment of ATO resistant
NB4 cells (by gradual exposure to ATO or by expression of the PML A216V) revealed
sensitivity of those cells to the combination VEN+S64315. Finally, in vivo CLDX model
revealed limited effect of VEN and S64315 monotherapy, while the combination was able
to effectively reduce the leukemic burden, resulting in increased overall survival
(vehicle:15 vs Combo:42 days).
Summary/Conclusion: The combination of BCL2 and MCL1 inhibition is a potential effective
treatment in ATO/RA resistant APL patients and could be clinically explored in this
category of patients with very poor prognosis.
P441: DIFFERENCES IN GENE EXPRESSION PATTERN AND IMMUNOPHENOTYPE OF BONE MARROW MULTIPOTENT
MESENCHYMAL STROMAL CELLS IN PATIENTS AT THE ONSET OF ACUTE LEUKEMIA AND AFTER ACHIEVING
REMISSION
A. Sadovskaya1,*, N. Petinati2, N. Kapranov2, N. Drize2, A. Vasilieva2, O. Gavrilina2,
E. Parovichnikova2
1Faculty of biology, Department of Immunology, Lomonosov Moscow State University;
2NATIONAL MEDICAL RESEARCH CENTER FOR HEMATOLOGY, Moscow, Russia
Background: The stromal microenvironment plays a key role in both maintaining normal
hematopoiesis and assisting malignant cells during leukemia development. In patients
with acute leukemia, the properties of multipotent mesenchymal stromal cells (MSCs)
are changed both at the onset of the disease due to interaction with tumor cells and
upon reaching remission due to chemotherapy. As a result, the ability of MSCs to maintain
normal hematopoiesis is reduced. A detailed study of these changes may allow the development
of rehabilitation measures for restoring the stromal microenvironment of patients.
Aims: The aim of this work was to study general changes in gene expression pattern
and immunophenotype of MSCs in patients with acute leukemia at the onset of the disease
and in remission.
Methods: The study included MSCs obtained from the bone marrow (BM) of 15 patients:
4 with ALL (1 male, 3 female, median age 26.5) and 11 AML (4 male, 7 female median
age 33), BM of 30 donors of appropriate age was used as control. All donors and patients
signed informed consent. MSCs were cultured by standard methods. Immunophenotypes
were studied by flow cytometry. Relative expression levels (REL) of several genes
were investigated by real time PCR.
Results: The study of the mean fluorescence intensity (MFI) of MSC markers revealed
a significant decrease in MFI of CD274 (PD-L1) in primary patients compared to healthy
donors and its absence on the cells of patients in remission. At the onset both MFI
of CD54 (ICAM-1) and the proportion of CD54+ MSCs were decreased. In remission these
parameters decreased even more, so the treatment reduced the adhesive capacity of
MSCs. The expression of CD105 was reduced at the onset of the disease and increased
after the end of treatment, but did not normalize.
In patients at the onset of the disease and in remission, compared with healthy donors,
the expression of IL1R1, PDGFRβ, ANG1 was reduced; expression of IL6, PPARγ, JAG1,
was increased (see Figure). Decreased level of IL8 and SOX9 normalized in remission.
The decrease in growth factor (GF) receptors PDGFRβ and IL1R1 (IL1β may act as a GF
for BM cells) in patients may be a result of negative feedback loops: GFs typically
act as auto- and paracrine regulators, and malignant cells often overexpress various
GF. The fact that these genes’ expression does not return to normal in remission may
be a result of damage from treatment. IL1β expression in patients was not different
from normal, but its increase in remission compared to onset implies stroma restoration
processes.
Another important effect is the alteration in MSCs’ interactions with stem cells:
JAG1 product helps to ensure stem cells’ quiescent state and bond with stroma. In
contrast, there is an increase in IGF1 expression at the onset, which can lead to
stem cell activation.
Alterations in the RELs of differentiation markers (SOX9, SPP1, PPARγ) indicate changes
in the predisposition of MSCs to differentiation. PPARγ is a multifunctional nuclear
receptor, and can inhibit proliferation. PPARγ upregulation may once again be a result
of negative feedback networks preventing MSCs’ uncontrolled proliferation in GF-rich
microenvironment.
Image:
Summary/Conclusion: The data suggest dramatic alterations in hematopoietic and stromal
cells interaction at the onset of acute leukemia that were not undone by achieving
remission. This is supported by decreased levels of endoglin and ICAM-1 and alteration
of gene expression pattern.
The materials supported by grants from the Russian Foundation for Basic Research project
19-29-04023 were used.
P442: SPINK2 PROTEIN EXPRESSION QUANTIFIED BY IMMUNOHISTOCHEMISTRY REFINES RISK STRATIFICATION
AND PREDICTS THERAPY OUTCOMES IN AML
H. A. Pitts1,*, C. K. Cheng1, J. S. Cheung1, M. K. H. Sun1, M. H. Ng1
1Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical & Cellular
Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong,
SAR, China
Background: Suboptimal prognostication, therapy refractoriness and relapse contribute
to the poor outcome of Acute Myeloid Leukemia (AML) patients. Leukemic stem cells
(LSC) are considered crucial drivers of relapse and therapy resistance. Clinicopathological
characterization of LSC-associated genes with convenient assessment tools such as
immunohistochemistry (IHC) is needed. Our initial screening of AML datasets for LSC-associated
genes identified Serine Protease Inhibitor Kazal type 2 (SPINK2) with high expression
in AML, particularly in LSC fractions. However, in-depth analysis of its clinicopathological
associations and prognostic utility in AML are lacking.
Aims: To assess SPINK2 expression in adult AML with IHC, and determine its clinicopathological
associations, prognostic impact and association with therapy response
Methods: We studied the expression of SPINK2 by IHC in 172 non-M3 adult AML patients
(median age:52yrs, range: 18-86yrs) treated at the Prince of Wales Hospital. The majority
(90.8%) were de novo type and 72.3% had intermediate-risk(IR) cytogenetics, with 88.5%
receiving Daunorubicin + Ara-C (DA 7 + 3) induction. SPINK2 expression was assessed
by 3 hematopathologists in a double-blinded manner and quantified by a composite score
(range: 0-16). Clinical data was collected for all cases.
Results:
SPINK2 staining in leukemic blasts was consistently cytoplasmic, and its protein expression
strongly correlated with mRNA levels by qPCR in a subset of 128 patients (r=0.716,
P<0.0001). The cohort was dichotomized at the median score 3, since this cut-off showed
strongest association with poor event-free survival (EFS) and overall survival (OS).
High SPINK2 (SPINK2
hi) patients accounted for 77/172 (44.8%) cases, and low SPINK2 (SPINK2
lo) patients for 95/172(55.2%).
SPINK2
hi associated with IR cytogenetics (P=0.014), normal karyotype (P=0.019) and NPM1
mutation (P<0.001), while SPINK2
lo with t(8;21) and CEBPA double-mutation (both P<0.001). Survival and treatment-response
analyses were performed on a subgroup of 137 patients who received DA 7 + 3 based
induction. SPINK2
hi patients had lower CR rates (73.8% vs 88.3%,P=0.028), higher 6-month relapse rates
(31.8% vs 9.1%,P=0.004), lower 5-yr RFS (25.8% vs 46.8%,P=0.004), EFS (16.6% vs 37.2%,P<0.001)
and OS (25.3% vs 51.2%,P<0.001). SPINK2 status also identified high risk patients
of the IR cytogenetic subgroup. Of these, SPINK2
hi patients had lower CR rates (68.6% vs 90.0%,P=0.013), higher 6-month relapse rates
(31.4% vs 6.9%,P=0.018), lower 5-yr RFS(27.0% vs 44.6%,P=0.018), EFS(15.4% vs 38.3%,P<0.001)
and OS(22.9% vs 51.5%,P=0.002). In this subgroup, median SPINK2 score was higher in
patients requiring ≥2 inductions to achieve CR vs. patients requiring 1 course (7
vs 2,P=0.009). Multivariate analyses in whole and IR cytogenetics cohorts showed the
poor prognostic effect of SPINK2
hi on EFS and OS independent of age, cytogenetic risk, mutations and achievement of
CR1, including stem cell transplantation given in CR (Table 1). In our NPM1
mut subgroup (N=41), SPINK2
hi status associated with poor EFS (HR:3.3,P=0.04) and OS (HR: 4.0,P=0.007) independent
of age and FLT3-ITD with high allelic ratio. Incorporation of SPINK2 with FLT3 status
refined ELN2017 risk definition, whereby 15/22 favorable-risk patients could be reclassified
as intermediate-risk.
Image:
Summary/Conclusion: High SPINK2 protein expression by IHC predicted increased chemoresistance
and early relapse risk in AML, and was an independent adverse prognostic biomarker,
particularly in patients with intermediate-risk cytogenetics and NPM1 mutations.
P443: IMPACT OF PHARMACOGENETIC VARIANTS ON CYTARABINE AND ANTHRACYCLINE EFFECT ON
OUTCOMES IN ADULT AML PATIENTS
Z. Pravdic1,*, M. Virijevic1,2, M. Mitrovic1,2, N. Pantic1, V. Gasic3, S. Pavlovic3,
N. Sabljic1, D. Pavlovic3, I. Marjanovic3, N. Suvajdzic Vukovic1,2
1Clinic of Hematology, UCCS; 2Faculty of Medicine, University of Belgrade; 3Institute
of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
Background: Acute myeloid leukaemia (AML) is primarily treated with combination of
cytarabine and anthracyclines. Despite high remission rates, the 5-year overall survival
(OS) is <50% and <20% for<60-year-old a ≥60-year-old patients, respectively. This
grim clinical outcome could be partly explained by the patients’ genetic variability
of proteins involved in metabolic paths of cytarabine and anthracyclines. Pharmacogenetic
variants of the main cytarabine membrane transporter SLC29A1 (solute carrier family
29 member 1), DCK (Deoxycytidine kinase) the first enzyme in cytarabine stepwise activation
process, genes coding main anthracycline efflux pump ABCB1 (ATP binding cassette)
and genes coding glutathione S-transferases GSTM1 and GSTT1, main cytosolic detoxifiers
od anthracycline-induced oxidative stress, are shown to influence clinical outcome
in AML patients.
Aims: to evaluate: 1) the effects of variants in pharmacogenes SLC29A1, DCK, ABCB1,
GSTM1 and GSTT1, on complete remission (CR) and relapse rate (RR), disease free survival
(DFS) and overall survival (OS), 2) the influence of demographic, laboratory and AML-related
parameters on CR, RR, DFS and OS.
Methods: 100 newly-diagnosed consenting adults (18-62 years old) diagnosed with AML,
except acute promyelocytic leukaemia in Clinic of Haematology UCCS from January 2015
to January 2018, were included in retrospective cohort. Patients received one or two
inductions ‘’3 + 7’’ cycles. In patients achieving CR either three consolidation cycles
with high/intermediate doses of cytarabine or allogenic SCT in selected patients were
performed. Demographic, standard laboratory and AML-related parameters (blood/bone
marrow blast percentage, cytologic, flow cytometry and genetic markers enabling ELN
risk stratification were collected from patients’ health records. Variants SLC29A1
rs9394992, DCK rs12648166, ABCB1 rs2032582 and GSTM1 and GSTT1 gene deletions were
detected by methodology based on PCR, fragment analysis and direct sequencing. The
study was approved by the Ethics Committee of the UCCS. The methods of descriptive
and analytic statistics were used, while survival analysis was done by the Kaplan-Meier
method using the Log-Rank test.
Results: 100 patients (53 males) were included in the study, with the median age of
51 (range 18-62). Median laboratory parameters were: WBC 15.5x109/L (range 1-348.8),
Hgb 97g/L (range 20-166), Plt 53.5 (range 1-422), LDH 249U/L (1-4169). CD34 was positive
in 56% patients. According to ELN 15, 55 and 30 patients were classified in favorable,
intermediate and adverse risk group, respectively. CR was achieved in 57 patients
(41 after the first and 16 after the second induction cycle). A total of 34% of patients
relapsed. Median DFS was 5.3 months (range 0.25-61.5), while median OS was 6 months
(range 0.3-75). There was a significant difference in median DFS between CC, CT and
TT genotype of SLC29A1: 13, 8 and 1.75 months, respectively (p=0.00037). Median DFS
was significantly decreased in null genotype of GSTM1 (9.5 (null) vs 18 months (non-null);
p=0.028). Significant difference in OS within ABCB1 genotypes TT, GG, GT and AG+AT
was registered: 13, 7, 6.5 and 3 months, respectively (p=0.0036). Other parameters
did not differ significantly.
Summary/Conclusion: Our results, regarding impact of variants of SLC29A1, GSTM1 on
DFS and impact of variant ABCB1 on OS in AML patients, are in line with previous studies.
Although further studies, with larger cohorts, are needed, these pharmacogenetic variants
show a potential to become prognostic markers in AML.
P444: IDENTIFICATION OF FUNCTIONAL GENETIC DEPENDENCIES IN CEBPA-MUTATED ACUTE MYELOID
LEUKEMIA
L. Proietti1,*, E. Heyes1, T. Eder1, T. Brandstoetter2, G. Manhart1, V. Sexl2, F.
Grebien1
1Institute for Medical Biochemestry; 2Institute of Pharmacology and Toxicology, University
of Veterinary Medicine Vienna, Vienna, Austria
Background: C/EBPα is a key myeloid transcription factor that regulates the switch
between proliferating uncommitted cells and terminally differentiated cells. The CEBPA
gene is mutated in 10-15% of AML patients. CEBPA lesions are divided into C-terminal
mutations that abrogate DNA binding and N-terminal frameshift mutations, which cause
expression of an N-terminally truncated isoform, termed p30. A large fraction of AML
patients carries biallelic CEBPA mutations. In these patients, the shorter C/EBPα
p30 variant is the only functional isoform of C/EBPα. While we and others have identified
molecular mechanisms underlying the development and maintenance of CEBPA-mutated AML,
a comprehensive analysis of vulnerabilities that is associated with this disease subtype
is lacking.
Aims: We aimed to characterize the functional genetic dependencies of AML with biallelic
CEBPA mutations.
Methods: We generated an immortalized AML cell line by serial transplantation of fetal
liver cells carrying biallelic CEBPA mutations (CNC,
C
ebpa
N-terminal/C-terminal) followed by prolonged culture of leukemic cells. After stable
expression of Cas9, we performed a genome-scale CRISPR/Cas9 loss-of-function screen
in CNC cells and compared the results to genome-wide screening results from the HPC7
hematopoietic multipotent progenitor cell line, which were used as a representative
of the healthy, untransformed state.
Results: Transplantation of CNC cells into C57BL/6 mice induced an aggressive Mac1+/Gr1+/c-Kit+
leukemia within 7 weeks, similar to the initial model (Bereshchenko et al, 2009).
This immuno-phenotype was preserved upon prolonged in vitro culture. Analysis of the
CRISPR/Cas9 screen in CNC cells identified 2488 genes whose mutational inactivation
was associated with reduced fitness - 438 of these genes are “core” essential, as
they have been annotated to be required for the survival of both cancer and normal
cells. In HPC7 cells, the loss of 1674 genes caused cell depletion, including 388
core essential genes. Intersection of screening results from CNC and HPC7 cells revealed
1290 commonly depleted genes, of which 380 were core essential. We identified 308
genes that were exclusively required for the proliferation and survival of CNC cells.
This list of high-confidence candidates is enriched for genes involved in mRNA metabolism,
DNA repair and signaling pathways. We are currently in the process of validating prioritized
target genes using a variety of approaches.
Summary/Conclusion: This approach allows the acquisition of deeper insights into the
molecular mechanisms underlying the development and maintenance of leukemia with CEBPA
mutations. Multi-layered validation of high confidence candidates will be performed
in vivo and in vitro to identify leukemia-specific vulnerabilities that can serve
as starting points for rational targeting strategies.
P445: CRYPTIC TRANSLOCATION T(5;11)(Q35;P15) RESULTING IN NUP98::NSD1 GENE FUSION
IN ADULTS WITH DE NOVO ACUTE MYELOID LEUKEMIA (AML)
S. Ransdorfova1,*, J. Markova1, M. Valerianova1, J. Brezinova1, M. Onderkova1, I.
Mendlikova1, L. Lizcova2, L. Pavlistova2, K. Svobodova2, S. Izakova2, A. Jonasova3,
C. Salek1, Z. Racil1, Z. Zemanova2
1Institute of Hematology and Blood Transfusion; 2Center of Oncocytogenomics, Institute
of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and
First Faculty of Medicine, Charles University; 31st Medical Department, General University
Hospital and First Faculty of Medicine, Charles University, Prague, Czechia
Background: The NUP98::NSD1 fusion, a product of the cryptic translocation t(5;11)(q35;p15.5),
is a recurrent genetic change in cytogenetically normal patients with AML. It occurs
most frequently in children (16%) and young (2%) AML patients, very rarely in adult
patients. The coexistence of internal tandem duplication of FLT3 gene (FLT3::ITD)
found in more than 70% of NUP98::NSD1 positive patients is always associated with
a poor prognosis and results in high frequency of induction failure.
Aims: The aim of this study was to determine the incidence of the NUP98::NSD1 fusion
gene in adults with AML and NUP98 rearrangement.
Methods: We examined the bone marrow cells of 268 newly diagnosed AML patients using
conventional karyotyping in combination with FISH (Abbott, MetaSystems) and mFISH/mBAND
(MetaSystems). We used RT-PCR followed by direct sequencing to detect fusion genes.
We processed the PCR product by ExoSAP-IT and directly sequenced on ABI Prism 310
genetic analyzer using the Big Dye Terminator kit v. 3.1.
Results: In nine out of 268 cases we identified rearrangement of the 11p13-15 region.
We confirmed t(5;11)(q35;p15.5) with NUP98::NSD1 gene fusion in four of them (4/268;
1.5%). Sequence analyses proved the NUP98-NSD1 transcript, arising from fusion of
NUP98 exon 12 with exon 6 in NSD1 gene, in all four patients (2M/2F; FAB M4/M5b; age
42, 54, 64 and 64 years). FLT3::ITD mutation was detected in all of them. Specific
primers and a probe have been designed to monitor minimal residual disease during
therapeutic treatment of the patients. Out of 4 patients, two died (median OS 9 months)
and two patients are alive (4 and 1 year after allogenic bone marrow transplantation).
Summary/Conclusion: Our study demonstrated the occurrence of t(5;11)(q35;p15.5) with
NUP98::NSD1 gene fusion also in adults over 60 years of age. We confirmed the NUP98::NSD1
fusion gene in 1.5% adults AML patients. With respect to the poor prognosis of the
patients with NUP98::NSD1 fusion gene, we suggest pre-screening of NUP98 gene rearrangement
using FISH in cytogenetically normal AML patients with confirmed FLT3::ITD mutation.
Supported by MH CZ-DRO-VFN64165, DRO-UHKT00023736.
P446: USE OF MIRNA EXPRESSION PATTERNS REVEALS ACTIVIN RECEPTOR 2 PATHWAY IN MEDIATING
CHEMO-RESISTANT AML
P. Reichelt1,*, S. Bernhart2, F. Wilke1, U. Platzbecker1, M. Cross1, G. Behre3
1Department of Hematology and Cell Therapy, University Hospital Leipzig; 2Interdisciplinary
center for bioinformatics Leipzig, Leipzig; 3Clinic for Internal Medicine I, Municipal
Hospital Dessau, Dessau, Germany
Background: Acute myeloid leukemia (AML) is a highly aggressive and heterogeneous
disease and the most prevalent type of acute leukemia in adults. Standard chemotherapy
using cytarabine and idarubicin/daunorubicin usually achieves remission, but this
is commonly followed by chemoresistance and relapse. The treatment of chemoresistant
AML remains a major challenge that will require personalized approaches.
Aims: With this study, we aim to establish NGS microRNA expression profiling and pathway
analysis to identify pathways that are regulated differentially between chemo-sensitive
and -resistant AML as potential targets for targeted therapy.
Methods: MicroRNA expression profiles were analyzed by NGS (next generation sequencing)
of chemo-resistant and -sensitive subclones of AML cell line HL60. Bioinformatical
identification of candidate deregulated pathways was performed using miRTarbase and
stringdb. Pathway activity in cell sublines was assessed by protein expression, proliferation
and flow cytometric analysis, using western blotting, phosphlow and MTS assays. Paired
samples of AML pre- and post- relapse were used to validate gene expression changes
by qPCR and analyses were extended to the publically available dataset of the AML
patient cohort of the cancer genome atlas (TCGA).
Results: Our microRNA based bioinformatic approach predicted 27 KEGG (Kyoto Encyclopedia
of Genes and Genomes) pathways to be represented differentially between resistant
and sensitive cells (FDR<0.05). Prominent among these were significant changes in
signaling pathways of the TGFβ ligand family. Cell culture experiments and protein
expression analysis confirmed both a higher sensitivity to TGFβ induced growth arrest
and increased abundance of TGFβ signaling proteins in chemo-resistant HL60 cells.
Consistent differences in TGFß pathway gene expression were also detected in AML patients
of the TCGA dataset. Furthermore, our data suggest a shift of the predominantly expressed
isoform of activin receptor 2B to activin receptor 2A in chemo-resistance. Analysis
of paired bone marrow samples from AML patients at diagnosis and first relapse showed
the latter to be associated with higher expression of TGFβ receptors and the TGFβ
target protein CDKN2B as well as significant upregulation of activin receptor 2A.
Image:
Summary/Conclusion: Here we show how miRNA screening approaches can be used to predict
deregulated cellular signaling pathways as potential targets for the therapy of chemoresistant
AML. MicroRNA based prediction of targetable pathways combined with database screening
identified the activin receptor 2A/B as a potential modulator of chemoresistance.
P447: BCL2-INHIBITION TARGETS LEUKEMIC STEM CELLS IN ACUTE MYELOID LEUKEMIA INDEPENDENT
OF MONOCYTIC BLAST POPULATIONS
S. Renders1,2,*, A.-M. Leppä1, A. Waclawiczek1, C. Reyneri1, M. Janssen2, E. Donato1,
J. Unglaub2, C. Pabst2, R. Schlenk2, M. Hundemer2,3, S. Raffel2, T. Sauer2, C. Müller-Tidow2,
A. Trumpp1
1DKFZ/ Hi-Stem; 2Department of Hematology, University Hospital Heidelberg, Heidelberg;
3MVZ Hämatologische Diagnostik, Heppenheim, Germany
Background: Treatment with Hypomethylating agents (HMA) in combination with the BCL-2
inhibitor Venetoclax (VEN) has recently become the standard of care for AML patients
unsuitable for intensive induction chemotherapy and shows superior results to treatment
with Azacitidine (AZA) alone (DiNardo et al., 2020, NEJM). However, upfront resistance
and relapse remain major obstacles.
It has recently been proposed that monocytic differentiation associated with increased
expression of antiapoptotic-protein MCL-1 predicts resistance to AZA/VEN treatment
in AML (Pei et al., 2020 Cancer Discovery). However, other studies did not find impaired
outcome in patients with monocytic AMLs treated with HMA/VEN (DiNardo et al., 2021,
Lancet Oncology; Stein et al., 2021 Blood Advances).
Aims: We aimed to understand apoptotic dependencies linked to differentiation states
in AML subpopulations in combined with clinical data to understand and treatment effects
and resistance upon HMA/VEN therapy.
Methods: We performed retrospective analysis of >50 patients at Heidelberg University
hospital to identify risk factors of treatment failure. For functional investigation,
we studied 24 AML cell lines and generated Xenografts in NSG mice of 12 AML patients
to study mature and leukemic stem cell (LSC)-like primary AML subpopulations. We characterized
these samples further by RNA sequencing and BCL-2 family level assessment. To functionally
validate our findings, we performed ex vivo drug treatment and subpopulation specific
BH3 profiling.
Results: In our clinical cohort the only factors associated with HMA/VEN resistance
were previous myelodysplastic syndrome (MDS)/myeloproliferative neoplasm (MPN) and
complex karyotype, but not monocytic differentiation. The CD64+CD11b+, mature blast
population making up >50% of leukemic cells in monocytic and <20% in primitive samples
showed high levels of resistance to AZA/VEN therapy in both primitive and monocytic
leukemias but did not engraft when transplanted into NSG mice, arguing they do not
contain LSCs. In contrast, we found immature GPR56+ LSCs to be sensitive to AZA/VEN
treatment irrespective whether they were derived from monocytic or primitive primary
AMLs. LSCs from monocytic and primitive AMLs initiated disease in NSG mice, highlighting
that targeting LSCs is essential to treat AML. Next, we investigated expression of
BCL2, MCL1 and BCL-xL levels in the same primary patient samples and observed high
MCL1 expression in monocytic AML samples. However, MCL1 expression predominantly derived
from the mature population whereas LSCs expressed robust levels of BCL2 but comparatively
low levels of MCL1, independent of whether monocytic or primitive AMLs were analyzed.
Additionally, transcriptomes of LSCs from either monocytic or primitive samples did
not display significant differences. We next performed BH3 profiling and found LSC
to be dependent on BCL2 and mature blasts on MCL1 in all samples studied. In line,
ex vivo HMA/VEN treatment specifically eliminated LSCs. We further monitored blast
clearance in longitudinal patient samples and found robust loss of LSC within hours
in patients, while mature blasts persisted for days.
Summary/Conclusion: We find that LSCs, independent of the overall mature blast content,
are sensitive to HMA/VEN treatment and highlight that genetic risk factors and disease
history are relevant predictors of treatment failure.
P448: PROGNOSTIC IMPACT OF SOMATIC CEBPA BZIP DOMAIN MUTATIONS IN ACUTE MYELOID LEUKEMIA
F. G. Ruecker1,*, A. Corbacioglu1, F. Theis1, M. Christopeit2, U. Germing3, G. Wulf4,
M. Abu Samra5, L. Teichmann6, M. Lübbert7, M. W. Kühn8, M. Bentz9, J. Westermann10,
L. Bullinger10, V. I. Gaidzik1, E. Jahn1, M. Gröger1, S. Kapp-Schwoerer1, D. Weber1,
F. Thol11, M. Heuser11, A. Ganser11, H. Döhner1, K. Döhner1
1Department of Internal Medicine III, University Hospital of Ulm, Ulm; 2Medical Clinic,
Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University
Hospital Tübingen, Tübingen; 3Department of Hematology, Oncology and Clinical Immunology,
Heinrich Heine University, Düsseldorf; 4University Medical Center Göttingen, Göttingen;
5Department of Internal Medicine IV, University Hospital of Gießen, Gießen; 6Department
of Medicine and Polyclinic III, Bonn University Hospital, Bonn; 7Department of Hematology,
Oncology and Stem Cell Transplantation, University of Freiburg Medical Center, Freiburg;
8Department of Hematology, Medical Oncology and Pneumology, University Medical Center
Mainz, Mainz; 9Department of Internal Medicine III, Hospital of Karlsruhe, Karlsruhe;
10Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine,
Berlin; 11Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation,
Hannover Medical School, Hannover, Hannover, Germany
Background: Mutations of CEBPA (CEBPA
mut) are present in ~5-10% of newly diagnosed adult acute myeloid leukemia (AML),
and approximately half of the patients (pts) exhibit biallelic mutations (CEBPA
bi). CEBPA
bi defines a distinct entity within the 2016 WHO classification and is categorized
as favorable in the 2017 risk stratification by the European LeukemiaNet. CEBPA
mut can be divided into basic leucine zipper domain (bZIP) or transcription activation
domains (TAD) mutations, respectively. Recent studies have demonstrated CEBPA
bZIP mutations, in particular in-frame mutations (CEBPA
bZIP-inf), to be associated with favorable outcome, regardless of mono- or biallelic
status.
Aims: To evaluate the prognostic impact of CEBPA
bZIP, in particular CEBPA
bZIP-inf mutations in AML.
Methods: Investigating a cohort of 454 intensively treated CEBPA mutated AML pts entered
into the AMLSG BiO Registry study (NCT01252485).
Results: Of the 454 pts, 223 had CEBPA
bi and 231 monoallelic CEBPA mutations (CEBPA
sm) affecting bZIP in 78 pts or TAD in 153 pts. Genotypes differed significantly with
regard to clinical and genetic features: CEBPA
bi pts were younger than CEBPA
smbZIP and CEBPA
smTAD (median age in yrs: 52 vs 59 vs 60; P<.001), had a higher rate of de novo AML
(97% vs 81% vs 84%; P<.001), lower platelet counts (median G/l 36 vs 57 vs 52; P<.001),
higher peripheral blood (PB) blast counts (median 71% vs 60% vs 44%; P<.001), and
showed an inverse correlation with FLT3 internal tandem duplication (FLT3-ITD) and
NPM1 mutation (NPM1
mut) (6% vs 21% vs 35% and 0% vs 26% vs 50%; P<.001 each).
Outcome analysis revealed a significant improved overall (OS) and event-free survival
(EFS) for CEBPA
bi with no difference between CEBPA
smbZIP and CEBPA
smTAD (5-year OS: 62% vs 42% vs 51%; P<.001, 5-year EFS: 47% vs 27% vs 43%; P<.022).
Subgroup specific analysis within CEBPA
smbZIP revealed an improved outcome for CEBPA
bZIP-inf pts (n=46) (5-year OS: 52% vs 29%; P=.001, and 5-year EFS: 34% vs 18%; P=.018).
To further address the impact of CEBPA
bZIP-inf, pts were categorized as CEBPA
bZIP-inf (n=250), irrespective of the allelic status, vs all others (CEBPA
other) (n=204). CEBPA
bZIP-inf pts were younger (median age in yrs: 52 vs 62; P<.001), had a higher rate
of de novo AML (97% vs 81%; P<.001), lower platelet counts (median G/l 36 vs 53; P<.001),
higher white blood cell (WBC) (median G/l 25.4 vs 16.1; P=.029), and higher PB blast
counts (median 72% vs 46%; P<.001); FLT3-ITD and NPM1
mut were less common in CEBPA
bZIP-inf pts (9% vs 30% and 3% vs 44%; P<.001 each). CEBPA
bZIP-inf exhibited a significant improved OS (5-year OS: 60% vs 48%; P<.001) and EFS
(5-year EFS: 47% vs 37%; P=.007); in multivariate Cox models for OS and EFS, including
allogeneic hematopoietic cell transplantation (HCT) in first complete remission as
time-dependent covariate, age (HR: 1.50; P<.001), WBC (HR: 1.40; P=.018), and adverse
cytogenetics (HR: 2.33; P=.010) were unfavorable factors for OS, whereas NPM1
mut (HR: 0.63; P=.040), HCT (HR: 0.46; P=.010), and CEBPA
bZIP-inf (HR: 0.59; P=.007) revealed as favorable. For EFS, age (HR: 1.19; P=.002),
WBC (HR: 1.30; P=.028), and FLT3-ITD (HR: 1.83; P=.003) were unfavorable, whereas
NPM1
mut (HR: 0.65; P=.028) and HCT (HR: 0.33; P<.001) were favorable.
Summary/Conclusion: In this cohort of 454 CEBPA mutated adult AML pts, CEBPA
bZIP-inf was associated with specific clinical and genetic characteristics. Furthermore,
CEBPA
bZIP-inf pts had a significantly superior outcome irrespective of the allelic status.
This study confirms recent findings suggesting a prognostic role of this mutation
type.
FGR and AC contributed equally
P449: NOVEL, FIRST-IN-CLASS, SOMATIC IDH2 DELETION-INSERTION VARIANT C.516_518DELINSTGC
CONFERS BORDERLINE PHENOTYPE AND DOES NOT SHOW SUSCEPTIBILITY TO ENASIDENIB IN-VITRO
L. Ruhnke1,*, D. M. Poitz2, S. Herold3, S. Dressler4, C. Dill1, M. Peitzsch2, U. Oelschlägel1,
J. Frimmel1, D. Kunadt1, T. Kretschmann1, H. Altmann1, F. Stölzel1, C. Röllig1, T.
Chavakis2, G. Baretton3, K. Schäfer-Eckart4, M. Bornhäuser1
1Department of Internal Medicine I; 2Institute of Clinical Chemistry and Laboratory
Medicine; 3Institute of Pathology, University Hospital Dresden, Dresden; 4Department
of Internal Medicine V, Nuremberg Hospital North, Paracelsus Medical University, Nuremberg,
Germany
Background: Mutations in IDH2 are among the most common genetic alterations in patients
with acute myeloid leukemia (AML). So far, only substitution variants (also known
as single nucleotide variants (SNVs)) - in particular affecting the hotspots at codon
Arg140 and Arg172 - have been identified.
.
Aims: Here we report and functionally characterize a novel, first-in-class, somatic
IDH2 deletion-insertion (delins) variant affecting codons Arg172/His173 identified
in a patient diagnosed with AML with myelodysplasia related changes (AML-MRC).
Methods: The novel delins variant was identified performing amplicon-based targeted
next generation sequencing (NGS), Sanger sequencing was used for verification. For
functional evaluation high-performance liquid chromatography tandem mass spectrometry
(LC-MS/MS), global DNA methylation and DNA hydroxymethylation assays and in-vitro
drug testing was performed; AML patients without evidence of IDH1 or IDH2 mutations
and patients with IDH2
Arg172 mutation served as negative and positive controls, respectively.
Results: A 63-year-old male patient (unique patient number (UPN) 1) was diagnosed
with AML-MRC. Further work-up revealed an aberrant karyotype (47,XY,+11/ 46,XY,der(7)t(7;11)(q31;q12)/
46,XY), a partial tandem duplication of KMT2A (KMT2A-PTD) as well as variants in IDH2,
DNMT3A and NRAS (Figure 1.) (no CBFB::MYH11, PML::RARA or RUNX1::RUNX1T1 fusion, no
ASXL1, CEBPA, FLT3, NPM1, RUNX1 or TP53 mutation) (Figure 1A). He received induction
therapy with liposomal daunorubicin/cytarabine, achieved CR and underwent allogeneic
hematopoietic cell transplantation.
Of note, the IDH2 variant identified was a novel three base-pair deletion-insertion
variant (c.516_518delinsTGC) leading to the replacement of amino acids arginine and
histidine at codon 172 and 173 for serin and alanine (p.Arg172_His173delinsSerAla),
variant allel frequency was 44%. The novel delins variant could be validated performing
Sanger sequencing; parallel Sanger sequencing of buccal saliva DNA ruled out germline
origin (Figure 1B). Since there are no valid in-silico prediction tools for delins
variants, we decided to assess the potential pathogenicity of the novel alteration.
First, we applied LC-MS/MS to evaluate 2-hydroxyglutarate (2-HG) levels as compared
to BM-MNCs obtained from IDH1/IDH2
WT AML patients (UNP2-4) and IDH2MUT AML patients (UPN5-10). In line with previous
reports, 2-HG levels were increased in patients with IDH2 mutations as compared to
their IDH1/IDH2
WT counterparts (1000-fold). However, in UPN1 only slightly elevated 2-HG levels were
found (10-fold) (Figure 1C); no differences in isocitrate or ketoglutarate levels
were seen (data not shown). Similar results were obtained when performing DNA methylation
and DNA hydroxymethylation studies. While the percentage of methylated DNA in UPN1
was comparable to the amount of 5-methylcytosine (5-mC) seen in the IDH2 mutated group
(UNP5-10), 5-hydroxymethylcytosine (5-hmC) levels in UPN1 tended towards those observed
in the IDH2
WT cohort (UPN2-4) (data not shown). Finally, we performed in-vitro drug testing to
evaluate response to the IDH2 inhibitor (IDH2i) enasidenib. Interestingly, enasidenib
did not affect granulocytic differentiation of BM-MNCs as assessed by CD11b, CD15
and CD16 flow cytometry (Figure 1D).
Image:
Summary/Conclusion: Here, we report a novel, somatic delins variant in IDH2, which,
albeit affecting the known Arg172 hotspot, does not show susceptibility to the IDH2i
enasidenib in-vitro. Our report argues for functional characterisation of novel variants
to ensure a valid biomarker-driven therapy in AML patients.
P450: PRECLINICAL AND CLINICAL SIGNS OF RVU120 EFFICACY, A SPECIFIC CDK8/19 INHIBITOR
IN DNMT3A MUTATION POSITIVE AML AND HR-MDS
U. Pakulska1, N. Angelosanto2, M. Mikula3, H. Nogai2, R. Dudziak2, C. Abboud4, M.
Obacz1, K. Goller1, M. Cybulska3, M. Mazan1, M. Kozakowska1, T. Rzymski1,*
1R&D; 2Clinical Department, Ryvu Therapeutics, Kraków; 3Maria Skłodowska-Curie Institute
- Cancer Center, Warsaw, Poland; 4Washington University School of Medicine in St.
Louis, St Louis, United States of America
Background: CDK8 and its paralog CDK19 regulate transcription as a part of mediator
complex, that links enhancers with core promoters. AML likewise many other cancers
highjack CDK8 and CDK19 to maintain undifferentiated state and prevent apoptotic cell
death. First CDK8/CDK19 inhibitor RVU120 has reached clinical development phase Ib
(NCT04021368) in AML and HR-MDS patients.
Aims: It is now critical to establish relationship between preclinical and clinical
efficacy results, and molecular characteristics in order to identify actionable markers
predicting response to CDK8/CDK19 inhibitors.
Methods: Association of molecular profiles with responses to RVU120 has been performed
on genetically annotated AML PDCs, followed by flow cytometry and bioinformatic analysis.
PDCs were implanted intravenously into NSG-SGM3 mice and after disease onset animals
were treated orally with RVU120. Profiling of transcriptional response to RVU120 in
DMNT3A mutant cells has been performed by RNA-seq. The First in Human study of RVU120
is currently active enrolling R/R AML or HR-MDS patients, in a dose escalation design
aimed at exploring safety/tolerability and identify the RPD2. RVU120 is administered
orally, each other day for 7 total doses per 21 days cycle, until disease progression/unacceptable
toxicity. Response to the study drug is assessed according to Dohner 2017 criteria.
Results: Screening of AML PDC against RVU120 indicated high anti-cancer efficacy in
>40% of tested samples. Correlation of efficacy with genetic profile of samples indicated
specific enrichment of DNMT3A or NPM1 mutants in responder group. These results were
further corroborated in disseminated PDX AML model, showing complete clearance of
DNMT3A and NPM1 mutation- positive blasts and recovery of murine BM in animals treated
with RVU120. Transcriptomic analysis of DNMT3A and NPM1 positive AML cells indicated
distinct transcriptomic profiles, characterized by high expression of homeobox genes,
involved in determination of cell faith. Immunophenotyping of responder cells indicated
elimination of lineage committed blast cells.
At the date of this abstract submission, 13 patients have been enrolled into CLI120-001
trial, including 2 patients with DNMT3A mutations. Notably, first R/R AML patient
that achieved CR was positive for DNMT3A and NPM1 mutations. At study entry this patient
was progressing with pancytopenia, 65% BM blasts and extramedullary localization of
leukemia. BM showed a complete clearance of blasts at the end of 1st cycle of 75mg
dose, followed by hematological recovery and signs of monocytic differentiation. Importantly,
monocytic differentiation is observed as a pharmacodynamic effect of RVU120 in preclinical
studies. Skin leukemia lesions improved gradually up to a CR in cycle 7. Second DNMT3A
mutation patient with HR-MDS, escalated from 50 to 75 mg dose from cycle 7, continues
treatment at the cycle 24 with SD and erythroid hematological improvement.
Summary/Conclusion: AML PDCs with DNMT3and NPM1 mutations show differential sensitivity
to RVU120 treatment both in vitro and in vivo. Anti-cancer efficacy of RVU120 was
associated with transcriptomic reprogramming and lineage commitment. Preliminary evidence
of clinical response to RVU120 has been also shown in R/R AML and HR-MDS patients
positive for DNMT3A mutations. Further molecular studies in greater number of patients
under RVU120 treatment are ongoing and will provide evidence for predictive markers
of response to RVU120 in AML.
P451: PRMT2: AN ANTI-INFLAMMATORY EPIGENETIC FACTOR INVOLVED IN ACUTE MYELOID LEUKEMIA
AGGRESSIVENESS
C. Sauter1,*, J. Simonet1, C. Fournier1,2, M. Mounier1,3, M. Rebourgeon1, L. Brignoli1,
A. Aznague1,2, A. Largeot1, Y. Hérault4, G. Sauvageau5, F. Guidez1, M. Callanan1,2,
J.-N. Bastie1,6, L. Delva1, R. Aucagne1,2
1UMR 1231 Inserm, Équipe Epi2THM, Équipe Labex LipSTIC, Université de Bourgogne Franche-Comté;
2Unité d’Innovation en Génétique et Épigénétique en Oncologie (IGEO), Plateforme CRISPR
Genomics (CRIGEN), CHU François-Mitterrand; 3Registre des hémopathies malignes de
Côte d’Or, Université de Bourgogne Franche-Comté, Dijon; 4Institut de Génétique et
de Biologie Moléculaire et Cellulaire (IGBMC), Département of de Génomique Fonctionnelle,
UMR 7104 CNRS, UMR 964 Inserm, Université Louis Pasteur, Collège de France et Institut
Clinique de la Souris (ICS), Illkirch, France; 5Laboratoire de Génétique Moléculaire
des Cellules Souches, Institut de Recherche en Immunologie et Cancer, Université de
Montréal, Montréal, Canada; 6Service d’Hématologie Clinique, CHU François-Mitterrand,
Dijon, France
Background: Protein Arginine Methyltransferases (PRMTs) are epigenetic factors involved
in several cellular processes including regulation of gene expression through methylation
of histone tails. Overexpression or overactivity of the two major PRMTs (1 and 5)
have been previously identified in Acute Myeloid Leukemia (AML) patients and their
biological or pharmacological inhibition leads to a decreased leukemia progression
in mice (PRMT1, 4, 5). Among the nine members of this protein family, PRMT2 has been
less well characterized. It has been shown to be overexpressed in glioblastoma and
is responsible for the H3R8me2a epigenetic mark leading to transcription activation
of target genes. In addition, PRMT2 has also been demonstrated to have a role in the
inflammatory pathway but its effect is not fully understood yet. Moreover, it is well
known that severe chronic inflammation can contribute to the development of certain
cancers. Exacerbated inflammatory responses and aberrant myeloproliferation are thus
interconnected.
Aims: We are investigating the role of PRMT2 in the development of AML and we aim
to discover its precise role in the control of inflammatory processes.
Methods: We used a PRMT2 knockout mouse model (Prmt2KO), AML patient cohorts of Dijon
(Dijon university hospital, France) and Montréal (IRIC, QC, Canada). We also generated
a knockout human AML HL-60 cell line using the CRISPR-Cas9 system and a HL-60 cell
line overexpressing PRMT2 through the use of a Citrine-tagged fusion protein.
Results: Prmt2KO mice do not exhibit difference in bone marrow progenitor or mature
cell populations compared to wild type mice, indicating that PRMT2 has little or no
influence in the hematopoietic stem cell maintenance. In contrast, we observed that
Prmt2KO bone marrow-derived macrophages (BMDM) are more sensitive to lipopolysaccharides
(LPS) stimulation and express higher levels of pro-inflammatory cytokine mRNA, suggesting
a role of PRMT2 in the negative regulation of the inflammatory processes. Analysis
of datasets from a cohort of 371 AML patients (Guy Sauvageau, Leucegene project, IRIC,
Montréal, QC, Canada) reveals that patients with a lower PRMT2 expression display
a worse survival rate compared to patients with a higher PRMT2 expression. Gene Set
Enrichment Analysis of this cohort show that PRMT2 is involved in the inflammatory
response signaling pathway, thus confirming our hypothesis. In a cell model we show
that PRMT2 knockout cells exhibit higher levels of phosphorylated STAT3 protein compared
to wild type cells after LPS stimulation. PRMT2 could therefore be involved directly
or indirectly in the STAT3 pathway by preventing its phosphorylation and activation.
Ongoing transcriptomic and epigenetic studies could provide new clues for better understanding
the role of PRMT2 in an inflammatory context in AML and the association between inflammation
and leukemogenesis.
Summary/Conclusion: Taken together, our data suggest that PRMT2 has an anti-inflammatory
effect in mice and AML patients with a low PRMT2 expression inducing a higher inflammatory
phenotype, thus resulting to worse overall survival. PRMT2 could be involved in the
STAT3 signaling pathway by preventing its activation leading to increased inflammation.
P452: THE INTERACTOME OF CDK6 IN DIFFERENT ONCOGENIC CONTEXTS
L. Scheiblecker1,*, S. Nebenfuehr1, M. Zojer1, E. Doma1, V. Sexl1, K. Kollmann1
1Institute for Pharmacology and Toxicology, University of Veterinary Medicine Vienna,
Vienna, Austria
Background: The mammalian cell cycle is controlled by a complex signaling network
that is frequently deregulated in cancer. Cyclin-dependent kinases (CDKs) are the
key players in governing cell cycle progression.
CDK6 and its close homologue CDK4 are responsible for driving the cell cycle from
G1 to S-Phase. In complex with their regulatory subunits, the D-type cyclins, CDK4/6
phosphorylate the retinoblastoma protein family members (Rb) to induce G1 to S-Phase
progression and DNA replication.
CDK6, and not CDK4, plays an important role in hematopoiesis and is frequently found
overexpressed in hematologic malignancies. In the recent years, CDK6 has been shown
to regulate transcription by interacting with chromatin in a kinase dependent and
independent manner. This function is crucial in leukemia as well as hematopoietic
stem cells.
Aims: To better understand the transcriptional role of CDK6 in transformed and untransformed
cells we will analyze the interactome of CDK6. We hypothesize that different oncogenes
lead to the expression of different CDK6-dependent transcriptional programs and to
different CDK6-containing protein complexes on DNA level. We aim to identify common
and leukemia subtype-specific interaction partners of CDK6 that will represent novel
therapeutic vulnerabilities in combination with CDK6 inhibition.
Methods: To characterize the interactome of CDK6 we transformed the murine hematopoietic
progenitor cell line HPC7 with the BCR-ABL or the MLL-AF9 oncogene that give rise
to myeloid leukemia. We isolated CDK6-containing protein complexes by immunoprecipitation
followed by mass spectrometry. To extend our data and to get a specific set of interaction
partners involved in the transcriptional role of CDK6, we performed a nuclear fractionation
prior to immunoprecipitation and mass spectrometry. This enabled us to focus on interaction
partners solely in the nucleus.
Results: We obtained proteins interacting with CDK6 on a global cell level as well
as specific for the nucleus that represent co-factors of CDK6-mediated transcriptional
regulation. By intersecting the proteomics data from transformed and untransformed
HPC7 cells, we detected interaction partners of CDK6 that are common for all tested
leukemia entities, specific for leukemic subtypes and interaction partners which just
occur in untransformed cells. As a next step, the determined proteins will be subjected
to pathway analysis to reveal CDK6 dependent signaling networks. These proteins might
serve as valuable targets for potential combinatorial treatments together with CDK6
inhibition.
Summary/Conclusion: Combining our results with next generation sequencing data will
allow us deciphering the distinct transcriptional signatures regulated by CDK6 in
different leukemia entities. Our insights will provide a better understanding of malignancies
with enhanced CDK6 expression and may help to develop novel treatment strategies by
interfering with CDK6 interaction partners.
P453: CDK6 DEGRADATION IS IMPEDED BY P16INK4A AND P18INK4C IN AML
B. Schmalzbauer1,*, T. Thondanpallil1, G. Heller2, C.-M. Sperl1, I. M. Mayer1, V.
M. Knab1, S. Nebenfuehr1, M. Zojer1, A. C. Mueller3, F. Fontaine3, T. Klampfl1, V.
Sexl1, K. Kollmann1
1Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna;
2Department of Medicine I, Division of Oncology, Medical University of Vienna; 3CeMM
– Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna,
Austria
Background: Acute myeloid leukemia (AML) is a highly aggressive disease that comprises
a heterogeneous group of genetically distinct subtypes. AML is initiated by leukemic
cells, which are able to self-renew and give rise to malignant myeloid blasts. A key
regulator and prognostic biomarker for AML is the cell cycle regulator Cyclin-dependent
kinase 6 (CDK6) which represents a novel therapeutic target for the treatment of certain
subtypes of AML. CDK4/6 kinase inhibitors are extensively studied in several cancer
types but the beneficial effects may be limited by primary and secondary resistance
mechanisms. Pharmacological CDK6 degraders, which eliminate kinase -dependent and
-independent effects, represent an alternative therapeutic option. The exact mechanism
and efficacy of CDK6 degraders in AML subtypes remain unknown. The involvement of
p16INK4A in Palbociclib resistance and the role of INK4 proteins in AML disease progression
require a systematic investigation of INK4 proteins in the context of CDK6 degradation
in distinct AML subtypes.
Aims: We aim to elucidate CDK6 degrader efficacy in two CDK6-dependent AML subtypes
harboring either MLL-AF9 or AML1-ETO. We hypothesize an involvement of INK4 proteins
on CDK6 degrader efficacy. Understanding the exact mechanisms of the CDK6 degrader
will help to determine AML subtypes that would benefit from CDK6-targeted therapy.
Methods: We took advantage of a novel hematopoietic progenitor cell model (HPCLSKs)
to study CDK6 degrader efficacy in a genetically defined setting of AML subtypes.
We analyzed INK4 RNA expression of human AML patients and the HPCLSK cells using RNA-Seq
and Microarray data sets. We analyzed cell proliferation and cell cycle changes using
flow cytometry. CDK6 Co-immunoprecipitation followed by Mass Spectrometry was performed
to screen for CDK6 interaction partners that are enriched upon CDK6 Degrader treatment.
These findings were validated via immunoblotting. Human AML cell lines were used to
further validate the results in a different in vitro system and to translate them
to human settings.
Results: We show that efficacy of the CDK6 specific protein degrader varies among
AML subtypes and depends on low expression of the INK4 proteins p16INK4A and p18INK4C.
INK4 protein levels are significantly elevated in MLL-AF9+ compared to AML1-ETO+ cells,
contributing to the different CDK6 degradation efficacy. We demonstrate that CDK6
complexes containing p16INK4A or p18INK4C are protected from pharmacological degradation
and that INK4 levels define the proliferative response to CDK6 degradation. These
findings define INK4 proteins as predictive marker for CDK6 degradation – targeted
therapies in AML.
Summary/Conclusion: We here identify p16INK4A and p18INK4C as dominant CDK6 binding
partners that counteract pharmacological protein degradation in AML. Our data highlight
the need for novel CDK6 specific therapies targeting CDK6 sites not competing with
INK4 or CIP/KIP binding to overcome therapeutic limitations.
P454: VENETOCLAX AND GILTERITINIB SYNERGIZE IN FLT3 WILDTYPE ACUTE MYELOID LEUKEMIA
BY SUPPRESSION OF MCL-1 VIA COMBINED AXL AND FLT3 TARGETING
C. Schmidt1,*, M. Janssen1,2, P.-M. Bruch1,2, M. F. Blank1,2,3, C. Rohde1,2, A. Waclawiczek4,
S. Renders1,4, D. Heid1,2, S. Göllner1, L. Vierbaum1, K. Weidenauer1, S. Herbst1,
M. Knoll1, C. Kolb1, B. Besenbeck1, M. Fabre5,6, M. Gu5,6, R. Schlenk1, F. Stölzel7,
M. Bornhäuser7, C. Röllig7, U. Platzbecker8, C. Baldus9, H. Serve10, T. Sauer1, S.
Raffel1, C. Pabst1,2, G. Vassiliou5,6,11, B. Vick12,13,14, I. Jeremias12,13,14,15,
A. Trumpp4, J. Krijgsveld3,16, C. Müller-Tidow1,2, S. Dietrich1,2
1Department of Medicine V, Hematology, Oncology and Rheumatology, University hospital
Heidelberg; 2Molecular Medicine Partnership Unit (MMPU), University of Heidelberg
and European Molecular Biology Laboratory (EMBL); 3Division Proteomics of Stem Cells
and Cancer; 4Division of Stem Cells and Cancer, German Cancer Research Centre (DKFZ),
Heidelberg, Germany; 5Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus,
Hinxton; 6Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute, University
of Cambridge, Cambridge, United Kingdom; 7Department of Medicine I, University hospital
Carl Gustav Carus, Dresden; 8Medical Clinic and Policlinic I, Hematology and Cellular
Therapy, Leipzig University Hospital, Leipzig; 9Department of Hematology and Oncology,
University hospital Schleswig-Holstein, Campus Kiel, Kiel; 10Department of Medicine
II, Hematology-Oncology, Goethe University Hospital Frankfurt, Frankfurt am Main,
Germany; 11Department of Hematology, University of Cambridge, Cambridge, United Kingdom;
12Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München;
13German Research Center for Environmental Health (HMGU); 14German Consortium for
Translational Cancer Research (DKTK); 15Department of Pediatrics, Dr. von Hauner Children’s
Hospital, Ludwig Maximilians University München, Munich; 16Medical Faculty Heidelberg,
Heidelberg University, Heidelberg, Germany
Background: BCL-2 inhibition has been shown to be effective in older patients with
acute myeloid leukemia (AML) in combination with hypomethylating agents or low-dose
cytarabine. However, treatment resistance and relapse represent major clinical challenges.
Thus, there is an unmet need to overcome resistance to current venetoclax-based regimens.
Aims: In order to establish an alternative treatment approach for AML in elderly patients,
we aimed at identifying a highly synergistic drug combination partner for venetoclax.
Methods: We performed a high-throughput drug screening to identify the most effective
combination partner for venetoclax in AML. Overall, 64 anti-leukemic drugs were screened
in 31 primary high-risk AML samples in the presence or absence of venetoclax. We calculated
drug synergies, validated top venetoclax combination partners in AML cell lines and
primary patient samples and performed mechanistic analyses.
Results: Established anti-leukemic drugs exhibited only weak synergism with venetoclax
in high-risk AML samples. In contrast, when targeting distinct pathways such as FLT3
signaling, we observed increased synergistic activity. Gilteritinib exhibited highest
synergy with venetoclax in FLT3 wildtype AML, with especially high synergy values
in TP53 mutated samples. The combination of gilteritinib and venetoclax increased
apoptosis, reduced viability and abolished colony-formation potential in FLT3 wildtype
as well as venetoclax-azacitidine resistant cell lines and primary patient samples.
Proteomics revealed increased levels for proteins involved in FLT3 wildtype signaling
in specimens with low in-vitro response to the currently used venetoclax-azacitidine
combination. Mechanistically, venetoclax with gilteritinib decreased phosphorylation
of ERK and GSK3B with subsequent suppression of the antiapoptotic protein MCL-1. MCL-1
downregulation was associated with increased MCL-1 phosphorylation of serine 159 and
proteasomal degradation. Compared to other FLT3 inhibiting drugs, gilteritinib showed
highest synergism with venetoclax due to the additional inhibition of AXL that reinforced
the downregulation of MCL-1.
Gilteritinib and venetoclax proved efficacious in an FLT3 wildtype PDX model with
TP53 mutation. Furthermore, the drug combination reduced leukemic burden in four patients
with venetoclax-azacitidine refractory AML, who received gilteritinib and venetoclax
as an individual treatment approach.
Summary/Conclusion: In summary, our results identified gilteritinib as a potential
therapy combination partner for venetoclax in patients with FLT3 wildtype AML.
P455: BREAKING THE PUMP: TARGETING THE SODIUM-POTASSIUM PUMP AS A THERAPEUTIC STRATEGY
IN ACUTE MYELOID LEUKEMIA
C. Schneider1,2,*, H. Spaink1, G. Alexe1,2, N. V. Dharia1,2, D. Khalid1, S. Scheich3,4,
B. Haeupl4,5, T. Oellerich4,5, K. Stegmaier1,2
1Pediatric Oncology, Dana-Farber Cancer Institute, Boston; 2The Broad Institute of
MIT and Harvard, Cambridge; 3Lymphoid Malignancies Branch, National Cancer Institute,
National Institutes of Health, Bethesda, United States of America; 4Department of
Medicine II, Department for Hematology/Oncology, Goethe University, Frankfurt; 5German
Cancer Research Center and German Cancer Consortium, Heidelberg, Germany
Background: Acute myeloid leukemia (AML) is a heterogenous hematologic malignancy
with poor overall survival despite intensive cytotoxic chemotherapy and newer targeted
agents. With a 5-year survival rate of only 29% in adults with AML, new therapeutic
strategies are needed.
Aims: In order to identify novel, selective targets for AML, we used the Cancer Dependency
Map, which provides genome-scale CRISPR/Cas9 depletion screen data in hundreds of
cancer cell lines. Here, we identified ATP1B3 as a context-specific dependency that
could be therapeutically exploited.
ATP1B3 is the smaller glycoprotein subunit (beta) of the sodium-potassium pump (Na/K-ATP
pump). Together with the catalytic alpha subunit ATP1A1, it forms a heterodimer located
in the plasma membrane, regulating the electrochemical gradient through the transport
of Na and K ions across the membrane. The Na/K-ATP pump is of vital importance in
the maintenance of cellular homeostasis and membrane potential and because of its
function as a receptor and signal transducer. While the ATP1A1 carries out ion transport
and enzymatic activity, making it a common essential gene (a gene which ranks among
the most depleted genes in at least 90% of screened cell lines), ATP1B3 is not essential
in all cancer cells. This beta subunit seems to be crucial for intracellular transport
and stabilization of the alpha subunit in the membrane. The beta subunit has 4 paralogs,
showing similar expression patterns among tissues, with one exception, ATP1B1.
Methods: We used a diverse panel of functional genomic based assays, validating our
finding using CRISPR/Cas9 knockout and overexpression constructs for the Na/K-ATP
pump beta subunit paralogs. In an approach to analyze protein-protein interactions
we generated BioID constructs for a mass spectrometry-based analysis. For an in vivo
study we used a bioluminescent imaging (BLI)-based orthotopic mouse model of AML,
carrying non-targeting, ATP1B1 or ATP1B3 knockout guides.
Results: By using CRISPR/Cas9 knockout we could validate ATP1B3 as a selective dependency
and performing competitive growth assays we showed that loss of ATP1B3 in ATP1B1 low
expressing AML cells leads to synthetic lethality. The absence of both paralogs of
the beta subunit results in the loss of their common essential binding partner ATP1A1
in hematologic malignancies, while higher expression of ATP1B1 can stabilize ATP1A1
under the loss of ATP1B3 in solid tumors or in ATP1B1 overexpressing AML cells.
Next, we validated our findings in vivo. In a BLI-based orthotopic mouse model of
AML, we found that the AML model with ATP1B3 knockout showed lower leukemia burden
and normal spleen weight in comparison to a non-targeting or ATP1B1 knockout control.
To understand the specific role of ATP1B3 we produced BioID (proximity-dependent biotin
identification) constructs for ATP1B1 and ATP1B3. In ongoing studies, through this
mass spectrometry-based analysis of protein-protein interactions, we will gain a greater
insight into the protein partners of the Na/K-ATP pump.
Summary/Conclusion: Taken together, we identified ATP1B3 as a selective dependency
in AML. We propose that the elimination of ATP1B3 leads to the destabilization of
the sodium-potassium pump when ATP1B1 levels are low, making it a potential tumor-selective
therapeutic target for AML and other hematologic malignancies with low expression
of ATP1B1.
P456: A LONG READ SEQUENCING AND CRISPR-CAS9 BASED APPROACH FOR RAPID COPY-NUMBER
ALTERATION AND STRUCTURAL VARIATION DETECTION IN HEMATOLOGIC MALIGNANCIES
J. Schrezenmeier1,2,*, J. E. Straeng1, O. Blau1, A. Elashy1, A. Lazarides1, S. Skambraks3,
E. Jahn3, B. Gillißen1, C. Eckert4, A. Nogai1, I.-W. Blau1, K. Giannopoulos5, M. Heuser6,
K. Döhner3, L. Bullinger1, A. Dolnik1
1Hematology, Oncology and Tumorimmunology, Charité-Universitätsmedizin Berlin; 2Berlin
Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation
Academy, Berlin; 3Department of Internal Medicine III, University of Ulm, Ulm; 4Pediatric
Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany; 5Department
of Experimental Hematooncology, Medical University of Lublin, Lublin, Poland; 6Department
of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical
School, Hannover, Germany
Background: Current standard karyotyping methods are labor-intensive and have severe
limitations in terms of resolution and duration to results. For hematologic malignancies,
but not restricted to, this represents a relevant obstacle in translating molecular
findings into time critical treatment decisions.
Aims: To develop a sequencing based approach for rapid and scalable cytogenetics that
can be applied to a wide spectrum of hematologic malignancies to detect clinically
relevant molecular markers within 48hrs
Methods: For copy number alteration detection, whole genome sequencing was performed
on a GridION sequencer (Oxford Nanopore Technologies, ONT, using the SQK-LSK109 kit)
in a cohort of 53 hematologic neoplasms, including acute myeloid leukemia (AML, n=18),
chronic lymphocytic leukemia (CLL, n=8), multiple myeloma (MM, n=17) and pediatric
acute lymphocytic leukemia (ALL, n=10) samples. For the identification of balanced
alterations, transcriptome sequencing data were generated using ONT (SQK-DCS109 kit),
and we applied an in-house developed analysis pipeline based on minimap2 alignment
followed by Blast with an ensuing filtering algorithm based on orientation, inter-read
distance and length filtering. For targeted sequencing of structural aberrations,
we developed a CRISPR-Cas9 based tiling approach using sgRNA pools (Integrated DNA
Technologies, IDT). This approach allows to cover AML relevant targets such as t(8;21),
the KMT2A (MLL) breakpoint area, and the FLT3-ITD region as well as lymphoma relevant
regions such as the immunoglobulin heavy chain locus (library preparation was performed
using SQK-CS9109 kit).
Results: For copy number profiling a median whole-genome coverage of 2.6 was reached
(range 0.27-7 fold). ONT sequencing and conventional karyotyping approaches showed
a high concordance with Pearson correlation coefficients >0.95 for copy number alteration
comparisons between conventional cytogenetics and ONT sequencing results for all investigated
disease entities. Regarding the detection of structural aberrations transcriptome
sequencing and fusion gene analysis using the above described analysis and filtering
pipeline we could e.g. reliably detect the t(9;22) translocation from the K562 cell
line and the t(8;21) translocation from Kasumi-1. Additionally, using this method
we were able to correctly identify the primary AML sample harboring a t(8;21) translocation
that was among the n=12 AML patient samples. In addition to an RNA-based work-flow,
we established a CRISPR-Cas9 based DNA enrichment approach for the detection of recurrent
structural aberrations from specified genomic DNA loci. The enrichment of genomic
regions of interest using sgRNA libraries led e.g. to a median coverage of 44 reads
(range 5-136-fold) of the t(8;21) region-of-interest in primary AML samples, which
allow the detection of the exact coordinates of the breakpoints.
Summary/Conclusion: Long read based sequencing based copy number alteration and structural
variation detection based on combination of different long read sequencing approaches
like low-coverage whole genome sequencing, transcriptome sequencing and CRISPR-Cas9
based sequencing of genomic loci of interest represents a highly promising tool for
high resolution and high speed cytogenetics that has the potential to overcome many
limitations of conventional cytogenetics.
P458: TRANSCRIPTIONAL AND TRANSLATIONAL SIGNATURES OF TRIPLE MUTATED DNMT3AKO/FLT3ITD/NPM1C
MICE SHOW ALTERED RNAS PROCESSING AND CELL CYCLE PROGRESSION
A. Scialdone1,*, D. Sorcini1, A. Stella1, V. Tini1, A. Marra1, C. Rompietti1, F. De
Falco1, E. Dorillo1, F. M. Adamo1, E. C. Barcelos1, R. Arcaleni1, M. P. Martelli1,
P. Sportoletti°1, B. Falini°;° co-last author1
1Medicine and Surgery, University of Perugia, Perugia, Italy
Background: The top ranking recurrently mutated genes in AML are NPM1, FLT3 and DNMT3A
1,2. Co-occurrence of these three mutations is associated to a unique AML subset characterized
by high peripheral blood and bone marrow blast counts, normal cytogenetic and very
poor EFS and OS3. Nevertheless, the cooperative leukemogenic mechanisms of these three
mutations have not been deeply explored and a successful targeting treatment of AML
patientsDNMT3Amut/
FLT3
-ITD/
NPM1
c still represents an unmet clinical need.
Aims: To dissect how DNMT3A loss of function, FLT3 Internal Tandem Duplication (ITD)
and NPM1 mutations (NPM1
c) cooperate in driving AML onset and progression through a picture of the transcriptome
and the translatome of triple mutated AML cells.
Methods: Mx1-Cre-NPM1
c and/or Mx1-Cre-DNMT3A
KO and FLT3
ITD mutant mice were crossed to obtain the following lines: DNMT3A
KO/FLT3
ITD/NPM1
c, FLT3
ITD/DNMT3A
KO and FLT3
ITD/NPM1
c. Peripheral blood (PB) was periodically taken and analysed, previous anesthesia,
while bone marrow (BM) and spleen were harvested two months after pIpC induction or
at conclamate disease (n=7 each group). To investigate the mechanisms underlying the
triple mutated mice phenotype, we looked BM leukemic samples from all groups both
at transcriptional level by RNA sequencing (RNAseq) and at translational level (protein
synthesis capacity of AML blasts) by Ribosome footprinting (Riboseq)4. Unique Differentially
Expressed Genes (DEGs) (filtered by p-value<0.05 and FC >1.5) belonging to FLT3
ITD/DNMT3A
KO/NPM1
c
vs FLT3
ITD/DNMT3A
KO were identified by Venn diagram and used in enrichment analysis on Reactome database
and MSigDB (C6-oncogenic signature).
Results: The FLT3ITD/DNMT3A
KO/NPM1c, DNMT3A
KO/FLT3
ITD and FLT3
ITD/NPM1
c displayed a fully penetrant leukemic phenotype. As expected, FLT3ITD/ DNMT3A
KO/NPM1
c showed a more aggressive disease characterized by higher WBC counts, more pronounced
blasts infiltration of the spleen and shorter survival (>2 fold vs double mutants).
The FLT3ITD/DNMT3A
KO/NPM1
c mice displayed 1429 (842 UP and 587 DOWN) DEGs compared to FLT3
ITD/DNMT3A
KO mice. The enrichment analysis suggested that DNMT3A and NPM1 mutations, in a constitutive
FLT3
ITD expression context, cooperate to activate transcriptional programs sustaining
the proliferation and maintenance of blasts through concerted upregulation of a number
of pathways, including the Anaphase-Promoting Complex (APC), which regulates cell
cycle progression, the m-TOR-mediated autophagy inhibition, the Rho-GTPases effectors
(with Rac1 overexpression), the Hippo-mediated self renewal, the HOXA9 signature and
TP53-regulated apoptosis. Interestingly, genes involved in transport of mature transcripts
to cytoplasm and nuclear RNAs metabolism were also upregulated. In keeping with these
RNAseq findings, the Riboseq data showed that both the quote of processed transcripts
(15 vs 9%) and the percentage of processed pseudogenes were more abundant in triple
vs double mutated mice. On the other hand, the long intergenic noncoding RNAs (lincRNA)
fraction was reduced in triple mutants. This finding may reflect dysfunctional remodeling
of tumor microenvironment, contributing to AML cells immune escape in the BM5.
Image:
Summary/Conclusion: The leukemic DNMT3A
KO/FLT3
ITD/NPM1
c genotype is characterized by unique transcriptional and translational signatures.
The information about deregulated pathways and altered protein synthesis establishes
the rational basis for designing new therapeutic approaches in this poor prognosis
AML genotype.
P459: NUC-7738 REGULATES BETA-CATENIN SIGNALLING RESULTING IN REDUCED PROLIFERATION
AND SELF-RENEWAL OF AML CELLS
A. M. Shahid1,*, M. Elshani1, D. J. Harrison1
1School of Medicine, University of St Andrews, St Andrews, United Kingdom
Background: NUC-7738, a phosphoramidate transformation of 3’deoxyadenosine (3’dA),
is specifically designed to generate the active anti-cancer metabolite 3’-deoxyadenosine
triphosphate (3’-dATP) directly in cells, bypassing key cancer resistance mechanisms
of transport, activation and breakdown. NUC-7738 is currently in a Phase I/II clinical
study to assess safety and determine the recommended dose in patients with advanced
solid tumors and lymphoma. We have recently shown in cell lines a potential therapeutic
role for NUC-7738 in patients with acute myeloid leukemia (AML), whereby NUC-7738
induced myeloid cell differentiation and mitochondrial-mediated apoptosis. AML leukemia
stem cells (LSCs) are required for the initiation and maintenance of the disease.
Activation of the Wnt/β-catenin pathway is required for the survival and development
of LSCs and therefore, targeting β-catenin is a potential therapeutic strategy.
Aims: The aim of this study was to determine whether NUC-7738 regulates β-catenin
and the expression of its target genes in AML cells
Methods: AML cell lines KG1a, OCI-AML3, HL-60 and U937 were treated with NUC-7738
for 48 or 72 hours. LSCs were determined by the expression of surface markers CD34,
CD38 and CD123 via flow cytometry. Self-renewal was assessed by performing the colony
forming assay using MethoCult Enriched media. Cell proliferation was determined by
the Alamar Blue assay. Immunoblotting was performed for β-catenin, PI3K-p110, phosphorylated
AktSer473 and phosphorylated GSK3βSer9
Results: NUC-7738 reduced PI3K-p110, phosphorylated AktSer473 and phosphorylated GSK3βSer9
resulting in reduced β-catenin expression. NUC-7738 decreased the percentage of CD34+
CD38- CD123+ (LSCs) from 15% to 7% and significantly reduced the total number and
size of leukemic colonies. NUC-7738 inhibited cell proliferation in U937, OCI-AML3
and HL-60 cells, with IC50’s of 1.55 µM, 7 µM and 16 µM, respectively.
Summary/Conclusion: Self-renewal pathways, including Wnt/β-catenin, are key for the
initiation and long-term maintenance of LSCs. Through the ability to reduce PI3K-p110,
phosphorylated AktSer473, and phosphorylated GSK3βSer9 and suppress β-catenin signalling,
NUC-7738 exploits a key developmental process of AML, supressing the expansion and
survival of LSCs. These results indicate that targeting LSCs with NUC-7738 may offer
a successful therapeutic strategy for treating patients with leukemia.
P460: RECOMBINANT SLIT2 INHIBITS ACUTE MYELOID LEUKEMIA CELL PROLIFERATION IN VITRO
AND IN VIVO
L. Araujo de Albuquerque Simoes1,2,3,4,*, I. Weinhäuser2,5, D. A Pereira-Martins2,5,
C. A. Ortiz Rojas2, T. Mantello Bianco2, R. D. C. Cavaglieri6, E. Magalhães Rego2,3,4,7
1Center of Translational Research in Oncology, ICESP - Faculty of Medicine, USP, Sao
Paulo; 2Center for Cell Based Therapy, Fundação Hemocentro de Ribeirão Preto, Ribeirão
Preto; 3Medical Research Laboratory on Molecular Hematology (LIM31), University of
São Paulo; 4Hematology, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil;
5Department of Experimental Hematology, University Medical Center Groningen, Groningen,
Netherlands; 6Medical Research Laboratory on Molecular Hematology (LIM31), Faculty
of Medicine, University of Sao Paulo; 7Center of Translational Research in Oncology,
ICESP - Faculty of Medicine, USP, Sao Paulo, Brazil
Background: The SLIT/ROBO axis has been shown to play a key role in many physiological
processes such as organogenesis, axon guidance and angiogenesis. Additionally, several
studies conducted in solid tumors observed frequent hypermethylation of either Slit
or Robo at their promoter site. In the context of leukemia, Golos et al., 2019 showed
that the low expression of SLIT2 was associated with lower overall survival in adults
with acute myeloid leukemia (AML). Furthermore, it was demonstrated by our group that
the knockdown of SLIT2 in Acute Promyelocytic Leukemia (APL) cells leads to an increase
of cell proliferation in vitro and a more aggressive course of the disease in vivo
(Weinhäuser et al., 2020).
Aims: Given the evidence of the relevance of SLIT2 in APL, we opted to transfer our
gained knowledge to a more challenging leukemia subset and decided to study the role
of SLIT2 in AML.
Methods: We first evaluated the methylation pattern of SLIT2 in AML patients compared
to healthy donors by analyzing publicly available datasets (GSE58477, normal karyotype
blasts: 62, healthy CD34+: 10; GSE63409, LSC: 14, HSC: 5), followed by the assessment
of the level of SLIT2 in the bone marrow (BM) plasma of AML patients and healthy donors
by ELISA. Additionally, to functionally assess the biological role of SLIT2, we treated
AML cell lines (KASUMI1, MV4-11, and MOLM13) with recombinant SLIT2 (50ng/mL) in vitro.
We performed the knockdown of SLIT2 in AML cell lines (THP-1 and OCI-AML3) and evaluated
their proliferation capacity as well. Moreover, AML cells were treated with decitabine
and recombinant SLIT2. Finally, we evaluated the anti-leukemic effects of SLIT2 in
vivo. To do so NSGS mice were transplanted with luciferase-transduced MV4-11 cells
and the animals were either treated with vehicle (control group) or recombinant SLIT2
(25 ng/g of body weight) two days after transplant for a period of one week.
Results: Our analysis indicated increased methylation at the SLIT2 promoter site in
AML patients compared to healthy CD34+. In accordance with our analysis, we detected
decreased level of SLIT2 protein in the bone marrow (BM) plasma of AML patients (1.43 ng/mL)
when compared to healthy donors (3.51 ng/mL) (p<0.05). The functional role of SLIT2
was determined with the treatment of KASUMI1, MV4-11 and MOLM13 cells with recombinant
SLIT2 in vitro. Our results showed that SLIT2 treatment reduced the cell proliferation
and colony formation capacity and induced cell cycle retention in the G1 phase for
all AML cell lines. Contrarily, the knockdown of SLIT2 promoted increased cell proliferation
in THP-1 and OCI-AML3 cell lines. Moreover, we observed increased induction of decitabine-induced
apoptosis when MV4-11 and MOLM13 cells were treated in the presence of recombinant
SLIT2. Finally, the engraftment and progression of the disease of NSGS mice transplanted
with luciferase-transduced MV4-11 cells were monitored by the detection of luciferase
bioluminescent signals. Our results showed, that SLIT2 treatment was able to significantly
delay the progression of AML in vivo. Mice treated with SLIT2 presented improved overall
survival (vehicle 15d: CI 13-16d; SLIT2 19d: 95% CI: 16-22d. P value = 0.0320), decrease
leukemic infiltration in the BM and spleen, reduced spleen size (vehicle: 125 mg;
SLIT2: 100 mg P value= 0.04), indicating lower disease burden when compared to the
control group.
Summary/Conclusion: In conclusion, our results suggest that SLIT2 has tumor suppressive
functions in AML in vitro and in vivo highlighting the therapeutic potential with
low cytotoxicity of SLIT2 in AML.
P461: MYELOID KINOME INHIBITOR HM43239 OVERCOMES ACQUIRED RESISTANCE IN ACUTE MYELOID
LEUKEMIA MODELS
R. Bejar1,*, S. J. Baek2, N. Abbasi1, A. Krasny1, R. Sinha1, H. Zhang1, S. Howell3,
W. Rice1
1Aptose Biosciences Inc., San Diego, United States of America; 2Hanmi Pharmaceutical
Company, Seoul, South Korea; 3Moores Cancer Center, University of San Diego Health,
San Diego, United States of America
Background: Oral HM43239 is in development for the treatment of acute myeloid leukemia
(AML) because of its capacity to potently inhibit kinases that drive myeloid malignancies,
including diverse forms of the FLT3, SYK, JAK and c-KIT kinases. Wildtype FLT3 is
overexpressed in most AML patients, and approximately 30% of newly diagnosed adult
AML patients harbor internal tandem duplications (ITDs) or point mutations in the
tyrosine kinase domain (TKD). These mutations drive aberrant activation of downstream
proliferation pathways and are associated with a high risk of relapse. Likewise, the
c-KIT alternative receptor kinase, as well as the SYK and JAK1/2 intracellular kinases,
mediate oncogenic signaling in AML that can promote drug resistance to certain FLT3
inhibitors. HM43239 was developed to overcome shortcomings of other FLT3 inhibitors.
It inhibits a broad set of mutant and wildtype forms of FLT3, while simultaneously
disrupting downstream SYK, JAK/STAT5, ERK, and other rescue signaling pathways. This
rationale supports the development of HM43239.
Aims: Evaluate the activity of HM43239, an orally active drug, as a FLT3-focused myeloid
kinome inhibitor in human AML models.
Methods: Biochemical kinase assays were performed by RBC and Carna. The effects of
HM43239 on cell proliferation (IC50), growth rate (GR50) and concentration at half-maximal
effect (Growth Effective Concentration; GEC50) were determined using the MTS assay
with vehicle controls. Cell-based inhibition of target phosphorylation was assessed
by Western blot and flow cytometric analyses. The AML cell lines tested included MV-4-11,
MOLM-13, MOLM-14 and BAF3/ITD. In vivo efficacy was assessed using the MOLM-14 FLT3-Mutated
xenograft model.
Results: HM43239 inhibited the enzymatic activities of FLT3-WT, -ITD, and -D835Y variants
with IC50 values of 1.1, 1.8 and 1.0 nM, respectively. Binding K
d
values for FLT3-WT, -ITD, -D835Y, -D835H, -ITD/D835V and -ITD/F691L were 0.58, 0.37,
0.29, 0.4, 0.48, and 1.3 nM, respectively. HM43239 killed AML cells that harbor the
FLT3-ITD mutation (MV-4-11, MOLM-13 and MOLM 14) with IC50, GR50 and GEC50 values
ranging from 4 to 10 nM. In cell lines expressing wild type FLT3 (KG1, HEL92.1, SKM-1,
and THP-1) the values for these parameters ranged from 0.05 nM to 3 µM. Western blot
and flow cytometric analyses of MV-4-11, MOLM-14 cells and Ba/F cells transfected
with target proteins revealed that the phosphorylation of FLT3 and SYK was reduced
by 50-90% at HM43239 concentrations of 10 – 100 nM, and that these concentrations
produced >80% reduction in phosphorylation of ERK and JAK/STAT5 that function downstream
of FLT3. HM43239 inhibited FcγR-induced SYK and JAK/STAT5 activation in KG-1a (FLT3-WT)
cells that upregulate RAS signaling which is a mechanism of acquired resistance to
gilteritinib. In murine xenograft studies, HM43239 was more effective than gilteritinib
in both SC and orthotopic FLT3-F691L and FLT3-ITD/F691L mutant MOLM-14 models.
Summary/Conclusion: HM43239 inhibits wild type and mutant forms of FLT3 at low nM
concentrations and demonstrates in vivo efficacy at doses that are well tolerated.
Its ability to also inhibit SYK and, by reducing the activity of these upstream kinases,
to also impair the activity of EKR1/2 and JAK/STAT5 that participate in rescue pathways,
makes this a particularly interesting molecule with the potential of offsetting the
development of resistance that is common with other FLT3 inhibitors. A Phase 1/2 trial
of HM43239 in patients with AML is open and accruing patients (NCT03850574).
P462: IDENTIFICATION OF TARGETED THERAPIES DIRECTED TO ACUTE MYELOID LEUKEMIA MINIMAL
RESIDUAL DISEASE
N. van Gils1,*, F. Kessler1, M. Broux2, F. Denkers1, S. Demeyer2, J. Cools2, D. C.
De Leeuw1, J. Janssen1, L. Smit1
1Hematology, UMC Amsterdam, location VUmc, Cancer Center Amsterdam, Amsterdam, Netherlands;
2Center for Human Genetics, KU Leuven, Leuven, Belgium
Background: Despite good responses to intensive polychemotherapy in the initial treatment
phase, the biggest challenge in the treatment of acute myeloid leukemia (AML) is persistence
of residual therapy-resistant cancer cells (measurable residual disease, MRD) that
can develop into recurrence. Leukemia cells with stem cell features (“leukemic stem
cells”, LSCs) residing within MRD are thought to be at the origin of relapse. Current
knowledge gaps on residual leukemic cell persistence and ways to target MRD are hampering
progress in development of treatments successfully preventing relapse and increasing
AML cure rates.
Aims: To identify transcriptional and epigenetic characteristics and vulnerabilities
of AML MRD, guiding the design of “targeted” combination treatment strategies that
successfully eliminate relapse-initiating cells in individual AML patients.
Methods: We developed in vivo patient-derived xenograft (PDX) NOD/SCID/IL2Rγc-deficient
(NSG) mouse models mimicking MRD by 1) intravenous (IV) injection of primary AML diagnosis
samples and, as soon as leukemic cells were detectable in the peripheral blood, treatment
with combination chemotherapy, and 2) IV injection of AML patient samples at the stage
of MRD in NSG mice. Moreover, we selected samples from individual AML patients at
different time points, diagnosis, MRD and relapse, and purified by flow cytometry
the leukemic cells using the leukemia-associated immunophenotype (LAIP) expressed
on the AML cells. To elucidate the mutational landscape, transcriptional program and
chromatin accessibility involved in development and persistence of MRD, we performed
DNA-, RNA- and the assay for transposase-accessible chromatin (ATAC)-sequencing on
the purified leukemic cells.
Results: We revealed that in patient AML samples at the stage of MRD there is clonogenic
capacity of leukemic stem/progenitors (Figure 1A). Treatment with combination chemotherapy
significantly reduced leukemic burden in the bone marrow of NSG mice (Figure 1B top).
Interestingly, the number of CD34+CD38- leukemic cells in AML1 was hardly affected
by chemotherapy, whereas treatment significantly reduced the number of these cells
in AML2 (Figure 1B bottom), indicating that leukemic CD34+CD38- cells residing within
different patients have a heterogeneous response to polychemotherapy. Injection of
AML MRD from several patients resulted in leukemia engraftment (example Figure 1C).
For one case, marked as MRD-negative by flow cytometry, we even observed that MRD
could initiate high leukemia load in the mice (Figure 1D), while after injection of
diagnosis and relapsed AML from the same patient no engraftment was observed. Together,
our results implicated that AML cells after therapy, even if the patient is marked
as MRD-negative, could have high leukemia-initiating potential, which could have been
induced by chemotherapy.
Using RNA-sequencing we identified and compared the gene expression profile and chromatin
accessibility of leukemic cells at diagnosis, MRD and relapse from patients samples.
We identified several genes upregulated in AML MRD cells as compared to diagnosis
AML. Moreover, we showed increased chromatin accessibility, indicating open chromatin
structure and potential transcriptional activity, at several loci in MRD compared
to diagnosis AML, giving opportunities to therapeutically target MRD.
Image:
Summary/Conclusion: We generated AML MRD PDX mouse models, and identified transcriptional
and epigenetic characteristics of AML MRD that might guide the design of “targeted”
combination treatment strategies potentially eliminating relapse-initiating cells.
P463: RE-PURPOSING OF GENE SIGNATURES IN AML UNCOVERS NOVEL ENERGETICS-ASSOCIATED
MOLECULAR SUBTYPES
P. Strain1,*, E. E. Scanlon1, G. Jellema2, R. D. Kennedy1,2, K. I. Mills1, J. K. Blayney1
1Patrick G Johnston Centre for Cancer Research, Queens University Belfast, Belfast;
2Almac Diagnostics, Almac, Craigavon, United Kingdom
Background: AML is a highly heterogeneous disease with great diversity in clinical
features and patient response to treatments. Despite recent improvements in disease
understanding, treatments have remained unchanged for 30 years. This presents the
need to characterise the disease more fully and characterise its underpinning molecular
subtypes. The re-use of published and validated prognostic and predictive gene signatures
e.g. Cancer Hallmarks presents an invaluable in silico opportunity to uncover the
biological mechanisms underpinning treatment response in AML.
Aims: To develop an automated statistical analysis pipeline combined with machine
learning techniques to derive robust patient clusters representative of novel molecular
AML subtypes significantly associated with clinical variables and survival outcomes.
Methods: Analysis was carried out using a primary dataset (AML-OHSU: 451 AML patient
samples) and a validation dataset (TCGA-LAML: 200 AML patient samples). Both datasets
had been processed using Almac’s claraT platform, a software-driven solution which
provides a comprehensive overview of tumour profiles using gene expression signatures.
An automated analytical pipeline was developed using Consensus Clustering (CC), a
method that determines the number and membership of potential clusters, using different
combinations of 8 distances and 7 linkages within a dataset. Robustness was tested
via bootstrapping. This pipeline was used to stratify patients via a dimension reduction
approach whereby clustering was performed on 210 gene signatures categorised by 10
different hallmarks of cancer. Analysis was performed to identify clinical associations
within robust clusters that were linked to differences in survival.
Results: The automated clustering pipeline analysed a total of 1,314 stable clusters
across 10 cancer hallmarks in the AML-OHSU dataset. Stable clusters were subsequently
processed via log rank analysis (OS right-censored at 60 months) identifying 134 stable
clusters with significant differences (p-value <0.05) in survival outcome.
Stable clusters with significant survival differences were tested against 32 clinical
categorical variables present in the AML-OHSU dataset. The results were filtered for
a significant threshold (chi square p-value <0.05 and BH p-value <0.2). Here we found
gene signatures representative of the Energetics hallmark, incorporating 22 signatures,
to be one of the most frequently clustered throughout our results, ranking highest
where K=3.
A significant difference in overall survival probability (Log rank p-value: 0.033)
was found between clusters (Energetics, K=3). Patients in the poorest survival cluster
were characterised by a refectory induction response to treatment, having the lowest
number of fusions, a low frequency of NPM1 mutations and a high proportion of patients
above the age of 65.
To validate energetics results from the AML-OHSU dataset, CC was again performed using
gene signatures from the TCGA dataset that were representative of the energetics hallmark.
A significant difference between overall survival probability (Log rank p-value: 0.019)
was again found between stable clusters of the energetics hallmark (K = 3).
Summary/Conclusion: We have demonstrated that a novel analytical pipeline developed
here to analyse Hallmark-related gene signatures can aid in the discovery of new molecular
subtypes in AML associated with prognosis. We have subsequently validated these in
an independent dataset. The Energetics hallmark has not, to our knowledge, been linked
with AML prognosis before, and may suggest novel biology linked to treatment response.
P464: COMPARED TO CLASSICAL CYTOGENETICS OPTICAL GENOME MAPPING (OGM) DETECTS MULTIPLE
ADDITIONAL STRUCTURAL CHROMOSOMAL ABERRATIONS IN PEDIATRIC ACUTE MYELOID LEUKEMIA
(AML)
J. Suttorp1,*, J. L. Lühmann2, D. Steinemann2, D. Reinhardt1, N. von Neuhoff1, M.
Schneider1
1Clinic of Pediatrics III, University Hospital Essen, Essen; 2Department of Human
Genetics, Hannover Medical School, Hannover, Germany
Background: AML is the 2nd most frequently diagnosed blood cancer in children affecting
approximately 130 patients annually in Germany. Pediatric AML is a heterogeneous malignancy
that has multiple genetic aberrations. This is relevant for classification, prognosis
and selection of optimal therapy. To detect these alterations established procedures
(karyotyping, FISH, RNA-based techniques) are used with well-known limitations (resolution
of karyotyping and FISH, respectively; cell cultivation is necessary to generate metaphases;
cost-intensiveness; necessity of experienced and intensively trained staff). OGM is
an emerging technique addressing these limitations. Based on extraction of ultra-high
weight DNA followed by fluorescent labelling at the sequence CTTAAG a barcode-like
pattern of the genomic DNA is created. The DNA strands labelled in this way are stretched
and drawn as single ultra-long fragments through nanochannels. The results are translated
into molecule maps which are compared to the reference genome (resolution down to
500 bp) allowing the identification of genetic alterations. This method combines high
resolution with picturing almost whole chromosomes without cell cultivation.
Aims: The aim of this study was to test to what extend OGM might supplement classical
cytogenetics (CCG) to detect risk defining genetic aberrations at initial diagnosis
in pediatric patients with AML.
Methods: We analyzed 24 specimen of pediatric AML, MPAL and bilineage leukemia patients
by OGM starting from stored frozen material (bone marrow, peripheral blood) obtained
at initial diagnosis. Results of OGM were compared with karyotyping and FISH. Primer
walking and breakpoint spanning PCR were used to validate newly detected aberrations
by OGM in selected cases. Applying breakpoint-spanning PCR, validated aberrations
were used as markers for assessment of minimal residual disease (MRD) in selected
cases during AML treatment.
Results: Overall, we detected discrepant results between CCG and OGM in 17/24 (70%)
cases including 9 cases in which karyotyping detected aberrations which were not found
by OGM. However, these aberrations were not relevant for risk stratification. In general,
focusing on genetic aberrations important for risk stratification the results between
CCG and OGM were concordant in 23/24 (96%) cases. In a single case OGM was able to
detect a high-risk aberration which was missed by CCG. In total, 33 previously unknown
genetic alterations were identified by OGM. Out of these four newly detected variants
(2 deletions, 2 translocations) were validated by PCR. The translocations t(2;12)
and t(8,12) both affected gene ETV6 on chr. 12 with different fusion partners (chr.
2: AC064875.1; chr. 8: NSMCE2). In both translocations sequences at the breakpoints
were determined. Two deletions affecting genes RNF157 on chr. 17 and MEF2B on chr.
19 were validated. These aberrations had not been described before in pediatric AML.
In 2 cases - both without a previously identified MRD marker – the newly detected
translocations by OGM were used for MRD monitoring.
Summary/Conclusion: OGM considerably expands the range of techniques to optimize the
diagnosis of pediatric AML and has much potential to omit limitations of classical
cytogenetics. Furthermore, the results will contribute to a better understanding of
the leukemogenesis of pediatric AML. In addition, OGM offers the possibility to identify
new aberrations which can serve as patient specific MRD markers especially in cases
without a previously known MRD marker.
P465: A LIPID NANOPARTICLE DELIVERY SYSTEM FOR TARGETING THE LEUKAEMIC FUSION GENE
RUNX1/ETO BY SIRNA
L. Swart1,*, P. Derevyanko1, A. van Oort1, M. Ashtiani1, A. Krippner-Heidenreich1,
A. Koekman2, C. Seinen2, H. Issa3, H. Blair4, R. Schiffelers2, O. Heidenreich1,4
1Princess Maxima Center for Pediatric Oncology; 2Department of Central Diagnostic
Laboratory Research, University Medical Center Utrecht, Utrecht University, Utrecht,
Utrecht, Netherlands; 3Department of Pediatrics, University Hospital Frankfurt, Frankfurt
(Main), Germany; 4Wolfson Childhood Cancer Research Centre, Newcastle University,
Newcastle upon Tyne, United Kingdom
Background: The leukaemic fusion gene RUNX1/ETO initiates and drives leukaemogenesis
and, thus, constitutes an ideal cancer-specific target. Direct and exclusive targeting
of leukaemic fusion proteins using RNA interference is an attractive concept, but
has proven challenging because of the unfavourable pharmacokinetic properties of siRNA.
To address this challenge, we have developed lipid nanoparticle (LNP) formulations
that interfere with the expression in leukaemic cell lines and in primary cells. To
further harness their use in patient-derived AML cells, we are exploring and optimizing
targeted LNPs for effective siRNA delivery to primary leukaemic cells.
Aims: To develop and validate a delivery system for the therapeutic application of
fusion gene-specific siRNAs.
Methods: LNPs encapsulating siRE (active siRNA targeting the RUNX1/ETO fusion transcript)
and siMM (mismatch siRNA control with two nucleotides swapped in the sense strand)
were prepared using microfluidic mixing. Ligand or fluorescent dyes were conjugated
to polyethylene glycol (PEG) following a copper-free click chemistry approach. LNPs
were characterized by dynamic light scattering (DLS) and cryo electron microscopy
(EM). Functional experiments with patient-derived t(8;21) cells were performed in
co-culture with mesenchymal stromal cells under conditions that preserved and expanded
immature VLA-4 expressing leukaemic populations as determined by an 11 antibody panel
and flow cytometry analysis. Transcript and protein levels of RUNX1/ETO and target
genes were determined by bulk and single cell (sc) RNA-seq, real-time polymerase chain
reaction and western blotting.
Results: To monitor uptake of 50 – 60 nanometers sized LNPsiRNA we labelled the surface
with Cy3 and a ligand by a PEG post-insertion approach. To improve uptake and tissue
retention, we chose a modified short peptide ligand harbouring an LDV motif with high
affinity towards the very late antigen-4 (VLA-4) that is widely expressed across haematopoietic
cells. Decoration of the LNPs with the LDV resulted in an enhanced uptake in t(8;21)
cell lines within the first 8 hours. While undecorated LNPs were hardly taken up by
patient-derived material, the decoration with LDV resulted in an efficient uptake
and a concomitant knockdown of RUNX1/ETO transcript and protein after a single dose.
Sequential treatment leads to longer-lasting reduction of RUNX1/ETO transcript and
associated target genes (LAPTM5, CCND2, C/EBPA and ANGPT1) and loss of RUNX1/ETO protein.
Combined multiparameter flow analysis and scRNAseq of patient cells show that RUNX1/ETO
depletion causes massive shifts in cell populations and an almost complete loss of
immature populations (CD34+ CD38- CD117+ CD45RA+) associated with a gain in mature
(CD15+) populations, indicating a release from the differentiation block caused by
the fusion protein. Gene Set Enrichment Analysis (GSEA) of scRNAseq data confirm the
role of RUNX1/ETO in promoting stemness and cell cycle progression and highlight a
key function of this fusion protein in coordinating metabolism in support of leukaemic
propagation. Currently, we are performing in vivo biodistribution and efficacy studies
using orthotopic mouse models.
Summary/Conclusion: Decoration of LNPs with an LDV motif enhances uptake of fusion
transcript-specific siRNAs and enables efficacious knockdown of RUNX1/ETO in patient-derived
cells. The biologic consequences validate RUNX1/ETO as a highly promising and leukaemia-specific
target and highlight the LNP-mediated delivery of siRNAs as a promising new approach
for the treatment of fusion gene-driven leukaemias.
P466: AMPLIFIED EPOR/JAK2 GENES DEFINE A UNIQUE SUBTYPE OF ACUTE ERYTHROID LEUKEMIA
J. Takeda1,*, K. Yoshida1, M. Nakagawa1, Y. Nannya1, A. Yoda1, R. Saiki1, Y. Ochi1,
L. Zhao2, R. Okuda1, X. Qi1, T. Mori1, A. Kon1, K. Chiba3, H. Tanaka4, Y. Shiraishi3,
M.-C. Kuo5, C. Kerr6, Y. Nagata6, D. Morishita7, N. Hiramoto8, A. Hangaishi9, H. Nakazawa10,
K. Ishiyama11, S. Miyano4, S. Chiba12, Y. Miyazaki13, T. Kitano14, K. Usuki9, N. Sezaki15,
H. Tsurumi16, S. Miyawaki17, J. Maciejewski6, T. Ishikawa8, K. Ohyashiki18, A. Ganser19,
M. Heuser19, F. Thol19, L.-Y. Shih5, A. Takaori−Kondo20, H. Makishima1, S. Ogawa1
1Department of Pathology and Tumor Biology; 2Institute for the Advanced Study of Human
Biology, Kyoto University, Kyoto; 3Division of Genome Analysis Platform Development,
National Cancer Center Research Institute; 4M&D Data Science Center, Tokyo Medical
and Dental University, Tokyo, Japan; 5Division of Hematology−Oncology, Department
of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan,
Taiwan; 6Department of Translational Hematology and Oncology Research, Taussig Cancer
Institute, Cleveland Clinic, Cleveland, United States of America; 7Chordia Therapeutics
Inc., Kanagawa; 8Department of Hematology, Kobe City Medical Center General Hospital,
Kobe; 9Department of Hematology, NTT Medical Centre Tokyo, Tokyo; 10Department of
Hematology, Shinshu University Hospital, Matsumoto; 11Department of Hematology, Kanazawa
University, Kanazawa; 12Department of Hematology, Faculty of Medicine, University
of Tsukuba, Tsukuba; 13Department of Hematology, Atomic Bomb Disease Institute, Nagasaki
University, Nagasaki; 14Department of Hematology, Tazuke Kofukai Medical Research
Institute, Kitano Hospital, Osaka; 15Department of Hematology, Chugoku Central Hospital,
Hiroshima; 16Department of Hematology, Gifu University, Gifu; 17Division of Hematology,
Tokyo Metropolitan Ohtsuka Hospital; 18Department of Hematology, Tokyo Medical University,
Tokyo, Japan; 19Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation,
Hannover Medical School, Hannover, Germany; 20Department of Hematology / Oncology,
Kyoto University, Kyoto, Japan
Background: Acute erythroid leukemia (AEL) is a rare subtype of acute myeloid leukemia
(AML) characterized by erythroid predominant proliferation and classified into two
subtypes with pure erythroid (PEL) and erythroid/myeloid (EML) phenotypes based on
the degree of erythroid hyperplasia. Despite an intensive mutational analysis, the
mechanism of erythroid hyperplasia in AEL is still poorly understood and so are feasible
therapeutic targets.
Aims: To understand the mechanism of erythroid dominant phenotype of AEL and identify
potential therapeutic targets for AEL.
Methods: We analyzed a total of 124 adult AEL cases, where whole genome/exome sequencing
of 35 cases were followed by targeted-capture sequencing in all cases. RNA sequencing
was also performed in 23 cases. The mutational profile of AEL cases was compared to
that of 409 cases with non-erythroid AML (non-AEL). Patient-derived xenograft (PDX)
mouse models of AEL with JAK2 and/or EPOR amplification were established from 6 AEL
patients with these abnormalities. These models were tested for their response to
JAK1/2 inhibitor.
Results: In accordance with a recent report, AEL cases were classified into 4 genomic
groups (A-D), which are characterized by biallelic TP53 mutations and complex karyotype
(Group-A), mutated NPM1 (Group-B) and STAG2 (Group-C), and other mutations in histone
modifiers and transcription factors (Group-D). In particular, all but one PEL cases
belonged to Group-A. Also found in non-AEL cases, these group-defining lesions in
AEL were uniquely associated with focal gains/amplifications of EPOR, JAK2, and/or
ERG/ETS2 loci (Group-A), PTPN11 mutations (Group-B), and KMT2A-PTD (Group-C), which
might be responsible for the AEL phenotype. Highly enriched in PEL cases (7/13), EPOR/JAK2
focal gains/amplifications were implicated in their extreme erythroid hyperplasia
and associated with particularly poor prognosis, even compared to other Group-A cases,
who had shorter survival than those in other groups. As expected, JAK2/EPOR-amplified
cases showed upregulated STAT5 signaling compared to non-AEL cases, which however,
was also observed in other AEL cases, suggesting that upregulated STAT5 signaling
is a hallmark of AEL. Based on these findings, we tested the effect of JAK2 inhibition
on cell growth of EPOR/JAK2-amplified AEL cells in in vitro culture and xenograft
model. Of interest, ruxolitinib-mediated JAK2 inhibition resulted in significantly
suppressed in vitro cell growth of EPOR/JAK2-amplified AEL cells and prolonged overall
survival in 4 PDX models with phospho-STAT5 downregulation, although other 2 models
were resistant to JAK2 inhibition with persistent STAT5 activation.
Summary/Conclusion: AEL is a heterogenous subgroups of AML characterized in common
by upregulated JAK/STAT5 signaling. Gains/amplifications of JAK2/EPOR are frequent
in TP53-mutated cases, particularly those with the PEL phenotype, and could be exploited
as potential therapeutic targets using JAK2 inhibitors.
P467: FUNCTIONAL CHARACTERIZATION OF ABERRANT GATA1 PROTEIN COMPLEXES IN NORMAL AND
MALIGNANT HUMAN ERYTHROBLASTS
S. Tauchmann1,*, F. Otzen Bagger2, T. Bock3, R. Sivalingam1, T. Eder4, E. Heyes4,
A. Fagnan5, M. von Lindern6, T. Mercher5, F. Grebien4, J. Schwaller1
1Childhood Leukemia, University of Basel, University Children`s Hospital Basel, Department
Biomedicine, Basel, Switzerland; 2University of Copenhagen, Center of Genomic Medicine,
Copenhagen, Denmark; 3Proteomics Core Facility, Biozentrum, University of Basel, Basel,
Switzerland; 4Institute for Medical Biochemistry, University of Veterinary Medicine
Vienna, Vienna, Austria; 5INSERM U1170, Equipe Labellisée Ligue Contre le Cancer,
Institut Gustave Roussy, Université Paris-Saclay, Université de Paris, Paris, France;
6Department Hematopoiesis, Sanquin Research, Academic Medical Center, University of
Amsterdam, Amsterdam, Netherlands
Background: Terminal differentiation of erythroid progenitor cells is controlled by
the master transcription factor GATA1 acting in activating and repressive protein
complexes. In acute erythroid leukemia (AEL), cell differentiation is impaired leading
to accumulation of immature erythroblasts. Previous work in mouse models and primary
human cells suggested impaired function of abundantly expressed GATA1 protein in AEL
cells. Interestingly, GATA1 overexpression induced or enhanced terminal erythroid
differentiation of the human AEL cell line K562 or immortalised HUDEP2 erythroblasts,
respectively.
Aims: We hypothesize that impaired terminal erythroid differentiation in AEL might
be the consequence of aberrant GATA1 protein interactions.
Methods: We compared the proteomes of three human AEL cell lines (F36P, K562, KMOE2)
and primary AEL patient cells with HUDEP2 cells and primary human erythroblasts (hEBST)
that were capable of in vitroterminal erythroid differentiation, using a tandem mass
tag(TMT)-based approach (n=3/type). In parallel, we compared the GATA1 interactomes
by immunoprecipitation (IP) followed by mass spectrometry (MS) (n=3/type). In addition,
we performed a targeted CRISPR screen in Cas9-expressing K562 cells in which we monitored
growth kinetics upon mutational disruption of candidate genes and simultaneously assessed
changes in erythroid differentiation by measuring CD71 and CD235a surface markers.
Results: Unsupervised hierarchical clustering displayed a clear separation of AEL
proteomes from those of normal erythroblasts with 386 proteins higher expressed in
the first and 623 proteins more abundant in the latter group (logFC≥2;q<0.05). GATA1-IP-MS
analysis of nuclear lysates from all six cell types identified 1616 proteins, of which
126 were differentially associated with GATA1 depending on cell state. 54 proteins
preferably interacted with GATA1 in the AEL group, whereas 72 proteins were more enriched
in “normal erythroblasts” (q<0.5). GATA1 interactomes in hEBST and HUDEP2 cells highly
overlapped and were more similar to GATA1 partners identified in F36P and KMOE, than
those of K562 and primary AEL cells. Integrative analysis revealed 118 proteins that
were differentially expressed and/or precipitated by GATA1-IP, of which 49 were enriched
in malignant and 69 proteins in normal erythroblasts (q<0.5). To identify if GATA1-associated
proteins are involved in erythroid differentiation we functionally characterized them
in a CRISPR screen in K562 cells. 4 patterns were observed: inactivation of cluster
1 genes such as ZEB2 or IKZF1 did not change cell differentiation or growth, loss
of cluster 2 genes like RPS21 or RANGAP1 did not change differentiation, but impaired
cell growth/survival. Disruption of cluster 3 genes like SND1, or EIF5A resulted in
an increase in CD71 and/or CD235a expression without significant changes in cell growth,
and deletion of cluster 4 genes including SMC1A or ATP1A1 caused significant signs
of differentiation together with cell depletion over time. Overall, knockout of 41
genes caused significantly increased CD71 and/or CD235a surface expression, indicating
that these GATA1-interacting proteins affect erythroid differentiation.
Summary/Conclusion: Our work suggests that impaired erythroid differentiation of AEL
cells involves aberrant GATA1 protein complexes. Ongoing gain- and loss of function
validation experiments of selected candidate genes and chromatin assays will identify
novel regulators of terminal differentiation in normal and malignant erythropoiesis.
P468: MRD AS A BIOMARKER FOR RESPONSE TO DONOR LYMPHOCYTE INFUSION AFTER AL-LOGENEIC
HEMATOPOIETIC CELL TRANSPLANTATION IN PATIENTS WITH ACUTE MYELOID LEUKEMIA
K. Teich1,*, M. Stadler1, R. Gabdoulline1, C. Wienecke1, B. Heida1, P. Klement1, K.
Büttner1, L. Venturini1, M. Wichmann1, W. Puppe2, C. Schultze-Florey1, G. Beutel1,
M. Eder1, A. Ganser1, M. Heuser1, F. Thol1
1Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation; 2Department
of Virology, Hannover Medical School, Hannover, Germany
Background: Donor lymphocyte infusions (DLIs) can be employed for acute myeloid leukemia
(AML) patients as therapeutic or prophylactic treatment after allogeneic hematopoietic
cell transplantation (alloHCT). Application of DLI after alloHCT may induce graft-versus-host
disease (GVHD). Therefore, biomarkers that predict the effect of DLIs may be useful
to guide DLI use. Molecular measurable residual disease (MRD) is prognostic for the
response to standard chemotherapy as well as for targeted therapies.
Aims: To evaluate MRD as a prognostic marker for DLIs after alloHCT.
Methods: Patients with AML or myelodysplastic syndrome (MDS) aged ≥18 years undergoing
DLI at Hannover Medical School between 1998 and 2018 with available PB and/or BM DNA
samples were included. Patients were excluded if they had no detectable mutations
at diagnosis, had a second alloHCT in the observed time or if MDS patients had <10%
bone marrow blasts at diagnosis.
DNA libraries were prepared using the TruSight Myeloid Panel or Nextera Flex for enrichment
(Illumina) and sequenced on a MiSeq device. Error-corrected sequencing of patient
specific mutations was performed 30 days (Follow Up 30 days, FU30) and 90 days (FU90)
after the first DLI. All mutations detected at diagnosis were used for MRD monitoring.
Marker positivity was defined as variant allele frequency above the limit of detection
as not all patients were in complete remission (CR), in CR with incomplete hematological
recovery (CRi) or in morphologic leukemia free state (MLFS).
Results: The median time from alloHCT to DLI was 11.3 months (range 3.1 to 74.4 months).
Of 78 patients, 24 patients were marker negative and 54 patients were marker positive
before DLI (of the marker positive patients 23 were in CR/CRi/MLFS). As expected,
marker negative patients before DLI had a significantly better overall survival (OS),
event-free survival (EFS) and relapse-free survival (RFS) than marker positive patients.
From the 54 marker positive patients before DLI, 27 patients were in CR/CRi at FU90,
22 patients did not reach CR/CRi and five patients died before FU90. OS, EFS and RFS
were significantly improved in patients that were in CR/CRi at FU90. CR/CRi status
at FU90 correlated strongly with remission status before DLI (p<0.001).
Next, the prognostic effect of MRD in CR/CRi patients at FU90 (n=27) was analyzed.
18 patients reached MRD negativity at FU90 and nine patients stayed MRD positive.
Baseline characteristics such as patient sex, cytogenetic risk group, age at diagnosis,
the median time between alloHCT and DLI or use of prophylactic vs. therapeutic DLIs
were similar between MRD negative and positive patients. Treatment intensity until
FU90 was similar between the two groups. Remission status before DLI was not associated
with MRD negativity at FU90 (p=0.782). There was no significant difference in OS (p=0.593),
EFS (p=0.229) and RFS (p=0.226) between FU90 MRD negative and positive patients. Exclusion
of DTA (DNMT3A, TET2, ASXL1) mutations as MRD markers affected the number of patients
in CR/CRi at FU90 (20 patients MRD negative and five patients MRD positive), while
patient characteristics and outcome did not differ between the two groups.
Summary/Conclusion: Patients that achieve CR/CRi until FU90 after DLIs have a significantly
better outcome than the remaining patients. However, the MRD status in patients in
CR/CRi at FU90 had no prognostic effect in the patient cohort investigated here.
P469: CHEMORESISTANCE IN ACUTE MYELOID LEUKEMIA: A SINGLE-CELL RNA SEQUENCING APPROACH
C.-L. Teng1,*, P.-L. Cheng1, T.-H. Hsiao2, C.-H. Chen3
1Division of Hematology/Medical Oncology; 2Department of Medical Research, Taichung
Veterans General Hospital, Taichung; 3National Institute of Cancer Research, National
Health Research Institutes, Zhunan, Taiwan
Background: Our previous study demonstrated that myc, mitochondrial oxidative phosphorylation,
mTOR, and stemness were independently responsible for chemoresistance in AML.
Aims: The current study aimed to further identify the potential mechanisms of chemoresistance
of the “7 + 3” induction in AML using a single-cell RNA sequencing (scRNA-seq) approach.
Methods: Thirteen untreated de novo AML patients were enrolled and stratified into
the complete remission (CR) (n=8) and the non-CR (n=5) groups. We used scRNA-seq to
analyze the genetic profiles of 28950 AML cells from these 13 patients. A previously
published bulk RNA-seq dataset validated our results.
Results: Chemoresistant AML cells had more premature accumulation in early hematopoiesis.
The hematopoietic stem cell-like cells from the non-CR group expressed more leukemic
stem cell markers (CD9, CD82, IL3RA and, IL1RAP) than those from the CR group. Besides,
chemoresistant progenitor cells had impaired myeloid differentiation due to early
hematopoiesis arrest. Importantly, the AML cells analyzed by the scRNA-seq and bulk
RNA-seq harbored comparable myeloid lineage cell fraction, which internally validated
our results. Using the TCGA database, our analysis demonstrated that AML patients
with a higher expression of chemoresistant genetic markers (IL3RA and IL1RAP) had
a worse overall survival (p < 0.01 for IL3RA; p < 0.05 for IL1RAP).
Image:
Summary/Conclusion: The AML cells responsive and resistant to the “7 + 3” induction
had diverse cell origins according to specific genetic biomarkers using a scRNA-seq
approach. Besides, hematopoiesis arrest occurred earlier in chemoresistant AML cells.
This result provided an insight into a more understanding of chemoresistance in AML.
P470: NGS-BASED MINIMAL RESIDUAL DISEASE DETECTION IN PERIPHERAL BLOOD SHOWS GOOD
PROGNOSTIC VALUE FOR OS AND EFS IN PATIENTS WITH ACUTE MYELOID LEUKEMIA RECRUITED
IN THE UNIFY TRIAL
C. Thiede1,2,*, C. Schuster2, C. Krippendorf2, G. Koenen3, T. Medts3, P. Marques Ramos3,
L. Gou4, P. Montesinos5, W. Fiedler6, R. Müller7, J. Krauter8, S. Sica9, J. Westermann10,
M. Levis11, R. Stone12, J. Sierra13, H. Döhner14
1Medizinische Klinik und Poliklinik, Universitätsklinikum Carl Gustav Carus an der
Technischen Universität Dresden; 2AgenDix, Gesellschaft für angewandte molekulare
Diagnostik mbH, Dresden, Germany; 3Novartis Pharma AG, Basel, Switzerland; 4Novartis
Pharmaceuticals Corporation, East Hanover, NJ, United States of America; 5Hospital
Universitario i Politecnico La Fe Valencia, Valencia, Spain; 6Med. Klinik, Universitätsklinikum
Hamburg Eppendorf, Hamburg, Germany; 7Klinik für Medizinische Onkologie und Hämatologie,
Universitäts Spital Zürich, Zürich, Switzerland; 8Städtisches Klinikum Braunschweig,
Braunschweig, Germany; 9Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica
ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy;
10Charité University Medical Center, Berlin, Germany; 11The Sidney Kimmel Comprehensive
Cancer Center, Johns Hopkins University, Baltimore, MD; 12Dana-Farber Cancer Institute,
Boston, United States of America; 13Hospital Santa Creu i Sant Pau, Universitat Autonoma
of Barcelona, Jose Carreras Leukemia Research Institute, Barcelona, Spain; 14Universitätsklinikum
Ulm, Ulm, Germany
Background: Assessment of measurable residual disease (MRD) to evaluate the depth
of remission at the time of achieving morphological complete remission (CR) or CR
with incomplete blood count recovery (CRi) by multiparameter flow cytometry (MFC)
or quantitative polymerase chain reaction (qPCR) has been shown to be predictive of
outcome in patients (pts) with acute myeloid leukemia (AML), and is recommended according
to European LeukemiaNet guidelines. However, accurate MRD assessment is still limited
by lack of suitable markers in all pts (qPCR) and/or limited specificity/sensitivity
(MFC). Next generation sequencing (NGS) holds promise to overcome some of these limitations
and allows versatile and sensitive MRD assessment in almost all AML pts. However,
data on NGS-MRD in this setting are still limited.
Aims: To further extend the experience with NGS-based MRD assessment, we prospectively
collected samples to explore prognostic implications of MRD as detected by NGS at
CR/CRi in the context of a randomized, placebo-controlled phase 3 study (UNIFY; NCT03512197),
investigating midostaurin added to conventional 7 + 3 based chemotherapy in FLT3wt
pts. Following the recommendation by an independent DMC, the UNIFY study was stopped
in Sep 2019 due to futility. The current subset analysis was performed to investigate
the impact of NGS-MRD results on long-term outcomes.
Methods: Peripheral blood mononuclear cell (PBMC) or bone marrow mononuclear cell
(BMMC) samples were collected from pts treated in the UNIFY study. NGS-MRD was performed
on genomic DNA using targeted NGS-error-controlled sequencing method (Ion-Torrent
S5XL) with a sensitivity between 0.1 and 0.001%. Targets for MRD detection were identified
by NGS of BM samples taken at diagnosis (Archer Myeloid Panel) covering 74 genes frequently
mutated in myeloid disease. MRD positivity was defined as any mutation call >0.1%,
excluding mutations in genes associated with clonal hematopoiesis, ie., DNMT3A, TET2
and ASXL1.
Results: Total 731 BMMC (from 305 out of the total 501 pts) and 564 PBMC samples (from
204 pts) were collected and analyzed, of which 467 were paired (from 175 pts). The
10 most frequently used genes for MRD monitoring were NRAS, IDH2, RUNX1, SRSF2, TP53,
PTPN11, IDH1, BCOR, CEBPA and GATA2. Analyses in paired samples showed numerically
higher MRD+ rates in BMMC vs. PBMC (57% vs 46% MRD+ at the end of induction, respectively).
No differences in MRD rates were seen between treatment arms, and data was pooled
for analysis. A high correlation of variant allele frequency (Pearson’s r≥0.84) and
a high level of concordance in MRD calls were seen between BMMC and PBMC (91% overall
percent agreement). For pts who fulfilled the criteria of a successful induction treatment,
detection of MRD in either BMMC or PBMC in CR/CRi pts at the end of induction was
associated with significantly lower event free survival (EFS) and overall survival
(OS) compared to MRD− status (1-year EFS of 26% vs 72% in BMMC and 34% vs 71% in PBMC;
1-year OS of 73% vs 93% in BMMC and 67% vs 94% in PBMC) (Figure). Preliminary multivariate
analysis, controlling for age and sex, confirmed the independent prognostic value
of NGS-MRD for OS. Additional analysis is ongoing.
Image:
Summary/Conclusion: Taken together, these data indicate that MRD by NGS at the time
of CR/CRi in both BMMC and PBMC, is prognostic for EFS and OS. Although the results
should be interpreted with caution due to the premature termination of the study and
limited follow-up, these further support the use of molecular MRD assessment to predict
outcome in pts with AML.
P471: ERYTHROID/MEGAKARYOCYTIC DIFFERENTIATION BIAS IN BONE MARROW OF AML/MDS PATIENTS
AFTER DECITABINE TREATMENT
F. Tiso1,*, A. O. de Graaf1, L. I. Kroeze2, B. A. van der Reijden1, S. M. C. Langemeijer3,
K. M. Hebeda2, J. H. Jansen1
1Department of Laboratory Medicine, Laboratory of Hematology; 2Department of Pathology;
3Department of Hematology, Radboudumc, Nijmegen, Netherlands
Background: The hypomethylating agent Decitabine is used mainly in AML patients who
are not eligible for standard induction chemotherapy or as a bridge therapy before
a stem cell transplantation. Decitabine is an analogue of cytidine deoxynucleoside
and it is incorporated into the DNA as decitabine triphosphate. In the DNA decitabine
triphosphate inhibits the methylation of the DNA, leading to the reactivation of tumour
suppressor genes which have been epigenetically silenced, induces cell differentiation
and also apoptosis.
Aims: We studied early signs of morphological changes in bone marrow biopsies (BMB)
of AML/MDS patients after treatment with Decitabine and correlated these with the
mutational background.
Methods: We investigated 45 patients with AML/MDS who were treated with Decitabine.
Bone marrow biopsies were morphologically examined, and we assessed the mutational
status of the BMB. Sequencing was performed using a panel of 27 commonly mutated genes
in myeloid malignancies, using a targeted error corrected approach.
Results: We observed a striking erythroid/megakaryocytic differentiation bias after
treatment in 16/45 patients, whereas the other patient group (n=29) showed predominant
myeloid differentiation. Patients developing this erythroid/megakaryocytic bias showed
a trend towards a higher frequency of mutations in genes encoding for spliceosome
components (U2AF1, SF3B1 and SRSF2), in particular U2AF1 was found significantly (p-value=0.05)
more mutated. Furthermore, this group carried a significantly (p-value=0.03) lower
number of mutations in genes encoding for proteins involved in signalling and kinase
pathways (ETNK1, FLT3, CBL, JAK2, KIT, KRAS, MPL, BRAF, NRAS) compared to the group
with predominant myeloid differentiation.
Summary/Conclusion: A bias towards erythroid/megakaryocytic differentiation was observed
in the bone marrow of one third of the patients after treatment with Decitabine. This
bias was over-represented in patients with mutations in spliceosome component encoding
genes, particularly U2AF1. Patients with myeloid predominant Decitabine response harboured
an increased number of mutations in signalling and kinase proteins encoding genes.
P472: THE FLT3-ITD RECEPTOR CAN PROMOTE PROTEIN FOLDING IN THE ENDOPLASMIC RETICULUM
THROUGH THE OXIDOREDUCTASE ERO1
M. Turos Cabal1,*, A. M. Sánchez Sánchez1, N. Puente Moncada1, C. Rodriguez1, F. Herrera2,
V. Martin1
1Morphology and Cellular Biology, University of Oviedo, Oviedo, Spain; 2Department
of Chemistry and Biochemistry, University of Lisbon, Lisbon, Portugal
Background: Acute myeloid leukemia (AML) is an aggressive hematologic malignancy and
the most common myeloid tumour in adults. Internal tandem duplications (ITD) in FLT3
receptor appear in 30% of cases and is associated with poor disease outcome. This
receptor is synthesized and processed in the endoplasmic reticulum (ER) before being
transported to the plasma membrane, where it is activated by ligand binding. Upon
activation, it regulates cell differentiation, proliferation, and survival. Mutations
in FLT3, such as ITD mutations, not only constitutively activate this receptor, but
also prevent its correct processing in the endoplasmic reticulum. Therefore, it is
mostly retained in this organelle.
Aims: We decided to study if the aberrant accumulation of FLT3-ITD affects the functioning
of the ER.
Methods: Experiments were performed on wild-type and FLT3-ITD AML cell lines, as well
as on stably transfected FLT3 and FLT3-ITD Ba/F3 cell lines. Protein aggregation,
reactive oxygen species (ROS) and glutathione (GSH) levels were determined by Thioflavin
T, DCFH-DA and Thioltracker Violet fluorescence, respectively. Protein detection was
performed by Western Blot. Cell viability and death was analysed by cell count and
Trypan Blue exclusion.
Results: Protein folding in the ER generates reactive oxygen species (ROS). Antioxidant
molecules such as GSH are needed to maintain redox homeostasis. We found that FLT3-ITD
cell lines have lower levels of protein aggregates, as well as ROS and GSH. This data
could suggest that mutant cell lines have more efficient protein folding mechanisms.
In fact, among the ER proteins that are involved in the protein folding process, we
found the oxidoreductase ERO1 to be increased in FLT3-ITD cell lines. Supporting our
findings, we saw that FLT3-ITD cell lines were less sensitive to protein folding blocking
agents, DTT and 2-mercaptoethanol. This was reflected by a slight decrease in cell
proliferation compared to wild-type cell lines. We confirmed that the decrease in
cell viability in FLT3-ITD cell lines was not accompanied by cell death, as opposed
to wild-type cell lines. Under ER stress conditions, such as an accumulation of misfolded
proteins in the ER, the unfolded protein response (UPR) is activated. We decided to
study if there were differences in the basal levels of UPR by western blot. We found
FLT3-ITD cell lines to have lower levels of UPR, reflected by lower protein expression
of the active forms of PERK and IRE (p-PERK and p-IRE respectively), together with
the protein BiP, the major chaperone found in the ER and binds to misfolded proteins.
Altogether, our data suggest that FLT3-ITD cell lines have less ER stress, reflected
by lower levels of chaperones and UPR proteins, possibly because they rely on more
efficient protein folding mechanisms. To confirm the importance of protein folding
process in acute myeloid leukemia, we tested the effect of ERO1 inhibitor (EN460),
and found it to have a greater impact in FLT3-ITD cell lines. In order to determine
a possible relation between the ITD mutation in the FLT3 receptor and ERO1, we used
stably transfected Ba/F3 cell lines carrying the normal or mutated FLT3 receptor.
We found ERO1 to be overexpressed on FLT3-ITD Ba/F3 cell lines.
Summary/Conclusion: Current AML treatments are mainly based on the administration
of FLT3 inhibitors, with limited success. Our data suggest that FLT3-ITD cell lines,
with greater resistance to treatments, have more efficient protein folding mechanisms.
This process should be studied in more detail to determine its potential as a therapeutic
target for AML treatment.
P473: DEVELOPMENT OF POTENTIAL BIOMARKERS FOR IRAK4 INHIBITOR EMAVUSERTIB IN HUMAN
ACUTE MYELOID LEUKEMIA
A. Ugolkov1,*, M. Pilichowska2, C.-C. Li1, R. C. Hok1, M. Samson1, M. Lane1, R. Von
Roemeling1, R. Martell1
1Curis, Inc., Lexington, MA; 2Tufts Medical Center, Boston, MA, United States of America
Background: Acute myeloid leukemia (AML), the second most common form of leukemia
in adults, remains a highly fatal disease. Interleukin-1 receptor-associated kinase
4 (IRAK4) has been demonstrated as a potential therapeutic target in human AML. IRAK4-mediated
activation of NF-kappaB signaling pathway could play a critical role in NF-kappaB-regulated
survival and chemoresistance of cancer cells. The results of ongoing Phase 1 study
demonstrated clinical activity of IRAK4 inhibitor emavusertib (CA-4948) in patients
with relapsed/refractory AML and high-risk MDS. To support the development of companion
diagnostic for emavusertib, we developed an immunohistochemical (IHC) assay and explored
expression of potential biomarkers in bone marrow (BM) samples obtained from AML patients.
Aims: The objective of the present study was to analyze expression of IRAK4, NF-kappaB
p-p50 and NF-kappaB p-p65 by IHC staining in human AML with further evaluation of
these molecules as potential biomarkers for emavusertib therapy in AML patients.
Methods: We used IHC staining and immunoblotting to determine the expression of IRAK4,
NF-kappaB p-p50 S337 and NF-kappaB p-p65 S536 proteins in human leukemia cell lines
and clinical AML samples. Exploratory biomarker evaluations were performed using serial
sections of formalin-fixed paraffin-embedded BM clot samples obtained from 19 AML
patients. Eight BM AML samples were purchased from Analytical Biological Services
and 11 BM samples were obtained at the screening from evaluable patients with relapsed/refractory
AML enrolled in CA-4948-102 clinical trial.
Results: Using IHC staining, we found IRAK4 nuclear expression in blasts in 9/19 AML
cases. To the best of our knowledge, this is the first report showing nuclear accumulation
of IRAK4 in cancer cells. In support of our findings in clinical AML samples, we detected
IRAK4 protein expression in nuclear lysates prepared from leukemia cell lines THP-1,
HL-60 and K562. Using AML BM samples, we found that IRAK4 nuclear expression in blasts
was significantly correlated with activation of NF-kappaB as determined by nuclear
accumulation of NF-kappaB p-p50 and p-p65 in 9/19 cases. Cytoplasmic expression of
IRAK4 was detected in 2/19 cases whereas IRAK4, NF-kappaB p-p50 and p-p65 expression
was not detectable in 8/19 cases of AML. Clinical response data will be presented
in the context of these novel findings.
Summary/Conclusion: Our findings uncovered a previously unknown nuclear expression
of IRAK4 in leukemia cells. Our results demonstrated co-expression of nuclear IRAK4,
NF-kappaB p-p50 and p-p65 in blasts suggesting a potential novel mode of interaction
between IRAK4 and NF-kappaB in human AML. Although the role of nuclear IRAK4 in leukemia
cells remains to be investigated, our preliminary findings revealed new perspectives
into emavusertib treatment stratification and demonstrate the possibility of discovering
novel biomarkers through IHC analysis of clinical samples. Additional accrual is ongoing
to better define emavusertib biomarkers in AML patients.
P474: PIEZO1 AND THE THERAPEUTIC POTENTIAL OF MECHANORECEPTORS IN ACUTE MYELOID LEUKAEMIA
M. Velasco Estevez1,*, A. Fernandez1, A. Otero-Sobrino1, P. Aguilar-Garrido1, M. A.
Navarro-Aguadero1, R. Sanchez2, A. Gimenez Sanchez2, V. Garrido Garcia2, L. Carneros
Blanco2, J. Martinez-Lopez2, M. Gallardo1
1H12O-CNIO Haematological Malignancies Group, Clinical Research Unit, Centro Nacional
de Investigaciones Oncologicas (CNIO); 2Grupo de Hematología Traslacional, Hospital
12 de Octubre de Madrid, Madrid, Spain
Background:
Mechanotransduction is the process through which cells sense the mechanical input
in and out the cell, and translate it into biochemical signals adapt and respond.
In the past few years, it has been observed the important contribution of mechanical
input in cell differentiation, fate, stemness, senescence and cancer, amongst others.
Piezo1 is a mechanoreceptor discovered in 2010 and constitutes a cation channel activated
through stretch and shear flux leading to a calcium influx in the cell that activates
a myriad of different pathways. Interestingly, despite being a mechanoreceptor, Piezo1
can also be activated through a small molecule, Yoda-1, and blocked by a peptide,
GsMTx4, allowing the pharmacological study of this receptor. Mutations in PIEZO1 lead
to pathologies as Xerocitosis hereditaria and it has been linked to erythroid formation.
However, the expression and role of Piezo1 in haematological malignancies is yet to
be explored
Aims: We aim to analyse the expression of Piezo1 in Acute Myeloid Leukaemia (AML)
and healthy donors, and investigate the putative role of Piezo1 in cell differentiation
and fate as well as its therapeutic potential for AML.
Methods: Mouse embryonic stem cells (mESC) were cultured and treated with Yoda-1 and
GsMTx4. PIEZO1 expression in AML and healthy donors was analysed through RNAseq databases
(Bloodspot, GEPIA2 and MILE study) and corroborated by ICC staining of human samples.
Lastly, mononuclear cells in viability from AML or healthy donors were cultured in
vitro for dose-response curves to Yoda-1 and GsMTx4 and in methylcellulose to analyse
the phenotype and proliferation under those treatments. Both imaging and immunophenotyping
with flow cytometry were performed in those samples.
Results: When analysing the expression of PIEZO1 in AML, it was seen that Piezo1 was
higher in AML compared to healthy donors, and interestingly, there was a much higher
expression in AML inv(16), same chromosome as PIEZO1 is (Figure 1A). We observed that
activation of Piezo1 in mESCs led to a higher expression of stemness marker Oct3/4
(Figure 1B), suggesting that it is involved in the stemness state of haematopoietic
stem cells. Furthermore, we observed a dramatic effect of both Yoda-1 and GsMTx4 in
leukaemia haematopoietic cells (LSCs) from AML patients grown in methylcellulose,
where GsMTx4 caused an increment in the cell number and shifted the cell fate towards
the erythroid lineage, while Yoda-1 decreased the number of cells and ablated the
erythroid differentiation. Interestingly, such effects were barely significant in
cells from healthy donors, showing a wide window for treatment (Figure 1C-D).
Image:
Summary/Conclusion: Our study shows that Piezo1 changes with cell differentiation
and it could be a mechanism through which stem cells direct their fate. Furthermore,
Piezo1 is highly expressed in AML and its blockage leads to a shift towards the erythroid
differentiation while its activation blocks cell proliferation and differentiation,
which such effects being much more significant in AML samples than healthy donors,
suggesting a potential as therapeutic target.
This work was financially supported by CRIS contra el Cancer Association (NGO) AES
ISCIII (PI18/00295), ISCIII Miguel Servet (CP19/00140) and IF-Marie Sklodowska Curie
Actions grant (MAtChinG – 101027864).
P475: CONTROL LEUKEMIA BY INDUCING ANTI-CANCER IMMUNE REACTIVITY IN VIVO? POTENTIAL
OF A DC-TRIGGERED MECHANISM
G. Filippini Velazquez1,*, D. Amberger2, L. Klauer2, E. Rackel2, M. Atzler2, S. Ugur2,
C. Plett2, A. Rabe3, C. Kugler2, A. Rank1, M. Inngjerdingen3, T. Baudrexler2, B. Eiz-Vesper4,
C. Schmid1, H. Schmetzer2
1Department of Hematology and Oncology, Section for Stem Cell Transplantation, Augsburg
University Hospital, Augsburg; 2Medical Department III, Department for Hematopoietic
Cell Transplantation, Munich University Hospital, Munich, Germany; 3Institute of Clinical
Medicine, Department of Immunology, University of Oslo, Oslo, Norway; 4Institute of
Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover,
Germany
Background: There are virtually no treatment options for therapy-refractory or relapsed
AML/MDS and high rates of relapse in successfully treated patients. The combination
of the (clinically approved) immune-modulatory compounds GM-CSF+ Prostaglandine (PGE)1,
the combination referred to as KIT-M converts myeloid blasts into dendritic cells
of leukemic origin (DCleu). After stimulation with DCleu, antileukemic (T)cells are
activated.
Aims: Kit-M treatment may be an attractive tool for immunotherapy in myeloid leukemia.
Methods: Generation of DC from leukemic whole blood (WB) samples. Mixed lymphocyte
culture (MLC) followed by functional antileukemic assays.
Results: 1.
ex vivo
: Treatment of 65 leukemic WB samples with KIT-M does not induce blast proliferation,
but triggers generation of mature DC/DCleu and reduces tolerogenic DC. Kit treated
WB activates the adaptive and innate immune system after MLC (Tcell proliferation,
antitumor-supportive Tcells (TCRgd,Tb7), memory cells (Tcm,Tb7cm) and downregulates
immunosuppressive Tcells (Treg4 and 8). Moreover leukemia-specific (interferon g (g)
and/or degranulating (deg)) adaptive (g-degT4,T8,TCRgd,Tb7,Tcm) and innate cells (g-degNK,NKb7,CIKb7)
are increased and regulatory cells (g-degTreg4) downregulated. In addition, blast
lysis is increased vs control. Ex vivo achieved blast lysis correlates positively
with frequencies of mature DC/DCleu, leukemia-specific T3,T4,T8,TCRgd,Tb7 and NK cells
and negatively with Treg4 and 8. Blast lysis does not correlate with age, sex, ELN
risktype, blast counts, or response to chemotherapy.
2.
In vivo
- rats:
Kit-M treatment of 3 leukemically diseased (vs 3 control) rats (followed by sacrification
after treatment) leads to reduced blasts and Tregs in blood and spleen and increased
DCleu and memory-like Tcells.
3.
In vivo
- human:
Kit-M therapy was offered to a 72 year old pancytopenic male as an individual salvage
attempt (applied as continuous infusion), after discussion with the ethical commitee,
the patient’s information about the experimental nature of the treatment and his written
consent. The treatment was well tolerated and the patient improved clinically. Neutrophils
in WBC increased from 10% to 50%, thrombocytes reached 100 G/l after 24 days. Immune
monitoring showed a continuous increase of proliferating and non-naïve Tcells, NK,
CIK- and NKT-, TH17 cells, Bmem-cells and DC in PB. The production of IFNg producing
T-, CIK and NKT-cells was demonstrated, suggesting an in vivo production/activation
of (potentially leukemia-specific) cells. Immune stimulatory effects decreased after
discontinuation of therapy. After 4 weeks of treatment, the patient was discharged
in good clinical condition. Unfortunately, at two weeks from discharge, AML progressed
and the patient died few days later.
Summary/Conclusion: Treatment of WB ex vivo with Kit-M leads to activation of adaptive
and innate (leukemia-specific) immune reactive cells (and downregulated suppressive
mechanisms) via a DC/DCleu triggered mechanism – resulting in significantly improved
blast lysis compared to controls (independent of patients‘ risk classification, MHC,
age or sex). In vivo treatment of leukemically diseased rats or humans was well tolerated,
led to an increase of platelets and granulocytes and stable (low) blast counts in
PB – probably mediated by a (leukemia specifically) DC/DCleu activated immune system.
A dose defining clinical trial in carefully selected patients to confirm clinical
safety and underscore clinical efficacy is being prepared.
P476: LOSS OF CHD4 IN ACUTE MYELOID LEUKAEMIA GIVES RISE TO INCREASED DNA DAMAGE REPAIR
DEFECTS AND ALTERED CELL CYCLE PROGRESSION
D. Venney1,*, Y. Atlasi2, A. Mohd-Sarip2, K. Mills1
1Haematology; 2Queens University Belfast, Belfast, United Kingdom
Background: Acute Myeloid Leukaemias (AMLs) are an array of blood cell disorders arising
from alterations within myeloid precursors. Cancer genome studies highlighted alterations
within the nucleosome remodelling and deacetylation (NuRD) complex occur in over 20%
of all cancers. The NuRD complex controls nucleosome assembly and chromatin accessibility;
CHD4 is a core subunit of the NuRD which is involved in a multitude of functions including
epigenetic regulation, cell cycle progression and DNA replication and repair. CHD4
is linked to PARP dependent DNA damage response (DDR) pathway by rapid but transient
accumulation at sites of damage, preventing transcription which enables homologous
recombination (HR).
Aims: We aim to explore the function of CHD4-NuRD in AML using a CHD4 knockout (CHD4-/-)
isogenic cell line models created using CRISPR/Cas9 to assess the mechanism of function
of CHD4 within AML disease progression and evaluate if there is targetable therapeutic
potential.
Methods: CRISPR-Cas9 was used to generate isogenic CHD4-/- models in two representative
AML cell lines. CHD4-/- models underwent functional studies; proliferation, cell cycle
and protein expression assays were used to phenotype models before inducing DNA damage
at a clinically relevant dose and reassessing cell response. A high throughput drug
screen was performed to identify DNA damage compounds that showed significant sensitivity
in CHD-/- models and validation was assessed using single compound treatments and
co-culture assays.
Results: Analysis of publicly available TCGA AML transcriptomic data set (N=161),
found that AML patients (N=82) who express below median CHD4 levels have a poorer
prognostic outcome suggesting a link between stalled transcription enabling HR-based
DDR and patient survival (P=0.047). Analysis of the intermediate cytogenetic risk
group showed patients with a low CHD4 expression had a worse outcome; which was comparable
to the adverse risk group patients (P=0.032), indicating that loss of CHD4 may contribute
to a more aggressive phenotype AML in this group.
Phenotype analysis of CHD4-/- models showed DNA damage markers 53BP1 and PARP expression
increases significantly, highlighting the role of CHD4 within the DDR. This increased
DNA damage response was accompanied by reduced cellular proliferation in CHD4-/- cells.
Accordingly, cell cycle analysis showed a decrease in cells entering the G2/M phase
alongside an increase in cells in G1 and S phase suggesting a potential cell cycle
checkpoint deficiency causing stunted proliferation in CHD4-/- cells. Of note, we
did not observe a significant compensatory increase in CHD3-NuRD components expression
in the absence of CHD4 suggesting there is no compensation for the loss of CHD4.
We measured behaviour of CHD4-/- cells to stress-inducing conditions; in response
to irradiation and chemotherapy drugs commonly used in AML. CHD4-/- cells display
much stronger reduction in cellular proliferation post-2Gy irradiation compared to
control while cell cycle analysis showed further cellular arrest in G2/M. Furthermore,
a drug screen (160 DNA damaging compounds) showed an increased sensitivity of CHD4-/-
cells to compounds targeting DNA/RNA synthesis pathways, including Tubercidin and
Cytarabine, indicating that loss of CHD4 results in hyper-increase in damage.
Summary/Conclusion: Our data has identified a role of CHD4 in AML progression mediated
through loss of cell cycle progression control and an elevated response to DNA damage.
Therefore, targeting cells with lower CHD4 expression with compounds targeting DNA/RNA
synthesis may have an impact on patient outcome.
P477: PHENOTYPICALLY-DEFINED STAGES OF LEUKEMIA ARREST PREDICT MAIN DRIVER MUTATIONS
SUBGROUPS, AND OUTCOME IN ACUTE MYELOID LEUKEMIA
F. Vergez1,*, L. Largeaud1, S. Bertoli2, M.-L. Nicolau-Travers1, J.-B. Rieu1, I. Vergnolle1,
E. Saland1, A. Sarry2, S. Tavitian2, F. Huguet2, M. Picard2, J.-P. Vial3, N. Lechevalier3,
A. Bidet3, P.-Y. Dumas4, A. Pigneux4, I. Luquet1, V. Mansat-De Mas1, E. Delabesse1,
M. Carroll5, G. Danet-Desnoyers5, J.-E. Sarry6, C. Recher2
1Laboratory of Hematology; 2Department of Hematology, CHU Toulouse, Toulouse; 3Laboratory
of Hematology; 4Department of Hematology, CHU Bordeaux, Bordeaux, France; 5Stem Cell
and Xenograft Core, UPENN, Philadelphia, United States of America; 6UMR1037, INSERM,
Toulouse, France
Background: Normal blood cell maturation is organized according to a functional hierarchy
at the top of which are multipotent hematopoietic stem cells (HSC). Immunophenotypically,
human HSC are enriched in a population of Lin-, CD34+, CD38-, CD90+, CD45RA- cells.
Upon differentiation, HSC give rise to multipotent progenitors (MPP), which retain
the ability to produce all blood lineages but have lost their self-renewal capacity.
The classical model of hematopoiesis postulates that MPP are then orientated toward
either the lymphoid or myeloid lineage, developing into common myeloid progenitors
(CMP) or lymphoid-primed multipotent progenitors (LMPP) which can still produce defined
myeloid cell types. Along the myeloid pathway, CMP can differentiate into either granulocyte-monocyte
progenitors (GMP) or megakaryocyte-erythroid progenitors (MEP). GMPs finally differentiate
into granulocyte progenitors (GP) or monocyte progenitors (MP).
Aims: In this study, we hypothesized that the stages of leukemic arrest (SLA) in myeloid
maturation can be defined by flow cytometry by analogy with the stages of hematopoiesis
and they correlate with the oncogenic events implicated in the mechanism of differentiation
block.
Methods: Six stages of leukemia differentiation-arrest categories based on CD34, CD117,
CD13, CD33, MPO and HLA-DR expression by flow cytometry were identified in 2 independent
fully annoted cohorts of 2087 and 1209 AML patients. Next-generation sequencing was
performed in 409 AML patients.
Results: HSC/MPP-like AMLs display low proliferation rate, inv(3) or RUNX1 mutations,
and high leukemic stem cell frequency as well as poor outcome, whereas GMP-like AMLs
have CEBPA mutations, RUNX1-RUNX1T1 or CBFB-MYH11 translocations, lower leukemic stem
cell frequency, higher chemosensitivity and better outcome. NPM1 mutations correlate
with most mature stages of leukemia arrest together with TET2 or IDH mutations in
GP-like AML or with DNMT3A mutations in MP-like AML.
Image:
Summary/Conclusion: AML immunophenotyping can establish a new SLA classification that
strongly correlates with cellular behaviour of the leukemic bulk, predicts main genetic
subgroups early at diagnosis and outcome after intensive chemotherapy. Each SLA is
defined by specific oncogenic events whose penetrance may be dependent on the differentiation
stages of hematopoiesis and their gene expression. Identifying disrupted gene pathways
specific for each SLA should therefore form the basis for targeted therapies aimed
at inducing AML differentiation.
P478: THE GERMLINE VARIANT GFI1-36N PROMOTES GENETIC INSTABILITY IN LEUKEMIC CELLS
AND OPENS NEW THERAPEUTIC APPROACHES FOR ABOUT 15% OF ALL AML PATIENTS
J. Vorwerk1,*, D. Frank1, K. Sun1, F. Neumannn2, M. Heuser3, D. Kunadt4, W. E. Berdel1,
J.-H. Mikesch1, C. Schliemann1, G. Lenz1, A. K. Jayavelu5,6,7,8,9, C. Khandanpour1
1Department of Medicine A, University Hospital Münster; 2Fluorescence Microscopy Facility
Münster, Institute of Medical Physics and Biophysics, University of Münster, Münster;
3Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover
Medical School, Hannover; 4Department of Internal Medicine I, University Hospital
Dresden, Dresden; 5Department of Proteomics and Signal Transduction, Max Planck Institute
of Biochemistry, Munich; 6Clinical Cooperation Unit Pediatric Leukemia, German Cancer
Research Center (DKFZ); 7Department of Pediatric Oncology, Hematology, and Immunology;
8Hopp Children’s Cancer Center Heidelberg (KiTZ); 9Molecular Medicine Partnership
Unit, European Molecular Biology Laboratory (EMBL), Heidelberg University, Heidelberg,
Germany
Background: The zinc finger protein Growth Factor Independence 1 (GFI1) acts as a
transcriptional repressor regulating differentiation of myeloid and lymphoid cells.
In a single-nucleotide polymorphism (SNP) of GFI1, GFI1-36N, the amino acid serine
(S) is replaced by asparagine (N) at position 36. GFI1-36N has a prevalence of 7%
in healthy Caucasians and 15% in acute myeloid leukemia (AML) patients, hence predisposing
to AML. We have shown previously that this is particularly since the GFI1-36N protein
differs from the GFI1-36S variant regarding its ability to induce epigenetic changes
leading to a derepression of oncogenes. Now we have evidence that GFI1 is implicated
in DNA repair as AML blast cells of patients carrying the GFI1-36N SNP feature more
frequently complex-aberrant karyotypes and more mutations leading to an inferior prognosis.
Aims: Therefore, in this work, we explored whether presence of GFI1-36N alters DNA
repair activity and whether this can be targeted therapeutically.
Methods: We analyzed the association between GFI1-36N and chromosomal aberrations
in human AML samples. As a murine model of human AML, we knocked in the human GFI1-36S
or GFI1-36N variant into the murine Gfi1 gene locus and retrovirally expressed MLL-AF9
to induce AML. We then performed γ-H2AX immunofluorescence and flow cytometry to quantify
the number of DNA double-strand breaks (DSBs), confocal immunofluorescence microscopy
to detect DNA repair foci of key enzymes of non-homologous end joining (NHEJ) and
homologous recombination (HR) as well as proteomics and immunoblotting to determine
the level of DNA damage response enzymes and cell cycle kinases. Cell viability assay
and colony-forming unit assay were used to examine the response of GFI1-36S and GFI1-36N
leukemic cells to palbociclib treatment.
Results: A complex aberrant karyotype was found twice as often in patients carrying
the GFI1-36N SNP compared to GFI1-36S controls (38% vs. 20%, p
** = 0.0080). Murine GFI1-36N-MLL-AF9 cells showed 43 times more genetic and cytogenetic
aberrations than GFI1-36S-MLL-AF9 controls (p
*** = 0.0003). Upon exposure to irradiation, murine GFI1-36N leukemic cells featured
more DSBs and delayed repair. Regarding the two main DSB repair pathways, we did not
see a significant difference in the ability of GFI1-36N or GFI1-36S leukemic cells
to perform NHEJ, but a reduced HR capacity in presence of GFI1-36N. On a molecular
level, GFI1-36N was associated with less foci of the HR key enzyme RAD51, reduced
protein levels of the DNA damage response enzymes CHK1 and CDK2, higher levels of
the cell cycle propagating CDKs, and reduced levels of the CDK inhibiting CDKNs. As
especially CDK4 and CDK6 protein levels were significantly increased in GFI1-36N leukemic
cells, we subsequently treated malignant cells with the established CDK4/6 inhibitor
palbociclib. We observed that GFI1-36N leukemic cells were more susceptible to this
treatment, indicated by 49% less CFUs compared to GFI1-36S leukemic controls (p
** = 0.0063).
Image:
Summary/Conclusion: The findings suggest that the GFI1-36N SNP increases cell cycling
and reduces DNA repair activity. This could promote emergence of chromosomal aberrations
and hence contribute to genomic instability of leukemic cells (Figure 1). On a therapeutic
level, GFI1-36N could possibly be a marker for a specific subset of AML patients that
is sensitive to CDK4/6 inhibitors.
P479: DNA METHYLATION PROFILING REFINES THE PROGNOSTIC CLASSIFICATION OF ACUTE MYELOID
LEUKEMIA PATIENTS TREATED WITH INTENSIVE CHEMOTHERAPY
S. Vosberg1,*, A. Ohnmacht2, C. Moser3, A. Arneth2, V. Jurinovic3, K. H. Metzeler4,
M. C. Sauerland5, D. Görlich5, W. E. Berdel6, J. Braess7, S. Amler8, U. Krug9, W.
Hiddemann3, K. Spiekermann3, C. C. Oakes10, M. P. Menden2, T. Herold3, P. A. Greif3
1Department of Internal Medicine, Medical University of Graz, Graz, Austria; 2Institute
of Computational Biology, Helmholtz Zentrum Munich, Neuherberg; 3Department of Medicine
III, University Hospital LMU Munich, Munich; 4Department of Hematology, Cellular Therapy,
and Hemostaseology, University of Leipzig, Leipzig; 5Institute of Biostatistics and
Clinical Research; 6Department of Medicine, Hematology and Oncology, University of
Münster, Münster; 7Department of Oncology and Hematology, Hospital Barmherzige Brüder,
Regensburg; 8Friedrich-Loeffler-Institut, Greifswald-Insel Riems; 9Department of Medicine
III, Hospital Leverkusen, Leverkusen, Germany; 10Department of Internal Medicine,
The Ohio State University, Columbus, United States of America
Background: Multiple studies have shown that DNA methylation is frequently altered
in acute myeloid leukemia (AML).
Aims: Here we propose a prognostic classifier for the survival of AML patients that
describes DNA Methylation-based Risk Assessment (“DMRA”), trained and validated using
a total of 538 adult AML patients from two independent cohorts, all treated with intensive
cytarabine-based chemotherapy with curative intent, and compared it to the current
ELN 2017 classification.
Methods: Patients from the first cohort were either treated within one of two consecutive
clinical trials of the German AMLCG study group or documented within the AMLCG registry.
DNA methylation profiles of 377 diagnostic AML samples were generated using Illumina
Infinium MethylationEPIC BeadChip arrays, selected based on their chemotherapy regimen
(i.e. 7 + 3, S-HAM, HAM(-HAM), or TAD-HAM). The clinical annotation includes overall
survival (OS) and the ELN risk assessment. Our DMRA classifier was trained on the
most variable CpG sites genome wide, weighted upon their relevance to OS based on
ridge regression. Three prognostic subgroups (“low”, “medium” and “high” risk) were
identified using cutoffs by optimizing the prediction error to avoid over- and underfitting.
We validated our approach by 10-fold cross-validation within AMLCG data, ensuring
that samples are either used for training or validation at the same time, as well
as using an independent cohort of diagnostic AML samples, selected for cytarabine-based
“standard chemotherapy” (n=161, BeatAML).
Results: Overall, the three risk groups defined by DMRA reveal more distinct survival
characteristics between each other as compared to between ELN risk groups (Figure
1A,B), with 3-year OS of 64% / 44% / 31% by ELN vs. 64% / 39% / 15% by DMRA. Of note,
nearly half of patients were assigned to a different risk group using DMRA as compared
to ELN (n=240/538, 45%, Figure 1C).
Importantly, we were able to define more precise risk groups within ELN subgroups,
with significant differences in OS. Within ELN favorable AML, 22% of patients (n=44/203)
were classified with increased risk (3-year OS 70% vs. 44%, p=0.003 “low” vs. “medium”,
log-rank test, Figure 1D). Within ELN intermediate AML, 33% of patients (n=44/135)
were classified as low risk AML (3-year OS 58% vs. 38%, p=0.03 “low” vs. “medium”,
Figure 1E). Within ELN adverse AML, 71% of patients (n=142/200) were classified with
reduced risk (n=27 “low risk”, n=115 “medium risk”, 3-year OS 38% vs. 37% vs. 13%,
p=0.0003 “low” vs. “high”, p=0.0002 “medium” vs. “high”, Figure 1F).
Image:
Summary/Conclusion: Based on this approach, we were able to distinguish clinically
relevant prognostic subgroups of AML patients within well described (cyto-) genetic
risk groups. Patients with favorable (cyto-) genetics but high-risk epigenetics might
benefit from alternative consolidation treatment strategies, such as allogeneic stem
cell transplantation. On the other hand, patients with adverse (cyto-) genetics but
low-risk epigenetics might be considered for consolidation chemotherapy instead of
stem cell transplantation in first complete remission. Finally, patients with both,
adverse (cyto-) genetics and high-risk epigenetics, must be considered as very poor
risk subgroup with very short OS (median 228 days).
Our results demonstrate that DNA methylation profiling is a feasible and robust approach
for risk stratification in AML, able to refine current risk assessment based on (cyto-)
genetics alone. We propose that DNA methylation-based risk assessment may serve as
a promising complement for current standards in prognostic classification of AML.
P480: NKG2D-MEDIATED ANTI-TUMOR IMMUNITY CONTRIBUTES TO THE FAVORABLE PROGNOSIS IN
APL
H. Wang1,*, X. Zhang1, S. Gong1, H. Du1, N. Mei1
1First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
Background: Acute myeloid leukemia (AML) is an aggressive hematological malignancy
with a poor prognosis. Acute promyelocytic leukemia (APL) is a unique subtype of AML,
with PML-RARA fusion caused by t(15;17)(q22;q12) translocation, and is now considered
a highly curable disease. Exploring the difference between APL and non-APL AML, may
shed new insight on the treatment of AML. NKG2D is an activating receptor on NK cells
that recognizes ligands on the surface of tumor cells and plays a crucial role in
tumor cell killing. Prior studies have demonstrated that one mechanism of AML immune
evasion is downregulation of NKG2D ligands (NKG2DL) or secretion of receptor-blocking
soluble ligands. Whereas, reports on NKG2DL expression in APL are still lacking. In
this study, we investigate the differences of NKG2DL expression on APL and non-APL
cells.
Aims: Exploring the difference between APL and non-APL AML, may shed new insight on
the treatment of AML.
Methods: Bone marrow samples were derived from de novo APL and non-APL AML patients,
diagnosed according to French-American-British (FAB) criteria in The First Affiliated
Hospital of Xi’an Jiao Tong University. Flow cytometry was performed with the anti-NKG2DL
mAbs (MIC-A/B-PE, ULBP-1-APC, ULBP-2/5/6-PerCP). Leukemic blasts were identified using
the CD45/SSC gating procedure.
Non-APL AML patients were divided into high or low group based on the median expression
levels of NKG2DL for survival analysis.
Results: Totally 46 patients comprising of 23 APL and 23 non-APL AML were enrolled
in this study. There was no significant difference in the baseline characteristics
between the two groups. All non‐APL AML cells had low or no NKG2DL surface expression,
while most APL cells expressed high NKG2DL levels. In particular, just two APL patients
was negative for the NKG2DL but positive for CD34. The median percentage of NKG2DL
among non-APL AML cells and APL cells were 1.2% vs 20.3% in MIC-A/B (P<0.01), 1.1%
vs 54.8% in ULBP-1 (P=0.017), and 3.8% vs 36.35% in ULBP-2/5/6 (P<0.01). (Fig.1)
23 non-APL AML patients were enrolled for study inclusion; one patient dropped out
due to cerebral infarction. The median follow-up time was 17.2 weeks. The high ULBP-2/5/6
group showed a superior overall survival rate compared to the low ULBP-2/5/6 group
(P < 0.01), but there was no significant difference between high and low MIC-A/B groups
(P = 0.31) or between the high and low ULBP-1 groups (P = 0.52).
Image:
Summary/Conclusion: NKG2DL was decreased significantly on the surface of non-APL AML
cells compared with APL cells. These results may indicate that NKG2D-mediated anti-tumor
immunity contributes to the favorable prognosis in APL, while immune escape is an
underlying mechanism for unfavorable prognosis in non-APL AML. NKG2DL may be used
as a potential biomarker to predict prognosis of patients with non-APL AML, and upregulation
of its expression could improve the clinical efficacy. Further studies with larger
sample size and longer follow-up are required to confirm our conclusion.
P481: MP0533, A NEW MULTISPECIFIC DARPIN CD3 ENGAGER TARGETING THREE TUMOR ASSOCIATED
ANTIGENS, INDUCES SPECIFIC T-CELL ACTIVATION AND AML TUMOR KILLING IN VIVO
A. Croset1,*, M. Bianchi1, M. Franchini1, T. Lekishvili1, Y. Kaufmann1, A. Auge2,
M. Hänggi1, W. Ali1, C. Zitt1, S. Wullschleger1, M. Matzner1, C. Reichen1, S. Fischer1,
Y. Grübler1, A. Eggenschwiler1, T. Looser1, R. Watson1, P. Spitzli1, V. Kirkin1, D.
Steiner1, A. Goubier1
1Molecular Partners AG, Schlieren, Switzerland; 2Molecular Partners AG, Molecular
Partners AG, Schlieren, Switzerland
Background: AML treatment options are generally focused on chemotherapy followed by
allogeneic hematopoietic stem cell transplantation. However, many patients are ineligible
for these options and often receive palliative treatments with poor long-term survival
and there is an urgent need for improved therapeutic solutions. Newer therapies, including
T cell engagers (TCE) and chimeric antigen receptor (CAR) T cells that target specific
molecules overexpressed on leukemic blasts and stem cells are promising alternative
treatment options for AML. However, achieving efficacy and low toxicity remains challenging.
MP0533 is a multi-specific, half-life extended, T cell engaging DARPin in development
for treatment of patients with AML and higher risk myelodysplastic syndrome (HR-MDS).
MP0533 consists of a chain of 6 covalently linked DARPin domains. Three of these domains
engage CD33, CD123 and CD70 on the target cells, utilizing an avidity driven approach
to ensure that binding occurs preferentially when 2 or more TAAs are present. A fourth
DARPin domain targets CD3 for T-cell engagement. Lastly 2 additional HSA binding DARPin
domains prolong half-life in circulation. In vitro, MP0533 has demonstrated, in both
allogeneic and autologous settings, single to double digit pM potency against AML
cell lines and AML primary cells expressing any combination of at least 2 of the 3
targeted TAAs and low cytokine release.
Aims: To evaluate and confirm that the previously established in vitro and ex vivo
efficacy and safety profiles of MP0533 are also observed in an in vivo human PBMC
xenograft mouse model.
Methods: NXG mice were injected intraperitoneally with hPBMC from healthy donors and
xenografted with MOLM-13 cells subcutaneously two days after hPBMC injection. Therapeutic
treatment was initiated eight days after tumor implantation once tumors were established.
DARPins were injected intravenously three times per week for two weeks. Several readouts
such as T cell activation and human immune cell infiltration in tumors were performed
by flow cytometry three days after first treatment. Additionally, cytokine and chemokine
release were analyzed in serum four hours after the first treatment injection and
in tumor supernatant 3 days after the first treatment injection.
Results: MP0533 induced AML tumor killing in vivo (P value <0.0001 at day 18 compared
to PBS). Three days after the first injection, MP0533 recruited human immune cells
in the tumor (around 10% of hCD45+ cells detected by FACS, P value = 0.0044 compared
to PBS) and induced T cell activation (around 46% of CD4+/CD25+/CD69+ cells and 38%
of CD8+/CD25+/CD69+ cells detected by flow cytometry, P value <0.0001 compared to
PBS) which triggered cytokines and chemokines release in the tumor such as IL-6, INFγ
and TNFα. However, MP0533 didn’t induce significant cytokine release, including at
the highest tested dose, in mice serum four hours after the first injection.
Summary/Conclusion: We were able to generate a multi-specific CD3 engaging DARPin
molecule with tailored affinities towards different TAAs showing significant efficacy
in vivo that induced T cell activation without significant cytokine/chemokine release.
P482: NOVEL BISPECIFIC INNATE CELL ENGAGER AFM28 FOR THE TREATMENT OF CD123 POSITIVE
ACUTE MYELOID LEUKEMIA AND MYELODYSPLASTIC SYNDROME
J.-J. Siegler1,*, N. Schmitt2, J. Pahl1, T. Haneke1, I. Kozlowska1, S. Sarlang1, A.
Beck2, S. Knackmuss1, P. Ravenstijn1, U. Reusch1, J. Medina-Echeverz1, J. Endell1,
T. Ross1, D. Nowak2, C. Merz1
1Affimed GmbH, Im Neuenheimer Feld 582, Heidelberg; 2Department of Hematology and
Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
Background: Novel treatments for patients with acute myeloid leukemia (AML) and high-risk
myelodysplastic syndrome (HR-MDS) with relapsed or refractory (R/R), or measurable
residual disease are urgently needed. Targeting of the surface antigen CD123 expressed
on both leukemic blasts and stem cells (LSCs) of most patients allows for depletion
of both compartments, which is considered a prerequisite for deep anti-tumor responses
and induction of prolonged remission. AFM28 is a novel bispecific Innate Cell Engager
(ICE®) designed for bivalent high-affinity binding of CD16A on natural killer (NK)
cells, redirecting effector cell cytotoxicity to CD123+ tumor cells. Allogeneic NK
cell therapy is emerging as a next-generation treatment with demonstrated activity
in R/R AML and a very benign safety profile. Retargeting of NK cells with AFM28 may
be a particularly effective treatment strategy in patients with CD123+ AML and HR-MDS.
Aims: 1. Preclinical characterization of AFM28 pharmacology and toxicology in vitro
and in vivo to support clinical development.
2. Assessment of binding and activation of NK cells by AFM28 to test suitability for
combination development with allogeneic NK cell therapy.
Methods: Flow cytometry (FC) was used to assess binding and retention of AFM28 on
NK cells, NK cell activation (degranulation, IFNγ expression) and depletion of leukemic
cells from AML and MDS patient specimens. Quantification of secreted cytokines in
leukocyte cultures employed bead-based luminex methodology. In vivo efficacy data
were acquired by BLI measurement from a murine AML model. Cynomolgus data were acquired
from non-GLP dose range finding and a GLP toxicology study using standard methods
(ELISA, IHC, FC).
Results: AFM28 induced NK cell activation at low picomolar concentrations and mediated
efficacious depletion of CD123+ tumor cell lines and primary leukemic cells, including
cells with low CD123 expression. NK cell activation was induced more potently than
by an effector-enhanced IgG1 and was accompanied by more pronounced degranulation
and IFNγ production. Similarly, AFM28 induced depletion of leukemic cells in patient
bone marrow samples, without lysis of CD34+/CD123− cells, suggesting sparing of healthy
hematopoietic progenitors. Potent anti-tumor activity was confirmed in vivo in a murine
tumor model. In preclinical toxicology models, AFM28-induced target cell depletion
was associated with minimal release of inflammatory cytokines, including IL-6, TNFα
and IFNγ from leukocytes in vitro, and only low-level release of IL-6 in cynomolgus
monkeys, despite efficacious depletion of CD123+ basophils and plasmacytoid dendritic
cells (pDCs). Characterization of NK cell binding revealed high-avidity interaction,
long cell surface retention, and potent induction of NK cell-mediated tumor cell lysis
with no impact on NK cell viability.
Summary/Conclusion: These data demonstrate that AFM28 exhibits potent pharmacological
activity in vitro and in vivo and efficaciously induces NK cell-mediated depletion
of primary leukemic cells. Further, toxicology data suggest good tolerability with
a low risk of cytokine release syndrome. Together, these findings support the clinical
investigation of AFM28 as a potential novel treatment of CD123+ AML and HR-MDS. A
first-in-human trial investigating safety and activity of single agent AFM28 in patients
with R/R AML is currently in preparation. In addition, these data support the combination
of AFM28 with allogeneic NK cell therapy, which holds potential as an effective, novel
treatment strategy.
P483: STAT3 DRIVES IMMUNE EVASION OF MLL-AF9-DRIVEN ACUTE MYELOID LEUKEMIA
B. Zdarsky1, A. Witalisz-Siepracka1,*, S. Boigenzahn1, K. Fiedler1, D. Stoiber1
1Department of Pharmacology, Physiology and Microbiology, Division Pharmacology, Karl
Landsteiner University of Health Sciences, Krems, Austria
Background: Acute myeloid leukemia (AML) is a heterogenous disease with poor prognosis.
Although the treatment opportunities in AML expanded in recent years, relapse still
remains a major issue and novel therapies aiming to eradicate minimal residual disease
are needed. Natural killer (NK) cell immunotherapy is put forward as a potential strategy
fulfilling these unmet clinical needs. Importantly, NK cell function of AML patients
is largely diminished due to different mechanisms of tumor-mediated immune suppression
and escape. Signal transducer and activator of transcription 3 (STAT3) is a member
of the JAK-STAT signaling pathway driving transcriptional responses downstream of
many cytokines. To escape the immune surveillance, tumor cells tend to downregulate
NK cell responses and STAT3 was proposed to drive this process in solid cancers.
Aims: We aim to investigate the function of STAT3 in AML evasion from NK cell surveillance.
Methods: Using CRISPR/Cas9 we generated STAT3-deficient human AML cell lines carrying
the MLL-AF9 oncogene. The cell lines were characterized via flow cytometry, Western
blot and qPCR and used as target cells for a human NK cell line in cytotoxicity assays.
Results: Using publicly available databases, we show that STAT3 expression negatively
correlates with several NK cell-activating ligands in AML patients. Moreover, when
studying a panel of human AML cell lines with a range of STAT3 expression levels,
we observed a trend for the most efficient NK-cell mediated killing in cell lines
expressing the lowest levels of STAT3. AML cells lacking STAT3 showed no major impairment
in survival and proliferation under homeostatic conditions but were less resistant
to different types of cellular stress. Interestingly, loss of STAT3 significantly
enhanced killing by NK cells. The effect was also supported by elevated expression
of NK-activating ligands in STAT3-deficient cells. We are currently testing if STAT3
inhibition recapitulates the observed effect.
Summary/Conclusion: Our in vitro data indicate that targeting STAT3 in AML might be
a strategy to enhance NK cell-mediated killing. We plan to extrapolate our findings
into in vivo xenograft systems with adoptive transplants of primary human NK cells.
The study will provide important insights into AML evasion from NK cell-mediated surveillance
and potentially give a novel rationale for STAT3 inhibition in AML.
P484: WHOLE EXOME SEQUENCING (WES) CHARACTERIZES THE MUTATIONAL LANDSCAPE OF BLASTIC
PLASMACYTOID DENDRITIC CELL NEOPLASM (BPDCN)
H. Witte1,*, A. Kuenstner2, J. Schwarting3, V. Bernard4, H. Merz4, N. von Bubnoff3,
H. Busch2, A. Feller4, N. Gebauer3
1Hematology and Oncology, German Armed Forces Hospital Ulm, Ulm; 2Medical Systems
Biology Group, University of Luebeck; 3Hematology and Oncology, UKSH Campus Lübeck;
4Haematopathology, Haematopathology Luebeck, Luebeck, Germany
Background: Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and clinically
aggressive hematologic malignancy. Due to novel technologies pathogenetic concepts
of BPDCN underlie a dynamic process. However, its genomic features including novel
therapeutic vulnerabilities remain sparsely characterized.
Aims: The present study aimed to illustrate the mutational landscape of BPDCN.
Methods: To delineate the mutational landscape of BPDCN, whole-exome sequencing (WES)
of 47 cases was performed. Significantly mutated genes, oncogenic drivers and perturbed
pathways were compared with data from acute myeloid leukemia (AML) and chronic myelomonocytic
leukemia (CMML).
Results: Median age was 73 years (range 26 – 91) and 72.3% were male. Primary involvement
of the skin and the bone marrow was present in 28 (59.6%) and 15 (34.9%) cases, respectively.
A median of 98 mutations per sample was detected resulting in a tumor mutational burden
(TMB) of 2.57 mutations/Mb/sample. WES and consecutive gene set enrichment analysis
revealed an accumulation of oncogenic mutations in epigenetic regulators of gene-expression
(95.7% of cases; TET2, DNMT3A, KMT2D, SETD2, IDH2), RTK-RAS (93.6%; NRAS, MET, EGFR),
NOTCH (76.6%; NOTCH2, CREBBP, EP300) and WNT signaling pathways (59.6%; CTNNB1, MED12).
Moreover, WES identified a subset of shared myeloid drivers across the spectrum of
BPDCN, AML and CMML. In the era of precision oncology, exome profiling revealed several
potential therapeutic targets such as NOTCH2, NRAS, EGFR or EZH2.
Summary/Conclusion: To the best of our knowledge, we provide genomic profiling in
the largest WES cohort in BPDCN to date. Current results confirmed previous genomic
features associated with myeloid pathogenesis, identified additional significantly
mutated drivers and novel potential therapeutic vulnerabilities.
P485: TARGETING HISTONE LYSINE-SPECIFIC DEMETHYLASES BY JIB-04 IMPROVES RESPONSE OF
AML CELLS TO VENETOCLAX TREATMENT
K. Wohlan1,*, D. Fan2,3, J. Su2,3, A. G. Guzman1, M. A. Goodell1,4,5,6,7, R. E. Rau8
1Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston,
United States of America; 2Institute of Biomedical Big Data; 3School of Biomedical
Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical
University, Wenzhou, China; 4Program in Developmental Biology; 5Stem Cells and Regenerative
Medicine Center, and Center for Cell and Gene Therapy; 6Interdepartmental Program
in Integrative Molecular and Biomedical Sciences; 7Department of Molecular and Human
Genetics, Baylor College of Medicine; 8Department of Pediatrics, Baylor College of
Medicine, Texas Children’s Hospital, Houston, United States of America
Background: Acute myeloid leukemia (AML) is a heterogeneous, hematologic malignancy
that is associated with a poor prognosis. Available treatment options are limited,
especially for older patients and others who are ineligible for intensive chemotherapy.
The BCL2-specific inhibitor venetoclax has proven efficacy in AML treatment particularly
in combination with hypomethylating agents (HMA) or low-dose cytarabine. However,
data from a clinical phase II study indicated a 30% failure rate after venetoclax+HMA
treatment of newly diagnosed patients (PMID: 30361262), suggesting additional options
are needed for patients who do not respond to available treatments or develop resistance.
Compared to normal bone marrow histone lysine-specific demethylases are more highly
expressed in AML, suggesting they are potential targets for therapeutic intervention.
JIB-04 is a potent small molecule inhibitor for multiple members of the KDM 4, 5 and
6 families with KDM5A being the most sensitive. Combined pharmacological inhibition
of KDM6 and KDM4 family members has been shown to reduce proliferation and increase
apoptosis of primary AML cells of various subtypes (PMID: 29111428). Knockdown of
KDM4 and KDM5 proteins leads to proliferation inhibition, cell cycle arrest, and apoptosis
in AML cells (PMID: 34017391; PMID: 29602065). Together, these data highlight that
KDM4, KDM5, and KDM6 proteins play an important role in the survival of AML cells.
Aims: Describe the efficacy of JIB-04 alone and in combination with venetoclax for
the treatment of AML. Identify the molecular changes due to JIB-04 in AML cells of
varying genetic subtypes.
Methods: We treated AML cell lines, patient AML blasts, and CD34+ cells from healthy
donors with JIB-04, venetoclax, or the combination for 72 hours. Apoptosis was quantified
by annexinV/propidium iodide staining. Cell proliferation of AML cell lines treated
with JIB-04 was evaluated using MTT assay and transcriptome profiling was done by
RNA-Seq.
Results: Treatment of AML cell lines with low nanomolar doses (IC50 range from 8-60nM)
of JIB-04 resulted in inhibition of proliferation and apoptosis. The combination of
JIB-04 with venetoclax showed a strong synergistic effect in all AML cell lines including
AML cell lines with relative resistance to venetoclax (Figure 1A). Moreover, patient
AML blasts also showed a synergistic response to the drug combination compared to
single-drug treatments (Figure 1B). In contrast, viability of normal CD34+ hematopoietic
cells was not affected by JIB-04 treatment at a dose of 100nM that leads to apoptosis
in AML cells, indicating a therapeutic window.
Changes in the transcriptome after JIB-04 treatment were analyzed in AML-OCI3 and
MOLM-13 cells. In both cell lines, genes related to the mTOR pathway and stress response
were downregulated, indicating possible mechanisms reducing proliferation and increasing
apoptosis. Further experiments will be performed to confirm the identified regulated
genes as druggable targets in AML that improve venetoclax response.
Image:
Summary/Conclusion: While JIB-04 alone shows some anti-leukemic effect, this study
highlights the potential therapeutic benefit of combining JIB-04 and venetoclax in
AML. This novel drug combination synergistically inhibits cell growth and induces
apoptosis in AML cells with various subtypes. In vivo studies are currently underway
to show the efficacy of this drug combination in patient derived xenograft models.
P486: CD9 MARKS DIFFERENTIATION AND ANTI-TUMOR IMMUNITY IN PEDIATRIC ACUTE MYELOID
LEUKEMIA
Y. Xu1,*, K. Y. Y. Chan1, S. P. Fok1, C. K. Li1, K. T. Leung1
1Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
Background: Despite intensive treatment regimes, the clinical outcomes for children
with acute myeloid leukemia (AML) remain suboptimal, underscoring the need to decipher
the underlying pathology and translate into therapeutic modalities. Emerging evidence
suggest myeloblasts could evolve multiple machineries to evade immune patrol and hinder
immunotherapies.
Aims: We recently reported the importance of CD9 in pediatric ALL but its role in
AML remains unknown. In this disease context, we pursued to characterize its prognostic
significance, elucidate its regulation and function, and identify its role in leukemia
immunosurveillance.
Methods: Patients were stratified based on CD9 status for comparison of long-term
survival. Epigenetic control of CD9 was investigated by ChIP-sequencing and confirmed
by HDACi treatment. Impact of CD9 on leukemia aggressiveness was measured by competition
and colony formation assays. Influence of CD9 on leukemia progression was evaluated
in xenograft models, coupled with global transcriptome profiling of AML samples. Immunomodulatory
effect of CD9 was dissected by single-cell transcriptomics and validated by flow cytometry.
Immune-related CD9 interactors were identified by IP-MS, followed by proof-of-function
experiments in an immune-reconstituted mouse model.
Results: The expression of CD9 on blasts of AML patients (12.2%, n=82) was significantly
lower than those of ALL patients (90.4%, n=219, P<0.001) or stem cells from normal
bone marrow donors (48.4%, n=22, P=0.014). Among AML cases, the blasts of 32 patients
(39%) were CD9+. The 5-year relapse-free survival rate of CD9- patients was significantly
lower than CD9+ patients (34.1% vs. 61.2%, P=0.018).
A marked decrease of H3K9/27Ac occupancy in the CD9 locus was observed in AML than
in ALL cells (4.8-14.2-fold, P<0.05), and strongly correlated with CD9 repression
(r=0.585-0.719, P<0.01). Exposure of CD9- AML cell lines (n=8) or samples (n=9) to
panobinostat significantly elevated CD9 mRNA and protein expression (3.1-32.2-fold,
P<0.05), and restored activating histone acetylation marks (4.1-41.6-fold, P<0.05).
Enforced CD9 expression in MV4-11 cells significantly suppressed proliferation (P<0.01)
and colony formation (P=0.002). NOD/SCID mice receiving CD9+ AML exhibited a drastic
reduction of leukemic load by 70.7-91.8%. Molecular expression analysis revealed decreased
stemness (NES: -1.7, P=0.01) and increased monocyte (NES: 1.8, P=0.034) gene signatures
in CD9+ patient samples. Concordantly, a profound up-regulation of CD9 (9.4-51.1-fold,
P<0.01) was observed in PMA-mediated monocyte/macrophage differentiation but not in
ATRA-mediated neutrophil differentiation of myeloblasts.
Mechanistically, CD9 promoted basal and IFNγ-induced MHC-I/II expression (P<0.01)
through the JAK2/STAT5 axis. Inter-patient comparisons of bone marrow samples (n=31)
revealed a higher MHC-I expression in CD9+ AML (P<0.001). Interestingly, CD9 physically
bound to MHC-I/II, formed an immune complex and regulated intracellular trafficking.
In NSG mice, co-transplantation of human PBMCs mounted an effective immunity against
CD9+ but not CD9- AML (MV4-11 and MOLM-13), concomitant with a robust bone marrow
infiltration of cytotoxic T cells.
Summary/Conclusion: Our data provided molecular, cellular and clinical evidence showing
the plausible function of CD9 as a key driver intertwining differentiation and immune
recognition in pediatric AML, and inspired a new combinatorial epigenetic/immunotherapy
for this rare but aggressive malignancy.
P487: SMALL NON-CODING RNAS ASSOCIATE WITH CHROMATIN TO MAINTEIN PROPOGATION OF ACUTE
MEYLOID LEUKEMIA
H. Yun1,2,*, F. Zhou1,2, C. Rohde1,2, J. Zoller1, X. Yu1,2, S. Göllner1, C. Müller-Tidow1,2,3
1Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University
Hospital; 2Molecular Medicine Partnership Unit, European Molecular Biology Laboratory
(EMBL); 3National Center for Tumor Diseases (NCT), Heidelberg, Germany
Background: Acute myeloid leukemia (AML) is a hematopoietic malignancy characterized
by the clonal expansion of myeloid progenitors without terminal differentiation. The
interplay between genome and epigenome alterations drives aberrant gene network linking
to leukemia transformation. Altered transcription may occur at three-dimensional chromatin
level and be orchestrated by a combination of multiple regulatory factors. Although
some RNA molecules, especially non-coding RNAs, were reported to modulate transcription
in leukemia cells, the regulatory role of genome-wide RNAs on transcription through
chromatin association remains largely unexplored.
Aims: To identify novel RNA molecules as chromatin-association factors critical for
leukemogenesis through modulating transcription
Methods: We performed in situ Mapping of RNA-Genome Interactome (iMARGI) in a human
acute leukemia cell line – MV4-11 cells. Chromatin association of candidate RNAs was
verified using Chromatin Isolation by RNA Purification (ChIRP) by means of biotinylated
oligonucleotides for target capture. A series of ChIP-seq assays were performed in
multiple AML cell lines to profile chromatin occupancy by Fibrillarin (FBL), a catalytic
component of C/D box ribonucleoproteins (snoRNPs), as well as to observe the remodelling
of chromatin states marked by H3K27ac and H3K9me3 upon shRNA-mediated knockdown of
FBL. Loss-of-function experiments using antisense oligonucleotides were performed
on candidate chromatin-associated RNAs in AML cell lines, AML primary samples, and
CD34+ healthy bone marrow cells.
Results: In total, about 52.1 million RNA-chromatin interactions were identified out
of deep sequencing of four iMARGI libraries. Whereas the vast majority (50.5 million,
97%) of RNA-chromatin interactome were intra-chromosome, a small fraction (1.6 million,
3%) occurred inter chromosome. Though protein-coding mRNAs predominate the chromatin-associated
RNA species, other non-coding RNAs were also captured. Intriguingly, different from
protein-coding mRNAs and long intergenic non-coding RNAs (lincRNAs) which mainly form
intra-chromosome interactions, two small non-coding RNA classes, small nucleolar RNAs
(snoRNAs) and small nuclear RNAs (snRNAs), were found with prevalent frequency (62%
and 92%, respectively) of inter-chromosome associations.
SNORD3A (U3) and SNORD118 (U8) were C/D box snoRNAs ranked top by the number of their
associated interactions in MV4-11 cells as well as in other cell types. Chromatin
association of U3 and U8 was verified using ChIRP, with the observation of unique
chromatin binding for both U3 and U8, and of specific binding motifs of transcription
factors at their binding sites. ChIP-seq revealed that the snoRNP FBL occupied only
a small set of genes (n=118), and shRNA-mediated knockdown of FBL had a minimal impact
on H3K27ac and H3K9me3, suggesting that snoRNA-chromatin interactions may function
independently on FBL-associated snoRNPs.
ASO-mediated loss of U3 or U8 significantly impairs leukemia cells proliferation and
colony forming capacity in multiple AML cell lines and primary AML blast samples,
whereas only mildly affects CD34+ healthy cells.
Summary/Conclusion: We identified chromatin-associated snoRNAs U3 and U8 which were
essential for leukemia maintenance and may serve as therapeutic targets for leukemia
eradication.
P488: THZ1, A COVALENT CDK7 INHIBITOR, INDUCES APOPTOSIS IN ACUTE MYELOID LEUKEMIA
CELLS AND EXERTS SYNERGISTIC ANTILEUKEMIA ACTIVITY WITH AZACITIDINE
S. Zhang1,*, H. Liu1
1Beijing Hospital, Beijing, China
Background: Acute myeloid leukemia (AML) is a malignant blood cancer that develops
mainly in elderly adults and has dismal clinical outcomes. Azacitidine (AZA) is still
the preferred treatment for elderly patients unfit for intensive chemotherapy. However,
the remission rate of azacitidine monotherapy is dismal, which necessitates the multidrug
combination therapy. THZ1, a CDK7 inhibitor, has been proven to be effective in various
cancers by regulating transcription and the cell cycle and inducing cell death. Here,
we investigated the antitumor effect of THZ1 monotherapy and its combination treatment
with AZA.
Aims: To investigate the antitumor activity of the CDK7 inhibitor - THZ1 in acute
myeloid leukemia (AML).
Methods: We first examined the cell viability of THZ1 against THP-1, MOLM-13, OCI-AML3
cell lines using CCK-8 assays. We used flow cytometry to detect apoptosis after cells
were stained with Annexin V-FITC/Propidium Iodide (PI), and western blot to detect
the expression levels of proteins. We detect cell cycle by flow cytometry after cells
were stained with 50 µg/ml PI and then analyzed with ModFit LT 5.0. RNA-sequencing
analysis was performed to explore the potential mechanism of THZ1 in AML cells. In
the double-drug combination experiment, we used CCK-8 assays to detect the effect
of drugs on the viability of the three cell lines, used CompuSyn software to calculate
the combination index (CI), which indicates additive effects (CI = 1.0), synergism
(CI < 1.0), and antagonism (CI > 1.0).
Results: THZ1 decreased viability and induced apoptosis in THP1, MOLM-13, and OCI-AML3
cells in a dose- and time-dependent manner. Besides, THZ1 inhibited phosphorylation
of Ser2, Ser5, and Ser7 residues of RNA Pol II CTD and induced cell cycle arrest at
G0/G1 phase in AML cells. RNA-sequencing analysis revealed that THZ1 induced changes
in the expression of genes involved in apoptosis, the cell cycle, and DNA repair.
Combined treatment with AZA and THZ1 showed a synergetic effect (CI < 1.0). Western
blot analysis of apoptosis markers, including cleaved caspase3 and PARP1, demonstrated
that THZ1 increased the expression of these markers and potentiated AZA-related apoptosis.
Compared with monotherapy, combined treatment with AZA and THZ1 resulted in more Annexin
V positive cells detected by flow cytometry, which further verified the synergistic
effect of the two drugs. Western blot results indicate that combined treatment with
AZA and THZ1 downregulates MCL1 at the protein level without an apparent decrease
in BCL2 protein expression.
Image:
Summary/Conclusion: Our data demonstrate that the CDK7 inhibitor THZ1 induces the
apoptosis of AML cells and exerts synergistic antileukemia activity with azacytidine,
which provide the rationale for combination treatment with AZA and THZ1 for AML.
P489: THE LONG NON-CODING RNA LINC01547 PROMOTES PROLIFERATION, CLONOGENICITY, AND
CELL CYCLE PROGRESSION IN ACUTE MYELOID LEUKEMIA
S. M. N. Zimmermann1,2,3,*, M. F. Blank1,2,4, D. Heid1,4,5, M. Bruckmann1, C. Müller-Tidow1,3,4,
J. Krijgsveld2,3
1Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital
Heidelberg; 2Division Proteomics of Stem Cells and Cancer, German Cancer Research
Center (DKFZ); 3University of Heidelberg Medical Faculty; 4Molecular Medicine Partnership
Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL);
5European Molecular Biology Laboratory EMBL, Heidelberg, Germany
Background: Non-coding sequences account for about 98% of the human genome, and it
has become increasingly apparent that non-protein coding transcripts drive numerous
physiological and pathological cellular processes. Long non-coding RNAs play important
roles in cancer, but functions in leukemogenesis and leukemic maintenance, as well
as underlying mechanisms, remain to be elucidated.
Aims: In this study, we characterize the functional and mechanistic implications of
the long non-coding RNA LINC01547 in acute myeloid leukemia (AML).
Methods: We generated single-cell derived, biallelic LINC01547 knock-out clones in
Kasumi-1 cells using the CRISPR/Cas9 system with paired sgRNAs. Knock-out and control
cells were assessed for differences in proliferation and clonogenicity by cell growth
and methylcellulose assays. Alterations in proliferation, cell cycle progression,
apoptosis, and global protein synthesis were subsequently evaluated in flow cytometry-based
assays. Finally, we characterized the impact of LINC01547 loss on RNA and protein
homeostasis by RNA sequencing, as well as nascent and steady-state proteomics.
Results: Bioinformatic analyses of expression data suggested a role for LINC01547
in AML. LINC01547 was highly expressed in AML (TCGA data), and its expression is associated
with worse overall survival in AML.
Biallelic deletion of LINC01547 suppressed AML cell growth and colony formation capacity.
Similarly, EdU-based proliferation assays showed significantly reduced proliferation
upon loss of LINC01547. Overall protein synthesis, assessed by O-propargyl-puromycin
(OPP) assays, and apoptosis, analyzed by annexin V assays, were not significantly
affected.
Transcriptome and steady-state proteome analyses revealed that depletion of LINC01547
impacted the expression of multiple mRNAs and proteins. An overall weak correlation
between differential expression on the transcript and the protein level indicated
that LINC01547 might mediate its effects through both, transcriptional and post-transcriptional
mechanisms. In line, mass-spectrometric analysis of nascent and steady-state proteomes
upon LINC01547 depletion showed numerous alterations on the translational and total
protein level, whereas the respective mRNAs often remained unaffected. Especially
proteins involved in cell cycle regulation exhibited reduced translation rates upon
loss of LINC01547. Consistently, cell cycle assays showed that depletion of LINC01547
slowed cell cycle progression and led to accumulation of cells in G0/G1 phase. Overlapping
our transcriptome, total proteome, and nascent proteome data sets, we identify potential
downstream targets through which LINC01547 presumably exerts its newly uncovered pro-oncogenic
function.
Summary/Conclusion: We demonstrate that the previously uncharacterized long non-coding
RNA LINC01547 has wide-ranging functions in AML. Its overexpression in AML and negative
correlation with overall survival are substantiated by its uncovered implications
in cell proliferation, clonogenicity, and cell cycle progression. In-depth characterization
and overlap of its impact on transcriptome, nascent proteome and total proteome reveal
numerous genes and pathways affected by LINC01547 depletion. Further assessment of
LINC01547 and its downstream targets will improve our understanding of the mechanisms
underlying leukemogenesis and potentially reveal novel leverage points in AML therapy.
P490: SNORNA-DERIVED RNAS (SDRNAS) ARE IMPORTANT DRIVERS OF LEUKEMOGENESIS AND LEUKEMIC
MAINTENANCE IN AML
R. Zinz1,2,*, C. Rohde1,3, D. Heid1,3,4, M. Bruckmann1, M. Bornhäuser5, C. Röllig5,
U. Platzbecker6, C. Baldus7, H. Serve8, F. Zhou1, C. Pabst1,3, C. Müller-Tidow1,2,3,
M. F. Blank1,3,9
1Medical Department V, Hematology, Oncology and Rheumatology, Heidelberg University
Hospital; 2University of Heidelberg Medical Faculty; 3Molecular Medicine Partnership
Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL);
4European Molecular Biology Laboratory (EMBL), Heidelberg; 5Medical Clinic and Policlinic
I, University Hospital Carl Gustav Carus and Medical Faculty of the TU Dresden, Dresden;
6Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University
Hospital, Leipzig; 7Medical Department II, Hematology/Oncology, University Medical
Center Schleswig-Holstein, Campus Kiel, Kiel; 8Department of Medicine II, Hematology/Oncology,
University Hospital Frankfurt, Goethe-University Frankfurt, Frankfurt; 9Division Proteomics
of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
Background: Small non-coding RNAs play important roles in leukemogenesis. The canonical
function of small nucleolar RNAs (snoRNAs) is to facilitate 2’-O-methylation of rRNA
via a target site-specific sequence and thus promote ribosomal biogenesis and function.
Recently, we have shown that a subset of small non-coding RNAs, i.e. C/D box snoRNAs
are overexpressed in acute myeloid leukemia (AML) and required for AML1/ETO-driven
leukemia initiation and maintenance. Depletion of snoRNP protein components or even
single snoRNAs can potently inhibit AML cell growth or decrease clonogenicity, and
the levels of many snoRNAs positively correlate with leukemic stem cell frequency
in AML patients. Over the past years, it has been shown that snoRNAs can be further
processed into shorter snoRNA-derived RNA fragments (sdRNAs), but the expression patterns
of sdRNAs in AML, as well as mechanistic and functional implications, remain widely
elusive.
Aims: In this study, we investigated sdRNA expression patterns, functions and mechanisms
in AML and healthy hematopoiesis.
Methods: We prospectively characterized snoRNA and sdRNA expression in 96 AML patient
samples, as well as in healthy hematopoietic stem and progenitor cells and differentiated
white blood cells by small RNA sequencing. mRNA sequencing was performed to investigate
connections between snoRNAs, sdRNAs and the transcriptome. Further, we depleted AML
cells of different snoRNA loci by CRISPR/Cas9-based knock-out and performed lentiviral
rescue overexpression of the respective sdRNA or full-length snoRNA.
Results: We discovered that more than 120 snoRNAs are further processed into sdRNA
fragments in AML. Single snoRNAs often evolve into multiple sdRNA isoforms. Interestingly,
sdRNA expression varied significantly between different AML patients. Correlation
of sdRNA and full-length snoRNA expression with patient characteristics, clinical
outcome and mutational status revealed numerous implications. Those include e.g. a
distinct pattern of enriched sdRNAs in NPM1-mutated patients. Expression of several
sdRNAs was associated with poor overall, event-free and relapse-free survival in AML.
We identified sdRNAs that were overexpressed in AML as compared to HSCs and found
characteristic expression patterns among differentiated white blood cells. Notably,
ratios of sdRNAs and their host snoRNA were not stable across different samples, suggesting
an active mechanism regulating sdRNA processing and thus their downstream pathways.
Gene set enrichment analyses based on mRNA and sdRNA correlations suggested distinct
cell type specific properties.
Forced expression of several sdRNAs rescued the reduced clonogenic potential observed
upon depletion of the respective host snoRNA locus. This effect was often identical
or even more pronounced than the rescue with the respective snoRNA precursor. Of note,
overexpression of distinct sdRNAs in AML cell lines enhanced clonogenic potential.
Summary/Conclusion: SnoRNA-derived sdRNAs are a common feature of AML with characteristic
expression patterns determined by mutation status and clinical features. Targeting
certain sdRNAs or factors involved in sdRNA processing or function might constitute
promising novel therapeutic leverage points in AML.
P491: PREDICTIVE FACTORS OF DIFFERENTIATION SYNDROME IN PATIENTS WITH ACUTE PROMYELOCYTIC
LEUKEMIA
M. Ben Salah1,*, M. Bchir1, R. Berred1, R. Kharrat1, L. Aissaoui1, Y. Ben Abdennebi1,
R. Ben Lakhal1, H. Ben neji1, B. Meddeb1
1Hematology department, Tunis El-Manar University, Faculty of medicine of Tunis, Aziza
Othmana Hospital, Tunis, Tunisia
Background: Differentiation syndrome (DS) may be a life-threatening complication in
patients with acute promyelocytic leukemia (APL) treated with all-trans retinoic acid
(ATRA) and anthracycline chemotherapy. Identifying high risk patients enables a more
efficient follow up and an optimal prophylaxis strategy.
Aims: The aim of this study was to analyze the incidence, characteristics, and the
risk factors of DS occurring during induction treatment in APL patients treated with
ATRA and anthracycline.
Methods: We conducted a retrospective single-center study, including patients diagnosed
with APL between 2010 and 2019 in our department and treated with ATRA and chemotherapy
according to PETHEMA protocols LPA99 and LPA2005. Obesity, renal failure and a high
risk (according to the Sanz score) represented an indication for a DS prophylaxis
prescription, using whether prednisone (LPA99) or dexamethasone (LPA2005). A univariate
and a multivariate analysis were conducted, in order to identify predictive factors
of DS.
Results: Ninety patients were included in our study, with an average age of 34 years
old. According to Frankel’s diagnostic criteria, 16 patients (18%) experienced a DS,
with a mean age of 39 years [13-71 years], and a sex ratio of 0.6. Half of this group
was classified as high risk according to the Sanz score, 44% as intermediate risk,
and 6% as low risk. A severe differentiation syndrome was described in 7 patients
(44%). The syndrome occurred at a median of 4 days after the start of induction therapy
[2-26 days]. A Prophylaxis had been prescribed for 10 patients (62.5%): 8 were classified
as high risk, and 2 had obesity.
In order to identify the risk factors of DS, the following parameters were analyzed:
age, sex, performance status, Body Mass Index, clinical presentation at diagnosis,
treatment protocol, Sanz score, creatinine level, LDH level, hemoglobin level, white
blood cell (WBC) count, platelets, cytological type, PML-RARa variant, CD2, CD15,
CD34, CD56 expression, fibrinogen level, prothrombin time, presence of disseminated
intravascular coagulopathy (DIC), occurrence of bleeding and thromboembolic complications,
and corticosteroid prophylaxis.
The univariate analysis identified a statistically significant difference between
the group of patients with DS and the one without, for the following factors: WBC
greater than 50G/L (31,3% versus 8,1%; p=0,02), the presence of DIC at diagnosis (81.3%
versus 18.7%; p=0.01), the expression of CD34 (p=0.017), the LDH level (a median of
307 versus 590; p=0.026), and the treatment protocol LPA 99(11 patients (68.8%) treated
according to LPA99 versus 5 (31,3%) treated according to LPA2005; p=0.003). Upon multivariate
analysis, WBC count greater than 50 G/L (p=0,007), the presence of DIC at diagnosis
(p=0,03), and the treatment protocol LPA99 (p=0,02), remained independent risk factors
of DS (Table1).
Image:
Summary/Conclusion: Differentiation syndrome is a significant complication secondary
to the ground-breaking treatment of APL. It is necessary to accurately identify high
risk patients in order to elaborate effective risk-adapted prophylaxis and treatment.
P492: TAMIBAROTENE IN COMBINATION WITH VENETOCLAX AND AZACITIDINE IN PREVIOUSLY UNTREATED
ADULT PATIENTS SELECTED FOR RARA-POSITIVE AML WHO ARE INELIGIBLE FOR STANDARD INDUCTION
THERAPY (SELECT AML-1)
E. Stein1,*, S. de Botton2, A. Pigneux3, C. McMahon4, B. Ball5, G. Borthakur6, A.
Eghtedar7, S. Kambhampati8, J. Tache9, E. Wang10, H. Kelley11, A. Volkert11, K. Baker11,
Q. Kang-Fortner11, G. Hodgson11, C. Madigan11, E. Warlick11, D. Roth11, M. Kelly11,
D. Pollyea4
1Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center,
New York, United States of America; 2Institut Gustave Roussy, Paris; 3CHU de Bordeaux
- Hôpital Haut-Lévèque, Bordeaux, France; 4University of Colorado, Aurora; 5City of
Hope, Duarte; 6Department of Leukemia, Division of Cancer Medicine, MD Anderson Cancer
Center, Houston; 7Colorado Blood Cancer Institute, Sarah Cannon Research Institution,
Denver; 8HCA Midwest Research Medical Center, Sarah Cannon Research Institution, Kansas
City; 9BRCR Global, Plantation; 10Roswell Park, Buffalo; 11Syros, Cambridge, United
States of America
Background: RARA-positive (RARA+) AML is a novel genomically defined patient subset
with an actionable biological target for treatment with tamibarotene, an oral and
selective RARα agonist (McKeown 2017). RARA+ patients can be selected by a blood-based
biomarker test, with approximately 30% of newly diagnosed (ND) AML patients being
RARA+ (Vigil 2017). As a biologically targeted agent for patients with RARA overexpression,
tamibarotene has the potential to provide benefit irrespective of mutation or cytogenetic
risk classification. In RARA+ ND AML patients ineligible for standard induction therapy,
tamibarotene plus azacitidine (aza) led to a CR/CRi rate of 61% and a rapid onset
of response (de Botton 2020).
Approximately one-third of patients with ND unfit AML do not respond to front-line
standard of care venetoclax (ven)/aza (DiNardo 2020). Translational data suggest RARA
positivity enriches for monocytic features reported to be associated with ven resistance
(Fiore 2020, Pei 2020). This data suggests the RARA biomarker selects for patients
who may respond to tamibarotene and may be less likely to respond to ven/aza. Given
that tamibarotene plus aza has been generally well tolerated, with no increase in
myelosuppression compared to single agent aza (de Botton 2020), it is anticipated
that tamibarotene can be administered safely in combination with ven/aza.
Aims: This is a Phase 2, open-label, multi-center study in the U.S. and France comparing
the clinical activity of tamibarotene/ven/aza to ven/aza in treatment-naive RARA+
AML patients ineligible for standard induction chemotherapy. The primary objectives
are to characterize the safety of the combination and to compare the CR/CRi rate of
tamibarotene/ven/aza vs. ven/aza, with secondary objectives to compare CR rate, CR/CRh
rate, duration of response, and time to response. The overall response rate using
tamibarotene/ven/aza following ven/aza treatment failure will be explored. Clinical
activity will be characterized by ELN criteria (Dohner 2017).
Methods: This 3-part trial includes a safety lead-in, randomized efficacy study, and
salvage arm. Following the safety lead-in, approximately 80 patients will be randomized
1:1 to receive tamibarotene/ven/aza or ven/aza. In the salvage arm, tamibarotene will
be added for study patients randomized to ven/aza who experience progressive disease,
relapse, or treatment failure. Patients will be treated with aza at 75 mg/m2 IV/SC
daily on days 1-7, ven on days 1-28 per VENCLEXA USPI, followed by tamibarotene at
6 mg twice per day by mouth on days 8-28 of each 28-day cycle.
Results: Response rates and 95% exact binomial confidence intervals will be calculated
by treatment group.
Summary/Conclusion: The SELECT AML-1 trial (NCT04905407) opened in July 2021 with
ongoing enrollment.
P493: SINGLE CENTER EXPERIENCE OF VENETOCLAX (VEN) IN COMBINATION WITH FLAG-IDA IN
PATIENTS (PTS) WITH NEWLY DIAGNOSED (ND) AND RELAPSED/REFRACTORY (R/R) ACUTE MYELOID
LEUKEMIA (AML)
Y. Abaza1,*, T. Khan1, S. Dinner1, O. Frankfurt1, J. K. Altman1
1Northwestern University, Chicago, United States of America
Background: Despite the high complete remission (CR) rates achieved with intensive
chemotherapy in AML, relapse rates remain high. This underscores the need for novel
regimens capable of inducing deeper remissions via eradicating measurable residual
disease (MRD). Recently, FLAG-IDA + VEN was shown to be an effective regimen in AML
inducing high rates of MRD negative CR in both ND-AML and R/R AML allowing patients
to be successfully bridged to allogenic stem cell transplantation (allo-SCT) (DiNardo
2021).
Aims: Assess the clinical activity of FLAG-IDA + VEN in pts with high-risk AML
Methods: We conducted a single-center retrospective study to assess the clinical activity
of FLAG-IDA + VEN in a high-risk pt population. Standard cytogenetic (CN) testing
was conducted and molecular data was obtained using a 40-gene next generation sequencing
platform. Responses were based on the modified International Working Group criteria
and included CR, CR with incomplete platelet recovery (CRp), morphologic leukemia-free
state (MLFS), and partial remission (PR).
Results: From March 2020 to January 2022, 17 pts with AML (10 ND and 7 R/R) were treated
using FLAG-IDA + VEN (Table 1). Median age was 48 years (range, 21-68) with male predominance
(76%). Among the 10 pts with ND-AML (5 de novo, 3 AML-MRC, 2 treated-secondary), 3
had extramedullary disease (EMD). The most common ELN cytogenetic risk groups were
adverse (N=2), complex (N=4), and KMT2A-rearranged (N=2). Five pts (50%) harbored
a RAS mutation. Median time to initiation of therapy was 5 days (range, 2-14). Five
pts [KMT2A-r: 2; complex CN: 2; diploid: 1] achieved CR/CRp including 1 pt with EMD.
Median time to count recovery (ANC ≥ 500 and platelets ≥ 50,000) was 21 days (range,
21- 42). Number of cycles of consolidation given to date were 1, 1, 1, 2, and 5 cycles,
respectively, and none of these pts have received allo-SCT yet. MRD status was available
for 3 pts of which 2 achieved MRD negativity using flow cytometry (<10-3) and one
remained MRD positive (NPM1 PCR) after induction. With median follow-up of 4.7 months
(range, 1.6-7.6), 4 pts remain alive in CR/CRp and 1 pt [complex CN, del 17p, TP53
mutant (VAF: 90%)] relapsed after 2 cycles of therapy with no response to salvage
decitabine (DAC) plus VEN and died due to progressive disease (PD). Of the remaining
5 nonresponders, 2 had treated-secondary AML, 2 with adverse CN [inv (3), -(7)], and
2 pts had EMD. All 5 pts died due to PD including 3 pts who had no response to salvage
therapy.
Among the 7 pts with R/R AML, 6 were in salvage 1 and 1 pt received prior allo-SCT.
Responses were observed in all patients: 4 CR, 1 CRp, 1 MLFS, and 1 PR. Median time
to count recovery in R/R AML pts was 33 days (range, 24-78). Of the 5 pts who achieved
CR/CRp, 4 were bridged to allo-SCT and 1 pt died in CR after 3 cycles of consolidation
due to sepsis. Of the 4 SCT recipients, 2 pts relapsed 2 months post-SCT (1 died due
to PD, 1 alive receiving salvage therapy) and 2 remain alive in CR for 4+ and 9+ months,
respectively. The 2 pts who achieved MLFS and PR died due to PD and fungal pneumonia
after 5 and 1.5 months of starting therapy, respectively. After median follow-up of
5.3 months (range, 0.7-13.2), median overall survival for the entire population was
6.2 months and the median duration of response for all pts who achieved CR/CRp was
not reached.
Image:
Summary/Conclusion: FLAG-IDA plus VEN is a feasible and active regimen in AML. Longer
follow-up and larger multicenter studies are needed to confirm the efficacy of this
regimen.
P494: MYELOID LEUKEMIA IN PATIENTS WITH INFLAMMATORY BOWEL DISEASES: A RETROSPECTIVE
COHORT STUDY
A. Abomhya1,*, A. Razzaq2, S. Lukose1
1Internal Medicine, The Brooklyn Hospital Center, Brooklyn, United States of America;
2St. George’s University School of Medicine, West Indies, Grenada
Background: Inflammatory bowel diseases include ulcerative colitis and Crohn’s disease
and affect around 3.1 million adults in the United States (US). The data on the risk
of myeloid leukemia among patients with IBD are limited and there has been only a
couple of studies that evaluated the incidence of myeloid leukemia in this patient
population.
Aims: This study aimed to estimate the prevalence of myeloid leukemia in patients
with IBD. Secondary outcomes included mortality, length of stay, all-cause 30-day
non-elective readmission rate, and total cost of hospitalization.
Methods: This is a retrospective cohort study for patients with IBD in the US. We
queried the Nationwide Readmission Databases 2016-2018 using ICD-10-CM codes to identify
all adult patients admitted for IBD. Patients with a comorbid diagnosis of myeloid
leukemia including acute myeloid leukemia (AML), chronic myeloid leukemia (CML), Myeloid
sarcoma, and Unspecified Myeloid leukemia were identified. Figure 1 summarizes the
case selection process. Median and IQR were used to describe Continuous variables,
and proportions were used with categorical variables. Comparison between groups was
performed by Mann Whitney test for continuous variables and Chi-Square test for Categorical
variables. Multivariate regression analysis was performed to study the impact of comorbid
myeloid leukemia on inpatient mortality and non-elective readmissions. Statistical
analyses were performed using SPSS Version 25 (IBM Corporation, Armonk, NY, USA).
Results: We extracted 365,152 index hospitalization records for IBD, 1052 (0.3%) had
myeloid leukemia. Six hundred sixteen patients had acute myeloid leukemia (AML), 341
patients had chronic myeloid leukemia, 10 patients had myeloid sarcoma and 104 had
unspecified myeloid leukemia. IBD patients with myeloid leukemia were older (64; Interquartile
Range (IQR): 52-73 vs. 56; IQR: 38-70, P <0.001), more common to be males (50.8% vs.
49.2%, P <0.001) compared to IBD patients without myeloid leukemia. Having myeloid
leukemia was associated with increased length of stays in days (7; Interquartile Range
(IQR): 3-23 vs. 3; IQR: 2-6, P <0.001), increased median total charges ($74,413; IQR:
$31,700 - $259,676 vs. $32,586; IQR: $17,827 - $62,260, P <0.001). On multivariate
analysis; having myeloid leukemia was associated with increased mortality (Odds ratio
(OR): 6.544; 95% confidence interval (CI): 5.318-8.052, P <0.001) and higher odds
of all-cause 30-day non-elective readmission (OR: 1.4; 95% CI: 1.155-1.697, P= 0.001).
Image:
Summary/Conclusion: In our nationwide cohort of IBD patients, 0.3% had myeloid leukemia.
Given the associated mortality and morbidity, we recommend considering a hematological
consult for IBD patients with leukocytosis or leukopenia for further evaluation and
appropriate management. More research studies are needed to investigate the pathogenesis
of myeloid leukemia in IBD patients.
P495: PHASE 2 STUDY OF ASTX727 (DECITABINE/CEDAZURIDINE) PLUS VENETOCLAX IN PATIENTS
WITH RELAPSED/REFRACTORY ACUTE MYELOID LEUKEMIA (AML) OR PREVIOUSLY UNTREATED, ELDERLY
PATIENTS UNFIT FOR CHEMOTHERAPY
T. Abuasab1,*, Y. Alvarado1, G. Issa1, R. Islam1, N. Short1, M. Yilmaz1, N. jain1,
L. Masarova1, S. Kornblau1, E. Jabbour1, N. Pemmaraju1, G. Montalban-Bravo1, S. Pierce1,
C. DiNardo1, T. Kadia1, N. Daver1, M. Konopleva1, G. Garcia-Manero1, F. Ravandi1
1Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston,
TX, USA, Houston, United States of America
Background: ASTX727, is an oral formulation of the fixed dose combination of decitabine
and cytidine deaminase inhibitor cedazuridine (35 mg/100 mg).
Aims: To investigate whether a total oral therapy regimen of ASTX727+venetoclax (ven)
is feasible and safe.
Methods: Pts aged ≥18 years (yrs) with relapsed/refractory AML (R/R) or pts with AML
aged ≥ 75 or 18 -74 with comorbid conditions prohibiting intensive chemotherapy were
eligible to participate (frontline-FL). Other eligibility criteria included adequate
renal and hepatic function and an ECOG performance status (PS) of≤2. ASTX727 is administered
daily on days 1‐5 of each treatment cycle and ven on days 1‐28 of the 1st cycle after
a dose ramp up of 100-200-400 mg over 3 days (with tumor lysis prophylaxis precautions
and with ven dose adjustments as needed. A bone marrow exam is performed on day 21±3
of 1st cycle and ven is held if blasts <5% to allow count recovery. Cycles are repeated
every 4-8 weeks depending on count recovery and Ven is administered for 21 days in
subsequent cycles, with dose adjustments as necessary depending on count recovery.
Results: Between March 2021 and January 2022, 28 pts (15 FL and 13 R/R) have been
treated on the study. The median age is 75 yrs (range, 47-90) with FL cohort 81 and
R/R cohort 72. 9 FL pts (60%) were ≥80 and 5 (30%) 70-80 years. In R/R cohort 9 pts
(69%) were 70-80 yrs. The median PS is 2 (range 0-3) and in the R/R cohort, the median
number of prior treatments is 2 (range, 1-4). In the FL cohort 5 (33%) had normal
and 6 (40%) a complex karyotype; 3 had other. In the R/R cohort, 15% had normal, 46%
complex karyotype and 31% other. Mutations of note in the FL cohort were RUNX1 (33%),
ASXL1 (33%), DNMT3A (7%), TET2 (40%) and TP53 (20%).
The overall response (ORR) including complete response (CR), CR with incomplete count
recovery (CRi) and morphological leukemia free state (MLFS) in the FL cohort is 61%
(4 CR, 4 CRi, 1 MLFS and 3 non-responders). 3 pts received only one day of therapy
for severe adverse events unrelated to therapy (1 due to ischemic stroke, 1 septic
shock and 1 debilitation) and were not evaluable for response. In the R/R cohort,
the ORR rate was 45% (2 CR, 2 CRi, 2 MLFS with 5 non-responders and 2 not evaluable).
The median number of cycles received is 2 (range, 1-5) for both cohorts. With a median
follow-up of 5 months, the median survival for the FL cohort has not been reached
(range, 0.6 – 7.3) and is 7.2 (range, 0.8-7.3) months for the R/R cohort. Grade 3
or higher adverse events directly attributable to therapy were mainly myelosuppression-related
and included neutropenic infections in 3 (11%) and elevation of liver enzymes in 1
(4%) pt.
Image:
Summary/Conclusion: The combination of ASTX727+ven is feasible, particularly in the
advanced elderly population, and demonstrates significant efficacy in pts unfit for
chemotherapy both in the FL and R/R settings.
P496: CLINICAL CHARACTERISTICS OF SECONDARY MYELOID NEOPLASMS IN PATIENTS WITH INFLAMMATORY
BOWEL DISEASE
T. Abuasab1,*, S. F. Mohadam1, H. Hwang2, X. Wang2, K. Sasaki1, M. Yilmaz1, T. Kadia1,
C. DiNardo1, N. Daver1, N. Pemmaraju1, G. Borthakur1, F. Ravandi1, G. Garcia-Manero1,
K. Takahashi1
1Department of Leukemia; 2Department of Biostatistics, The University of Texas, MD
Anderson Cancer Center, Houston, TX, USA, Houston, United States of America
Background: Inflammatory Bowel Disease (IBD) comprises two major disorders: ulcerative
colitis (UC) and Crohn’s disease (CD). Patients (pts) with IBD are at an increased
risk of cancer secondary to long-standing intestinal inflammation and secondary to
immunosuppressive therapies, including but not limited to colorectal cancer, hepatobiliary
tract cancer, Hodgkin, and non-Hodgkin lymphomas. However, very little data is available
regarding the risk of secondary myeloid neoplasm (MNs) in pts with IBD except for
the well-known association between azathioprine with therapy related MNs.
Aims: To describe the clinical characteristics of secondary MNs in patients with IBDs.
Methods: Retrospective chart review of patients with MNs who were previously treated
for IBDs was performed. A descriptive statistic was performed to define the demographics,
clinical and biological characteristics of the pts included in the study.
Results: Between 2012 and 2020, 43 pts were identified to have developed a secondary
MN during or after the treatment of IBDs. Sixty-three percent (27/43) were female,
and 37% (16/43) were male, with most of the pts being of white ethnicity (35/43, 81%).
Seventy percent (30/43) of the secondary MNs arose after the therapy for CD, whereas
30% (13/43) arose after UC therapy. The median age at the time of MN diagnosis was
59 years (yrs) (range, 23-83) and latency from IBD diagnosis was 16 years (range,
0-56). Twenty-five pts were on active treatment for IBD at the time of MN diagnosis
(9 on biological agent, 11 on mesalamine, and 2 on azathioprine), whereas 18 were
in remission. In addition, 8 pts (19%) had secondary cancer before the MN diagnosis
(2 pts lymphoma, 2 pts skin cancer, ovary Ca, uterine Ca, schwannoma, and adrenal
tumor) and 3 of them received therapy (chemotherapy and/or radiation therapy).
Seventy four percent (32/43) of MN diagnoses were Acute myeloid leukemia (AML), 9
(21%) pts had myelodysplastic syndrome (MDS), and one each for MDS/myeloproliferative
neoplasm (MPN) and chronic myelomonocytic (CMML). Seventy percent of the patients
had cytogenetic abnormalities:13 pts (30%) with complex karyotype and 6 pts (14%)
with core-binding factor (CBF) abnormalities (5 with inv16 and 1 with t [8;21]). The
most common somatic mutations included: TP53 (N = 10, 23%), FLT3 (N = 7, 16%), RAS
(N = 8, 19%), TET2 (N = 7, 16%), and DNMT3A mutations (N = 6, 14%). Interestingly,
all the pts with CBF-AML had prior history of CD, two of them treated with anti TNF-α
treatment.
The median overall survival (OS) was 2.17 yrs for the whole cohort, and there was
no difference in OS between MNs arose from CD and UC (2.35 and 2.08 yrs for CD and
UC, respectively). Seventeen patients underwent allogenic stem cell transplantation,
which resulted in remission of IBDs in 13 pts (76%).
Image:
Summary/Conclusion: We described the clinical characteristics of secondary MNs after
IBD therapy. Secondary MNs after IBD therapy included higher than expected prevalence
of core-binding factor AML, which was strongly associated with prior history of CD.
This observation raises an important question about the association between CD and
CBF-AML. Of interest, allogeneic stem cell transplantation led to remission of both
MNs and IBDs, highlighting the role of allo SCT in autoimmune diseases.
P497: PROGNOSTIC IMPACT OF RAS AND C-KIT MUTATIONS (SINGLE VS. MULTIPLE) IN CORE-BINDING
FACTOR ACUTE MYELOID LEUKEMIA TREATED WITH FLUDARABINE, CYTARABINE, G-CSF (FLAG) BASED
REGIMEN
T. Abuasab1,*, J. Senapati1, T. Kadia1, F. Ravandi1, C. DiNardo1, N. Pemmaraju1, M.
Ohanion1, Y. Alvarado1, H. Kantarjian1, G. Borthakur1
1Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston,
TX, USA, Houston, United States of America
Background: Core-binding factor acute myeloid leukemia (CBF-AML), including inv (16)
and t (8;21) AML, represent 15%-20% of all adult AML patients (pts) and is generally
recognized as favorable AML subgroups. Despite being favorable, up to 30-50% of patients
with CBF-AML eventually relapse. Both RAS (particularly multiclonal) and KIT mutations
in CBF-AML have been associated with poor outcomes, but our initial analysis for patients
treated with a fludarabine-based regimen did not support a negative impact of these
mutations. In addition, we showed that the optimal reduction of disease defining transcripts
(RUNX1-RUNX1T1 and CBFB-MYH11) by PCR at end of induction and end of consolidations,
has the most impact on the relapse free survival of these patients.
Aims: We studied the prognostic impact, in a consecutive cohort of CBF-AML patients
treated uniformly, of kinase mutations (RAS and KIT) and the number (Single vs. multiple)
of mutations in addition to the impact of mutation status on attainment of optimal
PCR reductions at most informative pre-defined time points; end of induction and end
of treatment.
Methods: Pts aged ≥ 18 years (yrs) with de novo CBF-AML, treated with FLAG based regimen
with idarubicin or gemtuzumab ozogamicin between April 2013 and December 2019 at our
center were included in this analysis. RAS and KIT mutations were tested by next generation
sequencing. PCR for disease-specific transcripts was monitored at the end of C1 and
end of therapy (EOT). Optimal response was considered as PCR transcripts < 0.1% post
C1 and <0.01% at EOT.
Results: A total of 91 pts with CBF AML were evaluated with a median age of 52 yrs
(range, 22-79), 41 of whom were females (45%). Fifty-three pts (58%) had Inv 16 and
the rest had t (8;21). Twenty-six pts (29%) had FLT3 mutation (ITD and/or TKD), 32
pts (35%) had RAS mutation (19 pts with NRAS, 3 pts with KRAS, and 10 pts with both
mutation), and 30 pts (33%) had KIT mutation. Out of 32 pts with RAS mutation, 16
(50%) had single mutation and16 (50%) had multiple RAS mutations, while of 30 pts
with KIT mutation, 25 pts (83%) had single KIT mutation, and 5 pts (17%) had multiple
KIT mutations.
On regression analysis of presence of KIT or RAS mutation and their impact on end
of induction and end of treatment PCR, only KIT mutation was associated with lower
odds (0.33, 95% CI 0.11-0.9) of optimal EOT response. Amongst mutated pts, the presence
of single versus multiple KIT or RAS mutation did not affect optimal PCR responses.
At a median follow up of 53.1 months (mos), the median relapse free survival (RFS)
for the whole cohort was 27.7 mos (range,1.7-93.6).
RAS mutated pts had longer RFS compared to non-mutated pts (not reached vs. 43.78
mos, p=0.02); however, KIT mutation did not affect RFS. In addition, the number of
the RAS or KIT mutation (One mutation vs. multiple mutations) also did not impact
RFS.
Image:
Summary/Conclusion: KIT mutation might negatively affect attainment of optimal PCR
responses at end of FLAG based therapy, but it lacked significant impact on relapse
free survival. Single versus multiple KIT or RAS mutations also do not seem to have
an impact on therapy response.
P498: CLINICAL AND BIOLOGICAL MARKERS ASSOCIATED WITH LONG-TERM SURVIVAL FOR PATIENTS
WITH ACUTE MYELOID LEUKEMIA (AML) IN REMISSION AFTER CHEMOTHERAPY IN THE QUAZAR AML-001
TRIAL OF ORAL AZACITIDINE
A. H. Wei1,*, H. Döhner2, H. Sayar3, F. Ravandi4, P. Montesinos5, H. Dombret6, D.
Selleslag7, K. Porkka8, J.-H. Jang9, B. Skikne10, C. Beach10, T. Prebet10, G. Zhang10,
A. Risueño11, M. Ugidos Guerrero11, W. L. See10, D. Menezes10, G. J. Roboz12
1Department of Clinical Haematology, Alfred Hospital, and the Australian Centre for
Blood Diseases, Monash University, Melbourne, Australia; 2Department of Internal Medicine
III, Ulm University Hospital, Ulm, Germany; 3Indiana University Cancer Center, Indianapolis;
4Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston,
United States of America; 5Hospital Universitario y Politécnico La Fe, Valencia, Spain;
6Hematology, Hôpital Saint-Louis, Assistance Publique – Hôpitaux de Paris (AP-HP),
and Institut de Recherche Saint-Louis, Université de Paris, Paris, France; 7AZ Sint-Jan
Brugge-Oostende AV, Bruges, Belgium; 8HUS Comprehensive Cancer Center, Hematology
Research Unit Helsinki and iCAN Digital Precision Cancer Center Medicine Flagship,
University of Helsinki, Helsinki, Finland; 9Samsung Medical Center, Sungkyunkwan University
School of Medicine, Seoul, South Korea; 10Bristol Myers Squibb, Princeton, United
States of America; 11BMS Center for Innovation and Translational Research Europe (CITRE,
a Bristol-Myers Squibb Company), Seville, Spain; 12Weill Cornell Medicine and New
York Presbyterian Hospital, New York, United States of America
Background: In the randomized, phase 3 QUAZAR AML-001 trial, oral azacitidine (Oral-AZA)
significantly prolonged overall survival (OS) compared with placebo (PBO) (median
OS 24.7 vs 14.8 months [mo], respectively) in older patients (pts) with AML in first
remission after intensive chemotherapy (IC). At an updated data cutoff performed in
Sep 2020, 34.9% of pts in the Oral-AZA arm and 24.4% of pts in the PBO arm remained
alive at ≥3 y from randomization.
Aims: Assess clinical and biological variables associated with long-term survival
(LTS) in QUAZAR AML-001.
Methods: In all, 472 pts were randomized 1:1 to receive Oral-AZA 300 mg or PBO QD
×14d/28d within 4 mo of achieving first complete remission (CR) or CR with incomplete
blood count recovery (CRi) after IC. The primary endpoint was OS, time from randomization
until death, withdrawal of consent, or loss to follow-up. The LTS cohort comprised
pts who were alive ≥3 y from randomization as of Sep 2020 and the non-LTS cohort included
pts who died or were censored before 3 y. Variables assessed for association with
LTS were diagnostic (Dx [pre-IC]) features (AML subtype, cytogenetic risk, NPM1 and
FLT3 mutations [mut]); pre-study treatment (Tx) variables (response to IC [CR/CRi],
receipt of consolidation, number of consolidation cycles); baseline (BL) demographic
and disease characteristics, hematologic parameters (red blood cells [RBCs], hemoglobin,
platelets, and leukocyte subsets), and measurable residual disease (MRD) status; and
post-BL variables (MRD response [conversion from MRD+ at BL to MRD– on study], timing
of MRD– [MRD– response on-study vs BL MRD–], and receipt of transplant after Tx discontinuation
[D/C]). Associations of LTS with bone marrow immune parameters (CD3, CD4, and CD8
T-cell counts, and expression of PD-1/TIM-3 T-cell exhaustion markers) were investigated
in a subset of pts (n=108). Variables were compared within Tx arms (LTS vs non-LTS)
in univariate analyses with P values corrected for multiple testing. A logistic multivariable
regression analysis of the effects of prognostic BL covariates on LTS was performed.
Results: The LTS cohort included 83/238 pts (34.9%) in the Oral-AZA arm and 57/234
pts (24.4%) in the PBO arm. Within both arms, factors significantly associated with
LTS were intermediate (Int)-risk cytogenetics and NPM1
mut at Dx, and MRD response (MRD+ to MRD–) on study (Figure). MRD response rate was
2-fold higher with Oral-AZA vs PBO (37% vs 19%, respectively), and while early attrition
was more common in the PBO arm, most MRD responses occurred within 6 mo. Factors significantly
associated with LTS only in the PBO arm were BL (post-IC) MRD– status and receipt
of transplant after Tx D/C. No significant associations were observed between BL hematological
or immune parameters and LTS.
The multivariable analysis (MVA) confirmed Oral-AZA Tx as independently significantly
predictive of LTS vs PBO. Other covariates significantly associated with LTS in MVA
were Int-risk cytogenetics and NPM1
mut at Dx, and MRD– status at BL.
Image:
Summary/Conclusion: Oral-AZA Tx was significantly associated with LTS vs PBO. In univariate
analysis, Int-risk cytogenetics and NPM1
mut at Dx, and MRD response on-study, were significantly prognostic of LTS in both
arms, whereas MRD– status at BL (post-IC) was associated with LTS only in the PBO
arm.
P499: THE PHASE 1B OMNIVERSE TRIAL OF ORAL AZACITIDINE IN COMBINATION WITH VENETOCLAX
FOR TREATMENT OF RELAPSED/REFRACTORY OR NEWLY DIAGNOSED ACUTE MYELOID LEUKEMIA (TRIAL
IN PROGRESS)
A. H. Wei1,*, H. E. Carraway2, L. Taningco3, E. Laille3, J. Gong3, T. Prebet3, D.
Lopes de Menezes3, F. Ravandi4
1Department of Clinical Haematology, Alfred Hospital, and the Australian Centre for
Blood Diseases, Monash University, Melbourne, Australia; 2Leukemia Program, Taussig
Cancer Institute, Cleveland Clinic, Cleveland; 3Bristol Myers Squibb, Princeton; 4Department
of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, United States
of America
Background: For older patients (pts) with acute myeloid leukemia (AML) who are ineligible
for standard intensive chemotherapy (IC) regimens, traditional lower-intensity AML
treatment (Tx) options include low-dose cytarabine (LDAC) or hypomethylating agents
(HMAs; azacitidine [AZA] and decitabine), which are generally well tolerated but associated
with suboptimal outcomes vs IC [Vey 2020]. Therefore, there remains a need for less
toxic and more efficacious lower-intensity AML Tx regimens, especially agents that
can be administered in the outpatient setting, which may increase convenience and
reduce resource utilization.
Venetoclax (VEN) is an oral, selective, small-molecule BCL2 inhibitor that has demonstrated
considerable activity when combined with LDAC or HMAs in older pts with AML. In a
recent phase 3 trial, VEN + injectable AZA significantly improved response and survival
vs AZA alone in IC-ineligible pts with newly diagnosed (ND) AML [DiNardo 2020]. In
the USA, VEN is approved in combination with an HMA or LDAC for Tx of pts with ND
AML ≥ 75 years (y) of age or who cannot use IC due to comorbidities.
Oral-AZA (CC-486) is approved for pts with AML in first remission (CR1) after IC who
are ineligible for transplant or other curative therapies. In the randomized, phase
3 QUAZAR AML-001 trial of Oral-AZA in pts with AML in CR1 after IC, Oral-AZA 300 mg
QD for 14 days (d)/28d Tx cycle was generally well tolerated and associated with significantly
improved overall survival vs placebo. Gastrointestinal symptoms were the most common
adverse events reported with Oral-AZA [Wei 2020]. Oral-AZA has also shown clinical
activity in pts with active AML [Savona 2015].
As incorporation of AZA into DNA is S-phase-restricted, extended Oral-AZA dosing regimens
(> 7d/28d Tx cycle) increase drug incorporation into cycling tumor cells to prolong
epigenetic activity throughout the Tx cycle [Laille 2015]. An all-oral VEN + Oral-AZA
combination regimen allows for outpatient administration and thus improves pt adherence
[Eek 2016].
Aims: Describe the study design and objectives of the OMNIVERSE trial of Oral-AZA
+ VEN in older pts with ND AML or pts with AML relapsed/refractory (R/R) to prior
Tx.
Methods: OMNIVERSE (NCT04887857) is a multicenter, open-label, 2-part phase 1b trial
(Figure). The key objectives of the trial are to assess safety and determine the maximum
tolerated dose of Oral-AZA + VEN in pts with R/R AML (WHO 2016 criteria) ineligible
to receive further IC (part I), and then in pts with ND AML ≥ 75 y of age, or those
≥ 18–74 y of age ineligible for IC or HSCT due to comorbidities (part 2). Main eligibility
criteria include an ECOG performance status of 0–2 (ECOG 3 is allowed for pts ≥ 18–74
y of age with comorbidities) and an unfavorable cytogenetic risk profile for pts with
ND AML. The initial dose of Oral-AZA is 300 mg QD × 14d/28d cycle, with de-escalation
to 200 mg × 14d/28d cycle allowed for dose-limiting toxicities. VEN 400 mg is taken
orally QD in continuous 28d cycles (or for 21d/cycle for dose level –2). A modified
toxicity probability interval-2 design is used to evaluate the planned dose levels.
The sample size is dependent on the dose levels included in the study (≤ 18 pts/part).
Results: N/A
Image:
Summary/Conclusion: Study enrollment began in 2021. The trial is ongoing at clinical
sites in the United States and Australia.
P500: A REAL WORLD MULTI CENTRE STUDY OF CPX-351 REVEALS NO DIFFERENCE IN OVERALL
SURVIVAL WHEN COMPARED WITH FLAG-IDA AND 3 + 7 IN HIGH RISK AML.
C. Andrews1,*, I. AlNabhani1, T. Young1, E. G. Atenafu1, S. E. Assouline2, J. M. Brandwein3,
S. M. Chan1, S. Chow4, D. Khalaf5, V. Gupta1, D. D. H. Kim6, D. Maze1, M. M. Minden1,
T. Murphy1, D. Sanford7, A. D. Schimmer1, A. C. Schuh1, J. Sibai1, K. Yee1, H. Sibai1
1Division of Medical Haematology and Oncology, Princess Margaret Cancer Centre, Toronto;
2Division of Hematology, Sir Mortimer B. Davis-Jewish General Hospital, Montreal;
3Division of Hematology, University of Alberta, Edmonton; 4Odette Cancer Centre, Sunnybrook
Health Science Centre, Toronto; 5Division of Hematology, Juravinski Cancer Centre,
McMaster University, Hamilton; 6Hans Messner Allogeneic Blood and Marrow Transplant
Program, Princess Margaret Cancer Centre, Toronto; 7Leukemia/Bone Marrow Transplant
Program of British Columbia, University of British Columbia, Vancouver, Canada
Background: CPX-351 is a liposomal formulation of daunorubicin and cytarabine in a
fixed synergistic ratio of 5:1. It has been approved by both the FDA and EMA for use
in high-risk AML including therapy related AML (t-AML) and AML with myelodysplastic
related change (AML-MRC). However, this is little data of CPX-351 in a real-world
setting. There is also no data comparing CPX-351 to FLAG-IDA which is used in our
institution front line for high-risk AML
Aims: The objective of our multi-centre study was to assess outcomes of CPX-351 and
then compare these outcomes in patients who received FLAG-IDA and 3 + 7 for high risk
AML(tAML and AML-MRC).
Methods: Patients aged 18 and over who were treated with induction chemotherapy and
who met the WHO criteria for t-AML and AML-MRC were included in the study. Retrospective
data was collected from 10 centres throughout Canada for CPX-351. The Princess Margaret
Cancer Centre database was used to collect data for patients who received induction
with FLAG-IDA and 3 + 7. Targeted sequencing was performed on DNA samples using the
TruSight Myeloid Sequencing Panel. Overall survival (OS) and progression free survival
(PFS) rates were calculated using the Kaplan-Meier method.
Results: 76 patients treated with CPX-351 were identified with baseline characteristics
seen on Table 1. Targeted sequencing was performed on 72% of patients (55/76) and
the average number of mutations was 2(0-7). RUNX1 was the most commonly mutated gene
found in 22% (12/55), followed by ASXL1 mutated in 19% (10/55) and SRSF2 in 15%. Assessing
treatment responses, 53% (38/76) of patients achieved a complete remission (CR) or
a complete remission with incomplete recovery (CRi). The median CR duration was short
at 7.3 months. Median follow up was 7.78 months (range 0.2 to 20 months). OS was 57%
at 12 months and 38% at 18 months. PFS was 40% at 12 months and 23% at 18 months.
There were no differences in OS when stratified by ELN risk (p=0.46), adverse risk
cytogenetics (p=0.1485), or poor risk mutations such as RUNX1 (p=0.73), ASXL1 (p=0.47)
or TP53(p=0.53).Patients who received an ASCT had significant improvement of OS of
82% at 18 months compared to those who did not receive a transplant of 15% at 18 months
(Fig 2; p=0.0001).
Patients who received 3 + 7 and FLAG-IDA for the same indications as CPX-351 (tAML
and AML-MRC) were identified. Baseline characteristics are shown in Table 1. 69% of
patients receiving FLAG-IDA and 67% of those receiving 3 + 7 had adverse ELN risk
stratification. CR rates were highest with FLAG IDA with 75% achieving a CR or CRi.
68% of patients who received CPX-351 proceeded to ASCT compared with 59% with FLAG-IDA
and 57% with 3 + 7. OS at 18 months was 42% for FLAG-IDA and 39% for 3 + 7. There
was no statistical difference in OS when comparing CPX-351 to FLAG-IDA and 3 + 7 (p=0.855)
(Figure 1). Similarly, there was no difference in PFS when comparing all 3 inductions
(p=0.26). Patients who received a ASCT had significant improvement of OS of 59% at
18 months compared to those who did not receive a transplant of 24% at 18 months (p=<0.0001)irrespective
of what induction was used.
Image:
Summary/Conclusion: High risk AML remains an unmet clinical need which is supported
by our study which reveals no difference in OS or PFS when treated with either CPX-351,
FLAG-IDA or 3 + 7. Unsurprisingly, ASCT improved OS across all patients irrespective
of induction type. Further study with a prospective randomised control trial is required.
P501: CLI120-001 PHASE1B DOSE ESCALATION STUDY OF RVU120 IN PATIENTS WITH AML OR HIGH
RISK MDS SAFETY AND EFFICACY DATA UPDATE
C. Abboud1, Z. Jan Maciej2, G. Borthakur3, S. Solomon4, B. Howard5, T. Bradley6, E.
Mouhayar7, N. Angelosanto8,*, H. Nogai8, A. Glasmacher9, R. Dudziak8, K. Brzozka10,
T. Rzymski10, P. Littlewood11, E.-L. Maranda12
1Division of Oncology, Washington University In Saint Louis, Saint Louis, United States
of America; 2Department of Hematology and Transplantology, Medical University of Gdansk,
Gdansk, Poland; 3Department of Leukemia, MD Anderson Cancer Center, Houston; 4Blood
and Marrow Transplant Leukemia and Immunotherapy Unit, Northside Hospital, Atlanta;
5Sarah Cannon Research Institute, Sarah Cannon, Nashville; 6Division of Hematology,
University of Miami Health System, Miami; 7Department of Cardiology, MD Anderson Cancer
Center, Houston, United States of America; 8CLINICAL DEVELOPMENT, Ryvu therapeutics,
Krakow, Poland; 9Dep. of Internal Medicine III, University Clinic Bonn, Bonn, Germany;
10Research & Preclinical Development; 11DMPK Department, Ryvu Therapeutics, Krakow;
12Deaprtment of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw,
Poland
Background: CDK8 and its paralog CDK19 have central roles in maintenance of cancer
cell viability and undifferentiated state for a variety of tumor types. (Dannappel
et al. 2019; Rzymski et al. 2015; Philip et al. 2018). RVU120 (SEL120), a novel CDK8/CDK19
kinase inhibitor with significant efficacy in preclinical AML models, has shown clinical
efficacy in a currently ongoing phase Ib trial in patients with relapsed/refractory
(R/R) AML or HR-MDS (NCT04021368). This paper provides update with new available data
on disease evaluation from ongoing patients and further enrolment into next cohort
level 85 mg.
Aims: The primary objective of the study is to determine preliminary safety profile,
dose limiting toxicities (DLTs), maximum tolerated dose (MTD) and the recommended
phase 2 dose of RVU120 as a single agent. Secondary objectives include PK, antileukemic
activity and exploratory PD characterization.
Methods: The study comprises at least 7 dose escalating cohorts. The first 3 cohorts
followed an accelerated scheme, 1 patient enrolled/cohort from 10 to 50 mg dose levels,
from cohort 4 (75 mg) to 7 (100 mg) doses onwards a 3 + 3 design is followed. Data
from each cohort is evaluated by a data review committee (DRC). RVU120 is administered
orally every other day, for a total of 7 doses, in a 3-week treatment cycle until
disease progression/unacceptable toxicity. Adverse events are graded according to
NCI-CTCAE v.5.0. DLTs are assessed at completion of C1. Disease evaluation is performed
according to Dohner 2017 and Cheson 2006 response criteria for AML and MDS respectively.
PK parameters are calculated by non-compartmental analysis. Pharmacodynamic (PD) activity
is assessed by flow cytometry measure of pSTAT5 Ser725 levels, that are highly dependent
on the activity of CDK8 and CDK19 in AML/MDS cells.
Results: At data cut off of 23rd Feb22, 13 pts have been enrolled, median age 73 years
and median 2 previous lines of therapy, ECOG PS 2 in 4 pts, 1 in 7, 0 in 2. No DLTs
were observed, all 14 Serious Adverse Events, including 1 COVID19 death and 1 pancreatitis,
were not related to study drug (G1 fever, G2 Upper Respiratory Infection, G3: pseudomonas
sepsis; urinary tract infection; febrile neutropenia; lung infection, pain, hemoptysis,
pleural effusion, G5 pneumonitis, death NOS, pancreatitis). Cohort 1 pt, 10 mg dose
level, and cohort 2 pt, 25 mg, showed stable (SD) and progressive disease (PD) respectively
at the end of C1. Cohort 3 pt, an 81 YO male HR-MDS, escalated from 50 to 75 mg dose
from C7, is SD at C24D13 with Erythroid Hematological Improvement on C5, C7, C10,
C18. Cohort 4, 75 mg dose pt, a 62 YO male with AML DNMT3A pos, relapsing after Ven/Dec,
achieved CRi at the end of C1 and CR in C7, and progressed at the end of C8. Two out
of the remaining 4 pts treated at 75 mg reached SD (1 still ongoing at C3D15 and another
died on C3D20 while on SD), 1 pt died of COVID-19 pneumonitis on C1D18, 1 pt with
AML secondary to MPN was SD at C2 and progressed on C4. Two pt were treated at 110 mg
(cohort 5), 1 not evaluable died for pancreatitis and 1 was SD at the end of C1. 2
pt entered cohort 6, 85 mg, and will be evaluable at the end of March 2022.
Summary/Conclusion: Preliminary results from the first 6 cohorts have shown a favorable
safety and a predictable PK profile of RVU120. Meaningful PD activity and clinical
efficacy were observed at 50 and 75 mg doses. Enrollment is currently ongoing at 85 mg
cohort
P502: CLINICAL IMPLEMENTATION OF GERMLINE GENETIC TESTING FOR HEMATOLOGIC DISORDERS
S. Ansar1,*, J. Malcolmson2,3, K. M. Farncombe4, K. Yee1, R. H. Kim1,5,6, H. Sibai1
1Division of Medical Oncology and Hematology; 2Familial Cancer Clinic, Princess Margaret
Cancer Centre, University Health Network; 3Department of Molecular Genetics, University
of Toronto; 4Toronto General Hospital Research Institute, University Health Network;
5Division of Clinical and Metabolic Genetics, The Hospital for Sick Children; 6Department
of Medicine, University of Toronto, Toronto, Canada
Background: Up to 18% of adult hematology patients (pts) suspected of having an inherited
predisposition are found to have a germline mutation leading to a hereditary hematologic
disorder (HHD). Timely identification of these disorders leads to heightened surveillance
for malignancies, specific treatment regimens, donor selection, transplant conditioning
regimens, cascade testing in family members and supportive care. Unlike solid tumour
hereditary cancer syndromes, the investigations in HHD are more complex and there
is no formal consensus of referral criteria or genetic testing criteria. We describe
our initial experience in this patient population at our centre, the Princess Margaret
Cancer Centre (PM). We have established a workflow to initiate germline genetic testing
in pts under suspicion for a HHD. This involves a collaboration with a genetics clinic
to 1) capture pts based on their personal/family history of cancer and suggestive
genetic findings identified on bone marrow/blood, 2) procure a skin biopsy for fibroblast
culture and germline DNA extraction and 3) provide follow-up counselling and management
for positive results.
Aims: Our goal is to evaluate the positivity rate of germline mutations in adult hematology
pts who were referred for genetics assessment.
Methods: The PM is the largest leukemia Centre in Canada, and has seen over 3000 adult
pts with a malignant disorder from 2015-2021 (AML:1597, MDS:472, ALL:286, MPN:664
and marrow failure:22). We performed a retrospective chart review of all adult hematology
pts referred to cancer genetics service for HHD workup during this time period.
Results: 116 pts (66 male, 50 female) were suspected for a HHD and referred for germline
genetic testing on fibroblast DNA. 52 pts (45%) were ≤40y old and 64 pts (55%) >40y
old (age range 18-86y, median 53y). 71 pts (61%) were referred with a positive family
history of hematologic disorders (HD). In addition, 61 pts presented with a myeloid
malignancy (46 with MDS, AML, or CML, and 15 with a MPN), 42 presented with a lymphoid
malignancy (ALL, CLL, or lymphoma), 8 presented with bone marrow failure, and 5 presented
with other HD. 40 pts were found to have a germline genetic mutation, 7 of which were
associated with carrier status. In total, 33 (28.4%) referred pts were found to have
at least one actionable germline mutation. This corresponds to a positive genetic
result found in 30% of pts referred with MDS/AML, 33% of pts referred with a lymphoid
malignancy, and 18% of pts referred with bone marrow failure, MPN, or other HD. Additionally,
18 (55%) of these pts had a positive family history of HD, while 15 (45%) presented
with no family history of HD. Of these 33 positive cases, 13 (11.2% of all referrals)
occurred in a gene associated with a hematologic malignancy and resulted in a HHD
diagnosis. 20 pts were found to have a mutation in a gene associated with a non-hematologic
hereditary cancer syndrome. After the implementation of a HHD genetics workflow in
2018, we saw an increase in the total number of referrals to a genetics clinic over
the past seven years, with 25 pts (6 positive cases) referred between 2015-2018, and
91 pts (27 positive cases) referred between 2019-2021.
Image:
Summary/Conclusion: Our overall positivity rate was 28.4% which includes mutations
in any hereditary cancer gene, and 11.2% for HHDs. Our high pickup rate across all
age groups and the increase in the number of referrals to genetics service suggests
that more pts with HD, including older pts, would benefit from consultation with specialized
centres that are experienced in the evaluation and treatment of these disorders.
P503: MRD MONITORING DURING INTENSIVE CHEMOTHERAPY IN PEDIATRIC AML- DATA OF THE AML-BFM
GROUP
E. Antoniou1,2,*, S. Hahn1,2, S. Sendker1,2, N. von Neuhoff1,2, D. Reinhardt1,2, M.
Schneider1,2
1Clinic of pediatrics III, University hospital of Essen; 2AML-BFM study group, Essen,
Germany
Background: During the past decades, minimal residual disease (MRD) has been established
as a diagnostic tool, providing critical prognostic information in pediatric acute
myeloid leukemia (AML). To measure initial treatment response, immunophenotyping was
used by most cooperative study groups. However, known limitation of sensitivity and
specificity supported the continuous evaluation of qPCR-based MRD detection in pediatric
AML.
Aims: To determine the prognostic role of MRD monitoring by RT-qPCR in the pediatric
AML.
Methods: A total of 238 pediatric AML patients were monitored for MRD by reverse-transcriptase
quantitative PCR (RT-qPCR) assay from September 2012 to December 2020. Since the inclusion
to this study is based on the presence of quantifiable genetic markers, the patient
group is strongly biased. The treatment was conducted according to the AML-BFM study
2004 and 2012 as well as the register 12 and 17. All measurements were performed in
bone marrow samples or in peripheral blood at initial diagnosis. MRD monitoring included
the following aberrations: t(8;21), inv(16), inv(11), NPM1, FLT3-ITD, t(15;17), t(5;11),
t(9;11), t(6;9), WT1, t(1;11), t(11;11), t(1;22), t(11;12), t(17;19), t(3;5), t(4;10),
t(11;19), t(2;11), t(6;11), t(X;11), t(6;8), t(8;16) and t(8;22).
Results: The patients had a mean age at diagnosis of 9.2 years (range 10 days to 18.6
years) and 51 % were females. They were distributed to the following stratification
groups: standard risk (56 %), intermediate risk (27 %) and high risk (17 %). The event
free survival was 71 % (±3 %) and the OS reached 90 %(±2 %).
In core binding factor acute myeloid leukemia (CBF-AML) (n=91) a positive MRD level
persists in the majority of cases. Following the 3rd treatment block 64% of cases
remained MRD-positive (MRD threshold ≥1x10-5). However, MRD monitoring showed no prognostic
relevance as relapses were detected in 18 % (6/33) and 17 % (10/58) of MRD-positive
and negative cases, respectively.
For patients with NPM1 mutations (n=26) MRD-positivity and -negativity (threshold
of ≥1x10-6) showed a similar relapse rate of 10 %. This data are in contrast to reports
in adults (Ivey et al. NEJM 2016).
In patients with an AML and t(9;11) MRD-positivity after first and second induction
indicated a significant higher risk of relapse. After first induction MRD-positive
cases (n=19) had a relapse rate of 32 % compared to 13 % for negative cases (n=24).
After second induction the relapse rates were 63 % and 22 % for MRD-positive (n=8)
and negative (n=27) cases, respectively.
Summary/Conclusion: MRD monitoring is a powerful tool to improve risk group stratification
in pediatric AML. Despite its high sensitivity and specificity, the prognostic informativeness
of MRD monitoring through RT-qPCR varies widely among different genetic subgroups.
In CBL-AML or NPM1-positive AML, which are prognostically more favorable, persisting
MRD-positivity alone was not associated with an increase in the risk of relapse. In
contrast, MRD-positivity in AML with t(9;11) appears to be highly informative.
Further studies with larger number of patients are needed to further define AML subgroups
and thresholds for MRD-positivity in the pediatric AML.
P504: CLINICAL MUTATIONAL ANALYSIS BY NEXT-GENERATION SEQUENCING AND REAL-LIFE VALIDATION
OF THE REVISED 2017 EUROPEAN LEUKEMIANET GENETIC RISK STRATIFICATION IN AML PATIENTS
C. Aparicio Pérez1,*, F. Salas Hernandez1, A. C. Gonzalez Teomiro1, F. Jimenez Najar1,
I. Fernandez Camacho1, C. Martin Calvo1, J. Sánchez García1,2, J. Serrano López1,2
1hematology, Reina Sofia University Hospital; 2IMIBIC, Córdoba, Spain
Background: Recently, a census of mutated genes in AML has been described but their
value in clinical practice is not fully elucidated. The revised 2017 European LeukemiaNet
(ELN) recommendations for genetic risk stratification of AML have been widely adopted,
but have not yet been validated in large cohorts of AML patients in real life.
Aims: The objective of this work is to analyze the mutations detected by Next Generation
Sequencing (NGS) and to study whether the application of ELN 2017 scale improves prognostic
risk stratification with respect to ELN 2010 and Medical Research Council (MRC).
Methods: We included 112 adult AML patients diagnosed in our center between Jun2017
and Dec2021 and who were studied at diagnostic using the spanish PLATFOLMA PETHEMA.
The prognostic risk was established according to MRC, ELN2010 and ELN2017 classification.
Baseline demographic, disease characteristics, treatment procedures and mutations
by functional groupsare summarized in Table 1
NPM1 and FLT3-ITD were determined by melting curve analysis and standard PCR-EC technique
according to Thiede et al (Blood 2002) in ABI 3130 Analyzer (Thermofisher). For NGS,
the commercial panel Myeloid SolutionTM (Sophia Genetics) KAPA Kit amplification libraries
and sequencing on ILUMINA Myseq platform were used. Variant analysis was performed
using DDM software (Sophia Genetics).
Results: The majority of the patients (97.1%) had at least one mutation at diagnosis
detected by NGS. The median number of mutations was 2.36 (0-6). Grouped by functional
groups, the most frequent were those related to DNA methylation (44.6 %) and signaling/kinase
pathways (37.5%). The most prevalent were FLT3 (29 %), IDH1/IDH2 (27%) and TP 53 (19.6%)
followed by NPM1 and RUNX-1 (17%).
Patients older than 60 years, presented higher percentage of unfavorable ASXL1 (18.6%),
RUNX1 (20%) and TP53 (25.7%) mutations with respect to younger (4.8%, 11.9% and 9.5
%) being these differences statistically significant in ASXL 1 and TP 53 (χ2 p=0.044
and p=0.047 respectively). NPM1 (28.6%) and FLT3 (40.5%) mutations were significantly
more frequent in younger patients (χ2 p=0.017 and p=0.013 respectively), than in >60
years (10% and 17.1%, respectively).
Re-stratifying according to ELN 2017, the 53% of patients categorized within the intermediate
risk group according to MRC change prognostic group: 28.9% became redefined as unfavorable
risk and 24.1% as favorable.
In the overall series the overall survival analysis shows statistically significant
differences taking into account either ELN 2017 or ELN 2010 and MRC (T.logrank p=0.003,
p=0.018 and p=0.012, respectively). However, according to ELN 2017 there are greater
differences between intermediate and unfavorable groups than in the other classifications.
Statistically significant differences in overall survival according to ELN 2017 (T.logrank
p=0.028) are also found in patients receiving intensive treatment. The estimated overall
survival at two years is 72.%, 52.1% and 46.8% in the favorable, intermediate and
unfavorable group, respectively.
Image:
Summary/Conclusion: - NGS proves its usefulness by detecting more clinically relevant
alterations than conventional cytogenetic techniques and PCR, and stratifies a larger
group of patients as favorable and unfavorable.
- Our results validate the prognostic significance of the ELN2017 classification in
real life, both in the overall series and in candidates for intensive QT.
- ELN 2017 establishes greater survival differences between the intermediate and unfavorable
group than ELN 2010 and MRC redefining the intermediate group.
P505: SAFETY AND EFFICACY OF CASEIN KINASE 1Α AND CYCLIN DEPENDENT KINASE 7/9 INHIBITION
IN PATIENTS WITH RELAPSED OR REFRACTORY AML: A PHASE 1, FIRST-IN-HUMAN STUDY OF BTX-A51
B. J. Ball1,*, G. Borthakur2, A. Stein1, K. W. Chan3, D. Thai3, E. M. Stein4
1Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical
Center, Duarte; 2Leukemia, MD Anderson Cancer Center, Houston; 3BioTheryx, San Diego;
4Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, United
States of America
Background: Inactivation of p53 and overexpression of Mcl1 are common mechanisms that
cancer cells use to evade apoptosis. BTX-A51 is a novel, oral, direct inhibitor of
casein kinase 1α (CK1α), cyclin dependent kinase 7 (CDK7), and CDK9. CDK7 and CDK9
phosphorylate RNA polymerase II (Pol II) to enable transcriptional initiation and
elongation, particularly at large clusters of transcriptional enhancers termed super-enhancers
(SE). Preclinical studies have demonstrated that BTX-A51 robustly increased p53 protein
levels via CK1α inhibition and Mdm2 downregulation while preferentially decreasing
SE transcription of key oncogenes such as Myc and Mcl1, enabling selective apoptosis
of leukemia cells. Here, we report the interim results of the phase 1, first-in-human
(FIH) study of BTX-A51 in patients (pts) with R/R AML.
Aims: 1. To evaluate the safety and preliminary efficacy of BTX-A51 in patients with
R/R AML
2. To determine the pharmacokinetics and pharmacodynamics of BTX-A51
Methods: This is an open-label, multi-center, FIH Phase 1 study. The study utilizes
a hybrid accelerated titration with single pt cohorts and a Bayesian optimal interval
design to assess 9 potential dosing cohorts. Key eligibility criteria include age
≥ 18 years, R/R AML or R/R high-risk MDS, ECOG ≤ 2, adequate kidney and liver function,
WBC ≤ 25K/uL. PD studies include digital droplet PCR from PB to quantify target genes
and measurement of macrophage inhibitory cytokine level by ELISA from serum at serial
timepoints.
Results: As of 25 January 2022, 30 pts (28 with AML; 2 with HR-MDS) enrolled at dose
levels between 1 and 42mg; 2 pts remain on treatment. Monotherapy doses between 1
and 42 mg were administered orally 3 days/week (wk) (3 wk in a 28-day cycle) and at
21 mg (4 wk in a 28-day cycle). Baseline characteristics include median age 75 years,
median number of prior therapies 3, 97% received prior treatment with venetoclax,
97% had prior HMA, and 43% had prior induction failure.
The most common treatment-emergent AEs (TEAEs) were nausea (60%), vomiting (52%),
hypokalemia (48%), diarrhea (36%), and hypotension (36%). The most common Grade 3
or higher TEAEs were anemia (28%), febrile neutropenia (28%), platelet count decreased
(24%), and hypokalemia (20%). The only DLTs were grade 3 hepatic failure at the 42 mg
dose and grade 3 alkaline phosphatase elevation at the 21 mg dose. All events resolved
after holding study drug.
Plasma PK of BTX-A51 was roughly dose-proportional between 1 and 42 mg with accumulation
based on AUC between Day 1 and Day 5. Estimated half-life was between 18 and 55 hours.
Among the 30 pts with R/R AML and MDS, CR/CRi rate was 10% (3/30) with 1 pt at the
11 mg and 2 pts at the 21 mg dose levels attaining CRi. Bone marrow (BM) blast reduction
>50% occurred in 4 patients including the 3 responders, all at the 11 and 21mg dose
levels. All 4 pts with >50% BM blast reduction had RUNX1 mutations; 9 pt with RUNX1
enrolled in the trial. The median duration of response for pts achieving CR/CRi was
approximately 1.5 month. Responses were not observed in MDS pts. PD data will be provided
in the full presentation. Based on the clinical data from dose escalation, the RP2D
is 21 mg administered 3 days/wk for 4 wk of a 28-day cycle.
Image:
Summary/Conclusion: In this FIH study, BTX-A51 demonstrated an acceptable safety profile
and promising antileukemic activity in pts with heavily pretreated R/R AML. The 21 mg
dose administered 3x/wk for 4 wk was identified as the RP2D. RUNX1 mutations were
enriched among responders and pts attaining >50% BM blast reduction.
P506: ACHIEVEMENT OF MEASURABLE RESIDUAL DISEASE CLEARANCE IS A STRONGER PREDICTOR
OF PATIENT OUTCOME THAN TREATMENT INTENSITY IN NEWLY DIAGNOSED PATIENTS WITH ACUTE
MYELOID LEUKEMIA
A. Bazinet1,*, T. Kadia1, N. Short1, G. Borthakur1, S. Wang2, S. Loghavi2, J. Jorgensen2,
K. Patel2, C. DiNardo1, N. Daver1, Y. Alvarado1, F. Haddad1, S. Pierce1, M. Andreeff1,
E. Jabbour1, M. Konopleva1, H. Kantarjian1, F. Ravandi1
1Leukemia; 2Hematopathology, University of Texas MD Anderson Cancer Center, Houston,
United States of America
Background: Modern therapy for acute myeloid leukemia (AML) can consist of either
intensive or low-intensity regimens selected based on patient (pt) age/comorbidities.
Measurable residual disease (MRD) has emerged as a strong independent prognostic factor
in AML. It is unknown whether pts who achieve similar levels of MRD clearance experience
comparable outcomes irrespective of treatment intensity.
Aims: To establish the relative prognostic contribution of treatment intensity and
MRD status on overall survival (OS) and relapse-free survival (RFS) in newly diagnosed
AML pts who have achieved a first response.
Methods: We conducted a retrospective chart review to identify non-CBF, non-APL AML
pts treated at our institution between 2010 and 2021 with intensive (IA; int/high-dose
cytarabine + anthracycline-based, without venetoclax) or low-intensity (Lo + VEN;
low-dose cytarabine/hypomethylating agent-based, with venetoclax) regimens who had
achieved CR/CRi/MLFS and undergone MRD testing by multiparameter flow cytometry at
time of first response. Differences in baseline pt characteristics were evaluated
using the Chi-square or Wilcoxon-Mann-Whitney tests for categorical and continuous
variables, respectively. The Kaplan-Meier method was used to estimate median OS and
RFS, with pts censored at the time of stem cell transplantation (SCT). Multivariate
analysis was performed using a Cox proportional hazards model.
Results: We identified 635 pts meeting inclusion criteria. Baseline characteristics
are shown in Table 1. Compared to the IA-treated pts (n=385), pts treated with Lo
+ VEN (n=250) were significantly older, more likely to be male, less likely to achieve
MRD clearance, and more likely to be classified as adverse risk by ELN 2017. The SCT
rate in the Lo + VEN cohort was half that of the IA cohort (25% vs 50%). Median OS
was 51m, 24.6m, 15m, and 9.9m in the IA MRD(-), Lo + VEN MRD(-), IA MRD(+), and Lo
+ VEN MRD(+) groups, respectively. Median RFS was 27.8m, 15.4m, 7.3m, and 5.2m in
the IA MRD(-), Lo + VEN MRD(-), IA MRD(+), and Lo + VEN MRD(+) groups, respectively.
Pts within the same MRD category (+ or -) had significantly higher OS and numerically
higher RFS if treated with IA versus Lo + VEN. When the analysis was restricted to
pts aged ≥ 60 years (n=56 in IA, n=239 in Lo + VEN; Fig. A-B), the differences within
MRD categories did not reach statistical significance (median OS NR in IA MRD(-) vs
24.6m in Lo + VEN MRD(-), p=0.08; median OS 13m in IA MRD(+) vs 10.6m in Lo + VEN
MRD(+), p=0.50; median RFS NR in IA MRD (-) vs 15.4m in Lo + VEN MRD(-), p=0.07; median
RFS 25.1m in IA MRD(+) vs 5.3m in Lo + VEN MRD(+), p=0.24). The SCT rate was 29% (87/295)
in the age ≥ 60 cohort. Given the confounding effect of unbalanced pt characteristics
between the IA and Lo + VEN cohorts, we performed a multivariate analysis on the full
population (n=635) taking into consideration the effects of age, treatment intensity,
MRD status, and ELN category on OS and RFS (all significant by univariate analysis).
In the multivariate analysis for OS, ELN adverse risk was the strongest predictor
(HR 2.09, p<0.001), followed by MRD(+) status (HR 1.68, p<0.001), while treatment
intensity and age were not significantly predictive. For RFS, ELN adverse risk (HR
2.31, p<0.001) and MRD(+) status (HR 1.98, p<0.001) were also the only two significant
predictors.
Image:
Summary/Conclusion: In newly diagnosed AML patients, MRD status at time of first response
and ELN risk are stronger predictors of pt outcome than intensity of therapy received.
P507: AGREEMENT BETWEEN REAL-WORLD PHYSICIAN RESPONSE ASSESSMENT AND THE 2017 EUROPEAN
LEUKEMIANET (ELN) CRITERIA IN ACUTE MYELOID LEUKEMIA (AML): A COMPARATIVE ANALYSIS
P. A. Patel1,*, F. Hoff1, A. J. Belli2, E. Hansen2, C. Anderson2, A. Barcellos2, L.
L. Fernandes2, H. Foss2, M. He2, M. Schulte2, C.-K. Wang2
1University of Texas Southwestern Medical Center, Dallas, TX; 2COTA, Inc., New York,
NY, United States of America
Background: Real-world data (RWD) is a valuable resource to understand the experience
of oncology patients treated outside a clinical trial. Increasingly, RWD is being
used in support of regulatory decision-making to evaluate outcomes compared to what
is observed in clinical trials. In the real-world setting, physicians often do not
utilize the specific inclusion/exclusion criteria and objective response criteria
that are implemented in clinical trials. Rather, a descriptive assessment of response
and clinical benefit is often used. Additionally, response assessment frequency may
not adhere to the requirements from clinical trials. Subsequently, it is critical
that real-world, physician-assessed responses are investigated to understand the concordance
with objective criteria in order to contextualize endpoints reported using RWD.
Aims: To evaluate the concordance and performance of physician-assessed response in
the COTA real-world database to derived response using the 2017 ELN criteria as the
established standard for response assessment in clinical trials.
Methods: A total of 879 patients meeting the following criteria were identified in
the COTA database: diagnosed with AML on or after April 1st, 2017, age ≥18 years at
diagnosis, received systemic treatment after diagnosis, and had at least 28 days of
follow up or a post-therapy bone marrow assessment. The COTA database is a USA-based
dataset composed of longitudinal, de-identified data on the diagnosis, clinical management,
and outcomes of patients with cancer. Physician-assessed response was manually captured
as documented in electronic health records (EHRs). Derived response was defined as
2017 ELN response based on bone marrow findings and complete blood count (CBC) results
available in the EHR. Rates of agreement and disagreement between physician-assessed
and derived response to first line treatment were calculated. Overall response rate
(ORR) was estimated by both response definitions.
Results: The study population (n=879) had a median age of 67 years and were predominately
white (78%), treated in the community setting (65%), and adverse risk per ELN criteria
(42%). Overall agreement between response categories was 65.1% with the highest agreement
among CR (61.7%). Agreement by response category is shown in Table 1. In a selected
group of patients with all required bone marrow and lab values to derive ELN response
(n=435), the ORR (95% confidence interval) was 80.5% (76.4, 84.1) for derived responders
and 79.1% (75, 82.8) for physician-assessed responders.
Image:
Summary/Conclusion: In our real-world cohort of patients with AML, agreement between
derived response per 2017 ELN criteria and physician-assessed response was 65.1%.
Among patients with derivable ELN response, the ORR calculations were similar by both
response definitions. These findings are significant in showing that responses assessed
by physicians treating patients with AML in the real-world setting are comparable
to those derived via 2017 ELN criteria. Of interest, discordance in derived response
vs. physician-assessed response was most common in the setting of CRi and MLFS. These
two response types require the incorporation of CBC results and bone marrow cellularity.
Future research will investigate the outcomes of patients with discordant response
assessments and incorporate additional response categories including CR with partial
hematologic recovery (CRh) and CR without minimal residual disease (CR MRD-).
P508: REAL LIFE EXPERIENCE USING FRONT-LINE CPX-351 FOR THERAPY-RELATED AND AML-MRC:
RESULTS FROM THE SPANISH PETHEMA REGISTRY.
T. Bernal1,*, G. Rad2, A. de Laiglesia3, C. Benavente4, A. Garcia Noblejas5, D. Garcia
Belmonte6, R. Riaza7, O. Salamero8, A. Foncillas9, A. Roldán10, V. Noriega Concepcion11,
J. Perez de Oteyza12, J. M. Bergua Burgues13, S. Lorente de Uña14, A. de la Fuente
Burguera15, M. J. Garcia Perez16, J. L. Lopez Lorenzo17, P. Martinez18, C. Alaez19,
M. Callejas20, C. Martinez Chamorro21, J. Rifon Roca22, L. Amador Barciela23, M. Lopez24,
K. Gomez Correcha25, E. Lavilla Rubiera26, M. L. Amigo27, F. Vall-Llovera28, A. Garrido29,
M. Garcia Fortes30, D. de Miguel Llorente31, A. Aules Leonardo32, C. Cervero33, R.
Coll Jorda34, M. Perez Encinas35, M. Polo Zarzuela4, D. Martinez Cuadron36, P. Montesinos36
1Hematology, Hospital Universitario Central Asturias, ISPA, IUOPA; 2Hematology, Hospital
Universitario Central Asturias, ISPA, Oviedo; 3Hematology, Hospital Puerta de Hierro;
4Hematology, Hospital Clinico San Carlos; 5Hematology, Hospital La Princesa; 6Hematology,
Hospital Universitario Sanitas La Zarzuela; 7Hematology, Hospital Universitario Severo
Ochoa, Madrid; 8Hematology, Hospital Vall d´Hebron, Barcelona; 9Hematology, Hospital
Infanta Leonor; 10Hematology, Hospital Infanta Sofia San Sebastian de los Reyes, Madrid;
11Hematology, Complejo Hospitalario de A Coruña, A Coruña; 12Hematology, Hospital
Madrid Norte San Chinarro, Madrid; 13Hematology, Hospital San Pedro Alcantara, Caceres;
14Hematology, Hospital Vithas Xanit Internacional, Malaga; 15Hematology, M.D. Anderson
Cancer Center, Madrid; 16Hematology, Complejo Hospitalario Torre Cardenas, Almeria;
17Hematology, Fundacion Jimenez Diaz; 18Hematology, Hospital Doce de Octubre; 19Hematology,
Hospital Universitario La Moncloa; 20Hematology, Hospital Universitario Principe de
Asturias; 21Hematology, Hospital Universitario Quiron Pozuelo, Madrid; 22Hematology,
Clinica Universitaria de Navarra, Pamplona; 23Hematology, Complejo Hospitalario Pontevedra,
Pontevedra; 24Hematology, Hospital General de Valencia, Valencia; 25Hematology, Hospital
Juan Ramon Jimenez, Huelva; 26Hematology, Hospital Lucus Augusti, Lugo; 27Hematology,
Hospital Universitario Morales Messeguer, Murcia; 28Hematology, Hospital Mutua Tarrasa;
29Hematology, Hospital de la santa Creu i San Pau, Barcelona; 30Hematology, Hospital
Universitario Virgen de la Victoria, Malaga; 31Hematology, Hospital Universitario
de Guadalajara, Guadalajara; 32Hematology, Hospital Miguel Servet, zZragoza; 33Hematology,
Hospital Virgen de la Luz, Cuenca; 34Hematology, ICO Girona, Hospital Universitario
Dr Josep Trueta, Girona; 35Hematology, Hospital Universitario Santiago de Compostela,
Santiago de Compostela; 36Hematology, Hospital Universitari i Politecnic La Fe, Valencia,
Spain
Background: Acute Myeloid Leukemias arising after cytotoxic therapy (t-AML) or with
myelodysplasia-related changes (AML-MRC) share adverse risk features and poor outcomes
after standard 3 + 7 chemotherapy. In a randomized clinical trial, CPX-351 has shown
superior overall survival (OS) compared to standard anthracycline-cytarabine schedule
in these AML subtypes.
Aims: to evaluate the effectiveness of CPX-351 treatment in a real-world setting.
Methods: adult t-AML and AML-MRC patients who have been treated upfront with CPX-351
in 35 Spanish centers between 2018 and 2021. All patients were included in the PETHEMA
registry (NCT02606825). Primary end-point was OS. Secondary end points were complete
remission with or without hematological recovery (CR/CRi), proportion of minimal residual
negativity (MRD), rate of allogeneic hematopoietic stem cell transplant (HSCT) and
safety.
Results: CPX-351 was administered to 74 patients as first induction chemotherapy.
Median age was 67 (63-71) years, with 40% (30/74) of female patients. ECOG performance
status score was 0-1 in 85% (53/62) patients. A diagnosis of t-AML was present in
30% (22/74) patients. Hypomethylating agents were used for MDS or CMML phase in 15%.
Hematopoietic Transplant Comorbidity Index was low, intermediate and high in 14% (10/74),
43% (32/74) and 43% (32/74) patients. ELN17 risk classification was favourable, intermediate,
adverse and indeterminate in 9% (7/74), 38% (28/74), 46% (34/74) and 5% (4/74).
CR/CRi was obtained in 43% (32/74) patients after first cycle with CPX-351. Three
additional patients achieved Morphological Leukemia Free Status. A second induction
with CPX-351 was administered in 5 patients with 100% CR (CR/CRi after 2 cycles 37/74,
50%). MRD was evaluated in 97% (36/37) of CR/CRi patients, being low (<0.1%) in 47%
(17/36) of them. In univariate analysis, no factor was associated with response. However,
71% (5/7) response rate was observed in the favourable ELN17 genetic risk group compared
to 48% (32/67) in the non-favourable.
HSCT was performed in 27% (20/74) patients, in 1st CR/CRi in 70% (14/20), MLFS in
15% (3/20) and active disease in 10% (2/20). With median follow up of 11.9 months
(7.2-23.6) from HSCT, median OS was not reached (95% CI 9.5-NR months (Figure). In
univariate analysis no factors were significantly associated with OS after HSCT.
Median time to absolute neutrophil count ≥0.5 x109/L and platelets ≥50 x109/L were
30.5 (25-35) and 30 (26-39) days, respectively. Deaths within the first 30 days after
induction occurred in 12% (9/74) patients. Causes of early death: infection in 5 patients,
haemorrhage in 3 and respiratory failure in 1. With 6.1 (1.8-14) months of median
follow up, median OS was 10.5 (6.9-NR) months, with significant differences between
HSCT and non-HSCT patients [NR, (95% CI: 12.7-NR), vs. 6 months, (95% CI: 4-NR), P<0.001].
In univariate analysis, factors associated with lower OS were non-favourable ELN17
[8.9 vs 23.5, P=0.04]; ECOG≥2 [0.7 vs 12., P=0.002]; age ≥65 [7.9 vs NR, P=0.007]
and male [6.6 vs NR, P= 0.007]. In multivariate analysis, age above 65 years [HR 1.6,
P<0.001], male [HR -0.9, P=0.02] and ECOG≥2 [HR 1.9, P<0.001] were associated with
OS.
Image:
Summary/Conclusion: Our real-life cohort was comparable to the target population of
the pivotal trial including patients diagnosed with sAML (t-AML, AML-MRC and ELN17
high risk AML), obtaining similar outcomes than the CPX-351 phase 3 trial arm. To
optimize treatment outcomes CPX-351 should be preferably offered to t-AML and AML-MRC
patients who are suitable for HSCT.
P509: CLONAL HEMATOPOIESIS-ASSOCIATED MUTATIONS AS MEASURABLE RESIDUAL DISEASE MARKERS
IN ACUTE MYELOID LEUKEMIA PATIENTS FOLLOWING ALLOGENEIC STEM CELL TRANSPLANTATION
L. Bischof1,*, J. Ussmann1, D. Brauer1, D. Backhaus1, L. Herrmann1, G.-N. Franke1,
V. Vucinic1, K. H. Metzeler1, U. Platzbecker1, S. Schwind1, M. Jentzsch1
1Hematology, Leipzig University Hospital, Leipzig, Germany
Background: Clonal hematopoiesis (CH)-associated mutations (mut) are frequent in acute
myeloid leukemia (AML), appear early in leukemogenesis, and often persist in remission
after chemotherapy. Following allogeneic hematopoietic stem cell transplantation (HSCT),
which replaces the patients’ hematopoiesis, CH mut may present useful measurable residual
disease (MRD) markers.
Aims: To evaluate patient specific CH-associated mut as MRD markers in AML patients
(pts) after allogeneic HSCT.
Methods: We analyzed 31 AML pts with CH-associated mut present at diagnosis (DNMT3A:
n=17; SRSF2: n=9; IDH2: n=7; ASXL1: n=4; TET2: n=2, and JAK2: n=1), 23 pts had 1,
7 had 2, and 1 patient had 3 CH mut. All received a myeloablative (36%), reduced-intensity
(48%), or non-myeloablative (16%) HSCT (median age 58, range 32-72 years). 84% were
transplanted in complete remission (CR) or CR with incomplete recovery (CRi). European
Leukemia Net (ELN) risk was 23% favorable, 37% intermediate, and 40% adverse. Mut-specific
digital droplet PCR assays were developed using a competitive probe-approach. MRDpos
was defined as variant allele frequency (VAF) ≥2% for ASXL1 & ≥0.05% for all other
analyzed mut. 209 samples with a median of 6 (range 1-13) blood or bone marrow samples
per patient were available after HSCT with a median of 1.2 years follow-up time for
pts alive.
Results: Prior to HSCT, 89% of pts remained CH-associated mut positive in CR/CRi.
11 pts relapsed after HSCT (35%), all with the same CH-associated mut present at diagnosis
(median VAF at relapse 15.6, range 0.2-41.2%). Of those, 6 pts had at least 1 MRD
sample available prior to relapse. In 5 pts, impeding relapse was predicted by a CH-associated
mut MRD conversion with a median VAF of 0.27 (range 0.06-1.0)% at a median of 103
(range 14-199) days prior to relapse. The lead time was longest for a DNMT3A mut patient
(199 days) & shortest for a SRSF2 mut patient (14 days). For the patient relapsing
without prior CH MRD positivity, the last available sample was taken 169 days before
relapse & was CH MRDneg. 20 pts retained remission during follow-up. Of those, 17
pts remained CH MRDneg in all samples (median samples per analyzed mutation 6, range
1-13). 2 pts had 2 MRDpos samples directly after HSCT but became MRDneg in subsequent
samples. 1 patient received Enasidenib maintenance after HSCT with fluctuating CH-MRD
values after HSCT (SRSF2 & IDH2, VAFs <0.05-0.34% & <0.05-0.14%, respectively).
Of the pts with MRD samples within the first year after HSCT available, pts with at
least 1 MRDpos sample after HSCT had a significantly higher cumulative incidence of
relapse (CIR, P=.008, Figure 1A) & shorter relapse-free survival (RFS, P=.02, Figure
1B). 8 pts had concurrent NPM1 mut (n=4) or RUNX1 mut (n=4), which showed concordant
MRD results in 7 pts (4 remained MRDneg, 3 concurrently converted MRDpos). 1 patient
suffered a NPM1 MRDneg relapse which was predicted by 2 re-detected CH-associated
mut (DNMT3A mut & IDH2 mut, 112 & 199 days prior to relapse, respectively, Figure
1C).
Image:
Summary/Conclusion: Prior to HSCT most CH-associated mut remain detectable in pts
in CR/CRi. However, pts with at least one CH MRDpos sample following allogeneic HSCT
had higher CIR & shorter RFS. All relapsing pts were positive for the same CH-associated
mut present at diagnosis. 5 of 6 showed increasing VAFs prior to hematologic relapse
& the majority of pts retaining remission remained CH MRDneg. While CH-associated
mut in chemotherapy treated AML pts have limited MRD value, these mut are feasible
MRD markers after HSCT & may inform relapse preventing therapy decisions.
P510: THE IMPACT OF POST-REMISSION GRANULOCYTE COLONY-STIMULATING FACTOR USE IN THE
PHASE 3 STUDIES OF VENETOCLAX COMBINATION TREATMENTS IN PATIENTS WITH NEWLY DIAGNOSED
ACUTE MYELOID LEUKEMIA
C. DiNardo1,*, K. Pratz2, P. Panayiotidis3, X. Wei4, V. Vorobyev5, Á. Illés6, I. Kim7,
V. Ivanov8, G. Ku9, C. L. Miller10, M. Zhang10, F. Tatsch10, J. Potluri10, X. Schmidt10,
C. Recher11
1Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston;
2Abramson Cancer Center, University of Pennsylvania, Philadelphia, United States of
America; 3Haematology Clinic and BMT Unit, NKUA, Laiko General Hospital, Athens, Greece;
4The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou,
China; 5Department of Hematology, S. P. Botkin City Clinical Hospital, Moscow, Russia;
6Department of Hematology, University of Debrecen, Faculty of Medicine, Debrecen,
Hungary; 7Seoul National University Hospital, Seoul, South Korea; 8Almazov National
Medical Research Centre, Saint Petersburg, Russia; 9Genentech, South San Francisco;
10AbbVie, Inc., North Chicago, United States of America; 11Centre Hospitalier Universitaire
de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université de
Toulouse 3 Paul Sabatier, Toulouse, France
Background: In the randomized Phase 3 VIALE-A (NCT02993523) and VIALE-C (NCT03069352)
trials, venetoclax (Ven) + azacitidine (Aza) and Ven + low-dose cytarabine (LDAC),
respectively, improved outcomes vs low intensity chemotherapy in patients (pts) with
newly diagnosed acute myeloid leukemia (AML) ineligible for intensive chemotherapy
(IC). Evidence is limited on the use of granulocyte colony-stimulating factor (G-CSF)
and impact on outcomes in this pt population.
Aims: To explore outcomes in pts with post-remission G-CSF use in the VIALE-A and
VIALE-C studies.
Methods: In VIALE-A, pts received Ven 400 mg or placebo (PBO) daily plus Aza 75 mg/m2
on d 1−7 (28-d cycles). In VIALE-C, pts received Ven 600 mg or PBO daily plus LDAC
20 mg/m2 on d 1−10 (28-d cycles). G-CSF use was given per institutional practice.
Here, pts who achieved a best response of complete remission (CR) or CR with incomplete
hematologic recovery (CRi) were analyzed by post-remission G-CSF use.
Results: In VIALE-A, 190/286 pts (66%) treated with Ven+Aza and 41/145 (28%) pts treated
with PBO+Aza achieved CR/CRi; 93 (49%) and 10 (24%) pts with CR/CRi received G-CSF
post-remission (median time to first post-remission G-CSF use [range], 36 d [2−483]
and 35 d [4−127]), respectively. In VIALE-C, 69/143 pts (48%) treated with Ven+LDAC
and 9/68 (13%) pts treated with PBO+LDAC achieved CR/CRi; 30 (43%) and 2 (22%) received
G-CSF post-remission (median time to first post-remission G-CSF use [range], 30 d
[2−459] and 229 d [169−289]), respectively. In VIALE-A and VIALE-C, baseline demographics
were generally similar in Ven-treated pts regardless of post-remission G-CSF use (Table).
In VIALE-A, median duration of response (mDOR) for CR/CRi (95% CI) was not reached
(NR; 17.5−NR) and was 12.9 mo (7.9−17.3) in the Ven+Aza+G-CSF and Ven+Aza+non-G-CSF
groups, respectively (Table); DOR at 12 mo was 67% and 53%. In VIALE-C, mDOR was 10.8
(4.9−17.8) and 11.8 mo (5.9−NR) in the Ven+LDAC+G-CSF and Ven+LDAC+non-G-CSF groups,
respectively; DOR at 12 mo was 45% and 49%. In VIALE-A, median overall survival (mOS
[95% CI]) was NR (NR−NR) with Ven+Aza+G-CSF and was 21.1 mo (15.2−NR) with Ven+Aza+non-G-CSF;
12-mo OS rates were 83% and 71%. In VIALE-C, mOS was 20.8 (11.9−NR) with Ven+LDAC+G-CSF
and 13.7 mo (10.8−NR) with Ven+LDAC+non-G-CSF; 12-mo OS estimates were 68% and 57%.
Among 164 evaluable pts in VIALE-A, measurable residual disease response (MRD<10−3)
was achieved by 67, of whom 38 (57%) had post-remission G-CSF. In VIALE-C, MRD<10−3
was achieved by 9 of 64 evaluable pts, of whom 6 (67%) had post-remission G-CSF.
In VIALE-A, post-remission Gr ≥3 neutropenia and febrile neutropenia (FN) rates were
33% (n=31) and 39% (n=36) with Ven+Aza+G-CSF, and 29% (n=28) and 20% (n=19) with Ven+Aza+non-G-CSF,
respectively. Median durations of post-remission Gr ≥3 neutropenia and FN in VIALE-A
were 12.5 d and 8 d (Ven+Aza+G-CSF), and 16 d and 10.5 d (Ven+Aza+non-G-CSF). In VIALE-C,
post-remission Gr ≥3 neutropenia and FN rates were 53% (n=16) and 23% (n=7) with Ven+LDAC+G-CSF,
and 51% (n=20) and 8% (n=3) with Ven+LDAC+non-G-CSF, respectively. Median durations
of post-remission Gr ≥3 neutropenia and FN in VIALE-C were 15 d and 6 d (Ven+LDAC+G-CSF),
and 12.5 d and 29 d (Ven+LDAC+non-G-CSF).
Image:
Summary/Conclusion: In VIALE-A and VIALE-C, G-CSF was frequently used per institutional
practice post-remission in responding patients to manage neutropenia. Overall, there
was a trend towards shorter durations of Gr ≥3 neutropenia and FN with post-remission
G-CSF use, without evidence of negative impact on DOR or OS.
P511: RISK FACTORS AND INCIDENCE OF CARDIAC EVENTS IN A LARGE COHORT OF 525 ADULT
PATIENTS WITH NEWLY DIAGNOSED NON-M3 ACUTE MYELOID LEUKEMIA
B. Boluda1,2,*, A. Solana-Altabella2,3, I. Cano1,2, D. Martínez-Cuadrón1,2, E. Acuña-Cruz1,2,
L. Torres-Miñana1,2, R. Rodríguez-Veiga1,2, D. Martínez-Campuzano1, R. García-Ruiz1,
P. Lloret1, P. Asensi1, A. Serrano1, A. Osa-Sáez4, J. Agüero-Ramón-Llín4, M. Rodríguez-Serrano4,
F. Buendía-Fuentes4, J. E. Megías-Vericat3, B. Martín-Herreros1,2, E. Barragán1, C.
Sargas1,2, M. Salas5, M. Wooddell5, C. Dharmani5, M. A. Sanz1,2, J. De La Rubia1,
P. Montesinos1
1Hematology, Hematology Department, Hospital Universitari i Politècnic La Fe, València,
Spain; 2Instituto de Investigación Sanitaria La Fe; 3Pharmacy; 4Cardiology, Hospital
Universitari i Politècnic La Fe, Valencia, Spain; 5Daiichi Sankyo, Inc., Basking Ridge,
United States of America
Background: The incidence of cardiac morbidity and mortality in acute myeloid leukemia
(AML) population is not well known. AML patients (pts) receive potentially cardiotoxic
drugs such as anthracyclines, QTc prolonging drugs such as FLT3 inhibitors, azoles,
among others. Cardiac events can emerge in pts with previous cardiac diseases, or
due to the toxicity of chemotherapy and other concomitant medications. So far, no
previous studies have provided a holistic view of cardiac issues in a real-world series
of AML pts.
Aims: To estimate the cumulative incidence (CI) of cardiac events in a large series
of unselected pts with AML in a single Spanish institution, and to identify risk factors
associated with their development.
Methods: Between January 2011 and June 2020, 571 pts were consecutively diagnosed
with AML in Hospital La Fe, Valencia. The median follow-up of alive patients was 33
months. Clinical records were reviewed from diagnosis to death/last follow-up. Cardiac
events were coded according to CTCAE. There were 525 treated pts, 285 (54%) with intensive
chemotherapy and 240 (46%) with non-intensive therapy.
Results: Among 46 untreated pts, 7 (15%) died due to cardiac events. Median age of
525 treated pts was 65 years, 331 (58%) male, and 73 (13.9%) had prior relevant cardiac
comorbidities; 77 (14.7%) had FLT3-ITD mutation and 38 (7.2%) received front-line
FLT3 inhibitors. Overall, 488 cardiac events were recorded, 19 (3.6%) pts had fatal
cardiac events and 288 (54.9%) had any grade of non-fatal cardiac events. The 9-years
CI of grade 1-2 QTcF prolongation was 11.2%, 2.7% pts had grade 3 QTcF prolongation
>500ms, and no patient had grade 4 o 5. The 9-years CI of grade 1-2 cardiac failure
was 1.3%, grade 3-4 15%, and grade 5 2.1%. The 9-years CI of grade 1-2 arrhythmia
was 1.9%, grade 3-4 9.1%, and no pts had grade 5. Among 307 pts with cardiac events,
38 (12%) pts developed the first event before starting treatment, 132 (43%) during
first cycle, 31 (10%) during consolidation, 55 (18%) during non-intensive further
cycles, 14 (5%) during hematopoietic cell transplantation and 37 (12%) during follow-up.
In the univariate analysis, pts with prior cardiac antecedents had increased incidence
of fatal cardiac events compared with pts with no prior cardiac disease (CI at 9 years
20.1% vs 4.9%, p<0.001). Pts older than 65 years of age (64.4% vs 49.9%, p<0.001),
prior cardiac disease (73% vs 54.6%, p=0.004) or included in clinical trial (65.2%
vs 50.8%, p<0.001) had increased CI of non-fatal cardiac events. Multivariate analysis
showed that prior cardiac disease was associated with increased incidence of fatal
cardiac events [Hazard Ratio (HR) 1.9, p<0.001]. Age >65 (HR 2.2, p<0.001), prior
cardiac disease (HR 1.4, p=0.02) and non-intensive chemotherapy (HR 1.8, p=0.004)
were associated with increased CI of non-fatal cardiac event.
Median overall survival (OS) in the overall cohort was 11.4 months (9.6-13.4 months,
IC 95%). Among 285 intensive therapy pts, median OS was 22 months, 34 months in pts
without cardiac events (n=125), 43 months with grade 1-2 (n=52), 15 months with grade
3-4 (n=98), and 5.2 months with grade 5 (n=10) (p<0.001).
Image:
Summary/Conclusion: We observed a high CI of cardiac events (58.5%) in a real-world
series of patients undergoing AML therapy, associated with significant mortality due
to cardiotoxicity (3.6%), and decreased OS. Prior history of cardiac comorbidity was
associated with an increased risk of fatal cardiac events. Older age, cardiac comorbidities,
and non-intensive therapies were related to non-fatal events.
P512: PRELIMINARY RESULTS OF VEN-A-QUI STUDY: A PHASE 1-2 TRIAL TO ASSESS THE SAFETY
AND EFFICACY OF THE COMBINATION OF AZACITIDINE OR LOW-DOSE CYTARABINE WITH VENETOCLAX
AND QUIZARTINIB IN NEWLY DIAGNOSED
J. M. Bergua-Burgues1,*, R. Rodríguez-Veiga2, I. Cano2, F. Vall-llovera3, A. García-Guiñon4,
J. Gómez-Estruch5, M. Colorado6, I. Casas-Avilés1, J. Esteve-Reyner7, M. V. Verdugo8,
F. Ramos9, M. Valero10, E. Acuña-Cruz2, B. Boluda2, L. Torres-Miñana2, J. Martínez-López11,
E. Barragán2, R. Ayala11, D. Martínez-Cuadrón2, P. Montesinos2
1Hospital San Pedro de Alcántara, Cáceres; 2Hospital Universitari i Politècnic La
Fe, Valencia; 3Hospital Universitari Mútua Terrassa, Barcelona; 4Hospital Arnau de
Vilanova, Lleida; 5Hospital Clinico Universitario Virgen de la Arrixaca, Murcia; 6Hospital
Marqués de Valdecilla, Santander; 7Hospital Clinic Barcelona, Barcelona; 8Hospital
de Jerez, Jerez de la Frontera; 9Hospital Clinico de León, León; 10Hospital Arnau
de Vilanova, Valencia; 11Hospital 12 de Octubre, Madrid, Spain
Background: Venetoclax (VEN) combined with Azacitidine (AZA) or Low Dose Cytarabine
(LDAC) has emerged as new therapeutic option for unfit acute myeloid leukemia (AML)
patients (pts), but primary resistance is observed in roughly 40% of them, while relapses
occur in the vast majority. We speculate that adding a FLT3-ITD inhibitor could improve
the complete remission (CR) and overall survival (OS) rates in this setting.
Aims: To explore the safety and efficacy of VEN-AZA or VEN-LDAC regimens in combination
with Quizartinib (QUI) (VEN-A-QUI trial; EUDRACT2020-000406-28).
Methods: The target population comprised newly diagnosed pts aged ≥ 60 years old unfit
for intensive treatment, including those with secondary AML, with or without prior
exposure to AZA. The Phase I consisted in two arms, one with AZA (Arm A) and the other
with LDAC (Arm B) plus VEN combined with QUI to establish the recommended phase 2
dose (RP2D) of both triplets. Phase 1 scheme was based in 3 + 3 cohorts of patients
observing cycle 1 dose limiting toxicities. Once established the RP2D the phase 2
comprised randomized 1:1 assignment of 60 patients (48 FLT3 wild type and 12 FLT3-ITD
mut) to VEN-AZA-QUI vs. VEN-LDAC-QUI, comparing the CR/CRi rate of both arms. Secondary
objectives were to evaluate the CR/CRi after cycle 1 and 4, compare OS and RFS between
both triplets, quality of life, medical resources, exploration of biomarkers, and
immune recovery.
Results: Data cut-off for preplanned interim analysis included 57 pts screened and
45 enrolled, 16 in phase 1 and 29 in phase 2. Median age was 76,5 years (range 67-87),
males/females (28/23). Previous MDS or MPN was present in 28 pts (59%); and 22 (48%)
had previous treatment with AZA for MDS or MPN phase.
We included 16 pts in phase 1, 9 with AZA and 7 with LDAC. RP2D of QUI was 60 mg in
AZA arm and 40 mg in LDAC arm. No DLT was observed in arm B, and in arm A a brain
hemorrhage after more than 40 days of thrombocytopenia at dose of 60 mg. The safety
committee recommended performing an early (day 14-21) bone marrow assessment in cycle
1, leading to VEN interruption in case of aplastic morphology with grade 4 neutropenia
or thrombocytopenia. No grade ≥3 related non-hematological adverse events (AEs) were
noted during phase 1. The most frequent non-hematological serious AEs during phase
1 were infections (n=23), and gastrointestinal (n=20). No grade 3 QTc prolongation
was observed. Objective responses were CR+CRh+CRi 7 pts (44%), PR 1 (6%), death 4
(25%), and resistance/progression 4 (25%).
Twenty-nine pts (4 with FLT3-ITD mut) were enrolled in the phase 2 (15 in AZA and
14 in LDAC Arm). A median of 1 cycle (range 1-4) was administered at data cut-off,
with best response among 24 evaluable pts: CR+CRh+CRi 10 (42%), MLFS in 3 (12%), PR
5 (21%), death 4 (17%), and resistance/progression 2 (8%). The overall response (CR+CRh+CRi+MLFS)
was 54%. The more frequent non-hematological AEs were infections (n=35) and gastrointestinal
(n=31). Two cardiac failures, 1 chest pain and 1 atrial fibrillation were noted in
phase 2 (all of them unrelated to VEN or QUIZ). No grade 3 QTc prolongation was observed.
Summary/Conclusion: This interim report shows an overall response rate of 54% using
triplets (VEN-AZA-QUI or VEN-LDAC-QUI) for newly diagnosed unfit AML pts. However,
substantial toxicity and early death cases were observed. Of note, 59% of enrolled
pts had secondary AML, and 48% was exposed to AZA before inclusion. Final analyses
with more pts and follow-up will clarify the efficacy and tolerability of these triplets.
P513: CPX-351 TREATMENT FOR ACUTE MYELOID LEUKEMIA IN ENGLAND: REAL-WORLD OUTCOMES
IN ADULTS AGED <60 YEARS VERSUS ≥60 YEARS
A. Legg1,*, R. Muzwidzwa2, A. Lambova2, K. Styles2, P. Doubleday2, G. Medalla1
1Jazz Pharmaceuticals, Oxford; 2IQVIA Inc., London, United Kingdom
Background: CPX-351 (Vyxeos® liposomal) is a dual-drug liposomal encapsulation of
daunorubicin and cytarabine in a synergistic 1:5 molar ratio. Since 2018, NICE and
the EMA have recommended the use of CPX-351 for adults with newly diagnosed, therapy-related
acute myeloid leukemia (t-AML) or AML with myelodysplasia-related changes (AML-MRC)
due to prior myelodysplastic syndrome (MDS)/chronic myelomonocytic leukemia (CMML)
or de novo AML with myelodysplasia-related cytogenetic changes.
Aims: To compare the characteristics and overall survival (OS) of younger (18–60 years)
versus older (≥60 years) adults with AML who were treated with CPX-351 in England.
Methods: This retrospective study utilized the Cancer Analysis System database available
through England’s National Cancer Registration and Analysis Service. A diagnosis of
t-AML or AML-MRC between 01/01/2013 and 31/03/2021 was determined either directly
using International Classification of Diseases for Oncology, Third Edition (ICD-O-3)
codes or indirectly using nonspecific ICD-O-2, ICD-O-3, or ICD-10 AML codes in combination
with prior systemic anticancer therapy or radiotherapy (t-AML) or a prior diagnosis
of MDS/CMML (AML-MRC). OS was estimated from the diagnosis date or landmarked from
the hematopoietic cell transplantation (HCT) date.
Results: Overall, 211 patients with AML received CPX-351; their median (IQR) age was
65 (59, 70) years, 133 (63%) were male, 187 (89%) were White, 87 (41%) had secondary
AML (t-AML or prior MDS/CMML), 33 (16%) had other AML-MRC, and 91 (43%) had unspecified
de novo AML. Of the 60 (28%) patients aged <60 years, 12 were aged 18–44 years and
48 were aged 45–59 years. Of the 151 (72%) patients aged ≥60 years, 97 were aged 60–69
years and 54 were aged ≥70 years.
The cutoff date for OS was 31/08/2021, with a median (IQR) follow-up of 11.3 (4.7,
20.1) months. The estimated median OS (95% CI) was 12.9 (10.5, 17.5) months overall,
18.5 (11.0, not estimable) months for adults aged <60 years, and 11.2 (8.5, 15.9)
months for adults aged ≥60 years, with 2-year survival of 35%, 44%, and 32%, respectively
(Figure). Early mortality at 30 days was 6% overall, 3% for adults aged <60 years,
and 7% for adults aged ≥60 years. Among adults aged <60 years, HCT was reported for
29/60 (48%) patients, including 29/50 (58%) with ≥3 months of follow-up. Among adults
aged ≥60 years, HCT was reported for 55/151 (36%) patients, including 55/115 (48%)
with ≥3 months of follow-up. When landmarked from the HCT date, median OS was not
reached in either age group (Figure).
In a treatment patterns analysis, after receiving CPX-351, 14/60 (23%) adults aged
<60 years and 63/151 (42%) aged ≥60 years died without salvage therapy, and 19/60
(32%) adults aged <60 years and 37/151 (25%) aged ≥60 years were alive without receiving
subsequent therapy by the end of the study period. The most common salvage therapy
after CPX-351 was FLAG-based chemotherapy (aged <60 years: n=12/60; aged ≥60 years:
n=17/151).
Image:
Summary/Conclusion: This study provides real-world survival outcomes in younger and
older adults with AML who were treated with CPX-351 in England. These results suggest
CPX-351 is an effective treatment for patients with AML aged <60 and ≥60 years in
a real-world setting that bridged many patients to HCT, with promising post-HCT outcomes.
P514: V-FAST MASTER TRIAL: PRELIMINARY RESULTS OF TREATMENT WITH CPX-351 PLUS MIDOSTAURIN
IN ADULTS WITH NEWLY DIAGNOSED FLT3-MUTATED ACUTE MYELOID LEUKEMIA
J. McCloskey1,*, V. Pullarkat2, G. Mannis3, T. L. Lin4, S. A. Strickland5, A. T. Fathi6,
H. P. Erba7, S. Faderl8, D. Chakravarthy8, Y. Lutska9, V. Chandrasekaran9, R. S. Cheung8,
M. Levis10
1John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ;
2City of Hope Comprehensive Cancer Center, Duarte, CA; 3Stanford University Medical
Center, Palo Alto, CA; 4University of Kansas Medical Center, Kansas City, KS; 5Vanderbilt-Ingram
Cancer Center, Nashville, TN; 6Massachusetts General Hospital Cancer Center/Harvard
Medical School, Boston, MA; 7Duke University School of Medicine, Durham, NC; 8Jazz
Pharmaceuticals, Palo Alto, CA; 9Jazz Pharmaceuticals, Philadelphia, PA; 10Johns Hopkins
Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States of America
Background:: CPX-351 (Europe: Vyxeos® liposomal; United States: Vyxeos®), a dual-drug
liposomal encapsulation of daunorubicin and cytarabine in a synergistic 1:5 molar
ratio, is approved for newly diagnosed, therapy-related acute myeloid leukemia (AML)
or AML with myelodysplasia-related changes in adults in Europe and patients aged ≥1
year in the United States. In a phase 3 study in older adults with newly diagnosed,
high-risk/secondary AML, CPX-351 significantly improved overall survival and remission
rates versus conventional 7 + 3, with a comparable safety profile. Preclinical data
suggest CPX-351 may have synergistic activity with targeted agents, including the
FLT3 inhibitor midostaurin.
Aims: Herein, we report preliminary results for the cohort of adults treated with
CPX-351 + midostaurin in the V-FAST (Vyxeos – First Phase Assessment with Targeted
Agents) trial.
Methods: V-FAST is an open-label, multicenter, multi-arm, nonrandomized, phase 1b
master trial (NCT04075747) to evaluate the safety and preliminary efficacy of CPX-351
combined with targeted agents (midostaurin, venetoclax, enasidenib). Eligible adults
in the CPX-351 + midostaurin cohort were aged 18 to 75 years, had newly diagnosed
AML with a FLT3 internal tandem duplication (ITD) or tyrosine kinase domain (TKD)
mutation, were fit for intensive chemotherapy, and had an Eastern Cooperative Oncology
Group (ECOG) performance status of 0 to 2. The dose-exploration phase (3 + 3 design)
determined a recommended phase 2 dose of CPX-351 100 units/m2 (daunorubicin 44 mg/m2
+ cytarabine 100 mg/m2) on Days 1, 3, and 5 + midostaurin 50 mg twice daily on Days
8 to 21. There were no dose-limiting toxicities, and additional patients were enrolled
in the expansion phase at this dose.
Results: A total of 23 patients received CPX-351 + midostaurin and had sufficient
data to be included in the analysis (cutoff date: 20 January 2022). Patient baseline
characteristics are shown in the Table. Treatment-emergent adverse events (TEAEs)
in ≥40% of patients included febrile neutropenia (78%), nausea (65%), increased alanine
aminotransferase (57%), leukopenia (57%), thrombocytopenia (57%), headache (43%),
and hyponatremia (43%). All patients experienced a grade 3/4 TEAE, primarily hematologic
events. Nonhematologic grade 3/4 TEAEs in ≥2 patients included pneumonia (17%), lung
infection (13%), and hyperglycemia (9%). There were no grade 5 TEAEs and no deaths
on or before Day 60. Complete remission was achieved by 18/22 (82%) patients with
an evaluable remission assessment after the first induction cycle.
Image:
Summary/Conclusion: Preliminary results from the V-FAST trial suggest the combination
of CPX-351 + midostaurin is feasible, with a manageable safety profile and promising
remission rates in adults with newly diagnosed AML who have a FLT3 mutation.
P515: LOWER-INTENSITY CPX-351 + VENETOCLAX FOR PATIENTS WITH NEWLY DIAGNOSED ACUTE
MYELOID LEUKEMIA WHO ARE UNFIT FOR INTENSIVE CHEMOTHERAPY
G. L. Uy1, V. A. Pullarkat2,*, P. Baratam3, R. K. Stuart3, R. B. Walter4, E. S. Winer5,
S. Faderl6, V. Chandrasekaran6, Q. Wang6, D. Chakravarthy6, R. S. Cheung6, T. L. Lin7
1Washington University School of Medicine, St. Louis, MO; 2City of Hope Comprehensive
Cancer Center, Duarte, CA; 3Medical University of South Carolina, Charleston, SC;
4Fred Hutchinson Cancer Research Center, Seattle, WA; 5Dana-Farber Cancer Institute,
Harvard Medical School, Boston, MA; 6Jazz Pharmaceuticals, Palo Alto, CA; 7University
of Kansas Medical Center, Kansas City, KS, United States of America
Background: CPX-351 (Europe: Vyxeos® liposomal; United States: Vyxeos®) is a dual-drug
liposomal encapsulation of daunorubicin and cytarabine in a synergistic 1:5 molar
ratio. CPX-351 is approved for newly diagnosed, therapy-related acute myeloid leukemia
(AML) or AML with myelodysplasia-related changes in adults in Europe and patients
aged ≥1 year in the United States who are candidates for intensive chemotherapy (IC).
However, the appropriate dosage of CPX-351 in patients unfit for IC may be different
from the label dosage. Venetoclax (VEN; BCL-2 inhibitor) + low-dose cytarabine has
demonstrated efficacy in unfit patients with AML, and drug synergism/additivity in
preclinical studies provided a rationale for combining CPX-351 + VEN clinically.
Aims: Our study evaluates the safety and efficacy of lower-intensity CPX-351 + VEN
in adults with newly diagnosed AML who are unfit for IC.
Methods: This is an ongoing, open-label, phase 1b study (NCT04038437). Patients who
achieve at least partial remission after 1 or 2 cycles may receive up to 4 similar
cycles in the dose-exploration phase (DEP) or up to 8 similar cycles in the expansion
phase (EP). Patients are assessed for response (morphology, measurable residual disease
[MRD]) and monitored for safety and survival.
Results: The data include 31 patients enrolled by 15 September 2021, with a data cutoff
of 2 December 2021: 4 patients in the DEP at dose level 1 (CPX-351 20 units/m2 on
Days 1 and 3 + VEN 400 mg on Days 2 to 21 of each cycle), 7 patients in the DEP at
dose level 2 (CPX-351 40 units/m2 + VEN 400 mg), and a total of 20 patients in the
DEP and EP at dose level 1b (CPX-351 30 units/m2 + VEN 400 mg), which was established
as the recommended phase 2 dose. Patients were considered unfit for IC based on age
≥75 years (n=15) or health (Eastern Cooperative Oncology Group performance status
of 2 to 3 and/or comorbidities [n=16]). Median age was 74 years (range: 60, 90); 65%
were male; 77% had de novo AML; 58% had poor-risk disease; and 23% had a TP53 mutation.
Nonhematologic treatment-emergent adverse events (TEAEs) in ≥20% of patients were
diarrhea (26%), cough (23%), dyspnea (23%), and nausea (23%). Hematologic grade ≥3
TEAEs were reported in 17 (55%) patients; no nonhematologic grade ≥3 TEAE was reported
in >10% of patients. There were no deaths by Day 30; mortality at Day 60 was 13%,
with deaths due to myocardial infarction unrelated to therapy (n=1), worsening lung
infection (n=1), and disease progression (n=2). Median (interquartile range) recovery
times were 30 days (22, 34.5) to neutrophils ≥500/μL and 21 days (21, 27) to platelets
≥50,000/μL.
Complete remission (CR) or CR with incomplete neutrophil or platelet recovery (CRi)
was achieved by 16/28 (57%) patients with an evaluable remission assessment. All 16
of these patients achieved remission (CR or CRi) after the first treatment cycle.
MRD negativity was achieved by 12/16 (75%) patients with CR or CRi, primarily after
Cycle 1 (Cycle 1: n=8; Cycle 2: n=2; Cycle 3: n=1; Cycle 4: n=1). Survival data are
not yet mature.
Summary/Conclusion: Lower-intensity CPX-351 + VEN was generally well tolerated in
adults with newly diagnosed AML who are unfit for IC and showed promising initial
efficacy, with CR or CRi in the majority of patients.
P516: ADULT LANGERHANS CELL HISTIOCYTOSIS WITH THYROID GLAND INVOLVEMENT: CLINICAL
PRESENTATION, GENOMIC ANALYSIS AND OUTCOME
H. Cai1,2,*, T. Liu1, H. Cai1,2, M. Duan1,2, J. Li1,2, D. Zhou1,2, X. Cao1,2
1Department of Hematology; 2State Key Laboratory of Complex Severe and Rare Diseases,
Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking
Union Medical College, Beijing, China
Background: Langerhans cell histiocytosis (LCH) is a rare, clonal disorder derived
from CD1a-positive and CD207-positive immature myeloid dendritic cells. The clinical
presentations and outcomes of LCH are extremely variable. Thyroid gland involvement
is rare, and solitary thyroid gland involvement with LCH is extremely rare, with most
cases presenting as part of multisystem disease.Until now, little information exists
on the genomic analysis of LCH with thyroid gland involvement with or without coexisting
papillary thyroid carcinoma.
Aims: To evaluate the characteristics and treatment outcomes of adult Langerhans cell
histiocytosis (LCH) patients with thyroid involvement.
Methods: We retrospectively described the clinical, biological, and genomic characteristics
of a series of 36 LCH patients with thyroid involvement in our centre between January
2001 and December 2021.
Results: We retrospectively described the clinical, biological, and genomic characteristics
of a series of 36 LCH patients with thyroid involvement in our centre between January
2001 and December 2021. At the time of diagnosis, only 1 patient was classified as
having single-system LCH, and 35 patients were classified as having multisystem (MS)
LCH. Three patients had coexisting papillary thyroid carcinoma. Patients with thyroid
gland involvement had higher frequencies of pituitary (88.6% vs. 53.4%, P<0.001),
liver (45.7% vs. 20.7%, P=0.003) and lymph node (54.3% vs. 31.6%, P=0.012) involvement
and a lower frequency of bone (45.7% vs. 72.0%, P=0.003) involvement than patients
without thyroid gland involvement. Sixteen patients had abnormal thyroid function,
including 9 patients with primary hypothyroidism, 1 patient with central hypothyroidism,
and 6 patients with subclinical hypothyroidism. BRAF
V600E, BRAF
N486_P490 and MAP2K1 mutations were detected in 14.3%, 57.1% and 7.1% of patients,
respectively. After a 43-month median follow-up, none of the patients died, and 15
patients experienced reactivation. The median event free survival was 37.5 months.
Two of 6 patients with subclinical hypothyroidism had normal thyroid function, and
12 patients still had hypothyroidism after treatment.
Summary/Conclusion: As the largest adult LCH cohort with thyroid gland involvement
to date, we found that patients with thyroid gland involvement had different clinical
characteristics, genetic profiles and outcomes than patients without thyroid gland
involvement.
P517: PROSPECTIVE MULTICENTRIC STUDY ON INFECTIOUS COMPLICATIONS AND CLINICAL OUTCOME
IN 230 UNFIT AML PATIENTS TREATED IN FIRST-LINE WITH HYPOMETHYLATING AGENTS ALONE
OR IN COMBINATION WITH VENETOCLAX.
A. Candoni1,*, M. Piccini2, D. Lazzarotto1, V. Bonuomo3, M. Dargenio4, M. Riva5, L.
Mellillo6, C. Papayannidis7, M. Stulle8, G. Dragonetti9, M. I. Del Principe10, C.
Cattaneo11, C. Pasciolla12, R. De Marchi13, M. Delia14, M. C. Tisi15, M. E. Zannier1,
G. Nadali3, M. Sciumè16, A. Spadea17, C. Sartor7, D. Griguolo8, E. Buzzatti10, C.
M. Basilico18, C. Sarlo19, A. L. Piccioni20, R. Cairoli5, A. Olivieri21, L. Pagano9
1Division of Hematology and Stem Cell Transplantation, ASUFC, University of Udine,
Udine; 2Division of Hematology, Azienda Ospedaliero-Universitaria Careggi, Florence;
3Division of Hematology, AOUI, Policlinico GB Rossi, Verona; 4Division of Hematology,
Ospedale Vito Fazzi, Lecce; 5Dipartimento di Ematologia ed Oncologia, Niguarda Cancer
Center ASST Grande Ospedale Metropolitano, Milano; 6Division of Hematology, Lecce
and IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Lecce; 7Institute
of Hematology and Medical Oncology “L. and A. Seragnoli”, University of Bologna, Bologna;
8Division of Hematology, ASUGI, Trieste; 9Division of Hematology, Polo Onco-Ematologico,
Fondazione Policlinico A. Gemelli-IRCCS, Università Cattolica del Sacro Cuore; 10Hematology,
Fondazione Policlinico Tor Vergata, Roma; 11Section of Hematology, Spedali Civili,
Brescia; 12Haematology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari; 13Onco
Hematology, Department of Oncology, Veneto Institute of Oncology IOV, IRCCS, Padova;
14Hematology and Bone Marrow Transplantation Unit, Azienda Ospedaliero-Universitaria
Consorziale Policlinico-University of Bari, Bari; 15Cell Therapy and Hematology, San
Bortolo Hospital, Vicenza; 16U.O. Oncoematologia, Fondazione IRCCS Ca’ Granda Ospedale
Maggiore Policlinico, University of Milan, Milano; 17Hematology and Stem Cell Transplant
Unit, IRCCS Regina Elena National Cancer Institute, Roma; 18Division of Hematology,
ASST Sette Laghi, Ospedale Circolo e Fondazione Macchi, Varese; 19Hematology and Stem
Cell Transplantation Unit, University Campus Bio-Medico; 20Dipartimento di Ematologia,
Azienda Ospedaliera San Giovanni Addolorata, Roma; 21Division of Hematology, Azienda
Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
Background: The hypomethylating agents (HMAs) are an important therapeutic option
for older patients (pts) with AML and have become the backbone for combination regimens
(eg, with Venetoclax). However, there are very limited real-life prospective studies
regarding clinical outcome of these pts, including infectious complications and infection
related mortality (IRM) during treatment.
Aims: To investigate the infectious complications and clinical outcome in AML patients
treated with HMAs± Venetoclax (V) outside of clinical trials.
Methods: The recruitment of this prospective multicentric study (CE-Id-study:2908)
has been completed on December 31, 2020. We enrolled 230 AML pts with a median age
of 75 years (range 25-94); 157 pts (68%) had >2 relevant comorbidities. Of the 230
cases, 132 (57%) received a first-line therapy with a combination of HMAs+V while
98 (43%) were treated with HMAs monotherapy (azacitidine or decitabine). A total of
1550 cycles of HMAs have been administered (680/1550 with HMAs+V).
Results: The best response achieved, with HMAs treatment, was: CR in 44% of cases
(57,6% with HMAs+V and 25,5% with HMAs alone, P=0,0001), PR in 17% and SD in 14% of
cases (ORR 61%; 72% in HMAS+V and 46% in HMAs alone, P=0,0007). The microbiological
or radiological proven infectious complications (almost one) occurred in 160/230 (70%)
of pts, mainly pneumonia (in 42% of pts) and/or bacteremia/sepsis (one or more events
in 29% of pts). Febrile neutropenia (one or more episodes) occurred in 38% of pts
and 14 cases of Covid-19 (6%) were reported. After a median follow-up of 9 months
(1-24) from the start of HMAs therapy, 144 (63%) pts died and 86 (37%) were alive.
The 1 yr OS probability was 46% with a median OS of 10,3 months (11 months in HMAs+V
and 9 months in HMAs alone; P=ns). The primary causes of death were: progression of
AML (42%), Infection (26%-37/144), Infection+AML (24%), other causes (8%). The IRM
was 26% and 19/144 (13%) pts died of infectious complication while in CR/PR (16 in
HMAs+V group and only 3 in HMAs group; P=0,005). Data on antibiotic prophylaxis, hospitalization,
drug-doses modulation, are available and analyzed in this study.
Summary/Conclusion: The results of this real-life, multicentric, prospective study,
confirm a higher CR rate in pts treated with HMAs+V compared to HMAs alone (P=0,0001).
However, we found a high rate of infectious complications and IRM (26%) with a higher
infection related deaths in patients in CR/PR who were treated with HMAs+V (P=0,005).
Findings from this study highlight the critical relevance of infection prevention
in reducing infectious mortality, which adversely impacts the OS of this frail AML
population.
P518: PEDAL/EUPAL INTERNATIONAL COLLABORATION TO IMPROVE THE OUTCOME OF CHILDREN WITH
RELAPSED OR REFRACTORY ACUTE MYELOID LEUKEMIA
V. Ceolin1,2,3,*, S. Ishimaru1, F. Bautista1,4, B. Goemans1, E. A. Kolb5, L. Di Laurenzio6,
T. M. Cooper7, M. Bakker1, J. Wahlstrom8, G. Sunkersett8, G. Ku9, C. Zwaan1,3,4, D.
Reinhardt10, G. Nichols6
1Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands; 2Regina Margherita
Children’s Hospital, University of Turin, Turin, Italy; 3Sophia Children’s Hospital,
Erasmus University MC, Rotterdam, Netherlands; 4ITCC consortium, Paris, France; 5Nemours
Centers for Childhood Cancer Research & Cancer and Blood Disorders, Wilmington; 6The
Leukemia & Lymphoma Society, New York; 7Seattle Children’s Hospital, Washington; 8AbbVie
One Oncology; 9Genentech Inc, San Francisco, United States of America; 10University
Hospital of Essen, Essen, Germany
Background: The prognosis of children with acute myeloid leukemia (AML) has increased
stepwise over the last decades, and the overall survival (OS) is currently in the
75-80% range. However, at relapse, OS is approximately 40-50%. Over the last decade,
many novel drugs have been developed specifically for use in adults with AML and few
have been authorized for children. Given the medical need, effective and less toxic
therapies are urgently also needed for children to overcome the limitations of current
intensive chemotherapy.
Aims: The Leukemia & Lymphoma Society (LLS) Pediatric Acute Leukemia (PedAL) program
is a strategic initiative that aims to overcome the obstacles in treating children
with acute leukemia via a Transatlantic collaboration. The aim is to run a program
of early and late phase clinical trials evaluating the safety and the efficacy of
new agents in pediatric leukemia, eventually setting a new standard of treatment for
relapsed AML. In Europe this will be implemented as a master clinical trial with sub-trials.
The European Pediatric Acute Leukemia (EuPAL, including United Kingdom) consortium
was set up to coordinate the European efforts for the PedAL initiative.
Methods: In Europe, the current project consists of a registry protocol (EuPAL2021
Registry) and a Master protocol with sub-trials (ITCC-101). In North America patients
will be enrolled in the PedAL Screening Protocol - APAL2020SC. Any patient with known
or suspected relapsed or refractory AML, who is less than 22 years of age will be
eligible for inclusion in the registry in Europe or in the screening protocol in North
America, to provide confirmation of relapse and target expression using standardized
methodologies. In Europe, the EuPAL 2021 Registry will lead to harmonization of diagnostics
in country/collaborative group-specific centralized laboratories where the analysis
will be carried out as part of standard of care. In North America, Australia and New
Zealand APAL2020SC will utilize commercial laboratories (Hematologics, Foundation
Medicine). The ITCC-101 PedAL/EuPAL Master protocol is a complex clinical trial with
a stratification approach to allocate these patients to sub-trials, according to relevant
biomarkers and disease related information. After confirmation of relapse, patients
may be enrolled in one of the open sub-trials, provided the detailed eligibility criteria
are met. Each sub-trial will effectively run as one study performed on a global scale,
and data will be entered in one database. Sub-trials may not start at the same time
and may be added by a substantial amendment to the master. In North America each sub-trial
will be added as an amendment to the LLS Investigational New Drug. For the first wave
of sub-trials, Investigational Medicinal Product prioritization was mainly achieved
through international collaboration at the Pediatric Forum for AML organized by ACCELERATE
Strategy Forum in April 2020.
Results: The first sub-trial will be APAL2020D, an open-label Phase III randomized
multicenter trial to assess if venetoclax combined with FLA+GO (fludarabine, high-dose
cytarabine, and gemtuzumab ozogamicin) will improve overall survival compared to FLA+GO,
with the aim to establish the standard treatment for the 2nd relapsed AML patients
(NCT05183035) and will open in Q2/Q3 2022.
Summary/Conclusion: The PedAL/EuPAL international collaboration will determine, using
biology-based selection markers for treatment stratification, the standards of care
for AML in 1st and 2nd relapse and define the safety and efficacy parameters necessary
for the efficient evaluation of novel targeted therapies.
P519: TRANSFUSION INDEPENDENCE AMONG NEWLY DIAGNOSED ACUTE MYELOID LEUKEMIA PATIENTS
RECEIVING VENETOCLAX-BASED COMBINATIONS VS OTHER THERAPIES: RESULTS FROM THE AML REAL
WORLD EVIDENCE (ARC) INITIATIVE
O. Wolach1, J. S. Garcia2, P. Vachhani3, J. Zeidner4, C. Lai5, Y. Moshe6, M. Xavier7,
S. Lee8, T. Zuckerman9, D. A. Pollyea10, S. Abedin11, E. C. Chen2, S. Bathini3, W.
Edwards7, C. N. Bui12, W. W.-H. Cheng12, A. Svensson12, A. Guérin13, J. Maitland13,
E. Ma14, M. Montez14, M. Grunspan15, A. D. Goldberg16,*
1Rabin Medical Center, Petah Tikya, Israel; 2Dana-Farber Cancer Institute, Boston;
3O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham;
4University of North Carolina, Lineberger Comprehensive Cancer Center, Chapel Hill;
5University of Pennsylvania Perelman Center for Advanced Medicine, Philadelphia, United
States of America; 6Tel Aviv (Sourasky) Medical Center, Tel Aviv, Israel; 7Scripps-MD
Anderson, San Diego; 8Janssen Research and Development, Springhouse, United States
of America; 9Rambam Health Care Campus, Haifa, Israel; 10University of Colorado School
of Medicine, Aurora; 11Medical College of Wisconsin, Milwaukee; 12AbbVie Inc, North
Chicago, United States of America; 13Analysis Group Inc, Montreal, Canada; 14Genentech,
South San Francisco, United States of America; 15AbbVie Inc, Hod-Hasharon, Israel;
16Memorial Sloan Kettering Cancer Center, New York, United States of America
Background: Venetoclax (VEN), a novel BCL-2 inhibitor, is FDA approved in combination
with hypomethylating agents (azacitidine or decitabine) or low-dose cytarabine for
the treatment of newly-diagnosed (ND) acute myeloid leukemia (AML) in adults ≥75 years
or those who have comorbidities that preclude use of intensive induction chemotherapy.
This study is part of the ongoing AML Real world evidenCe (ARC) Initiative which aims
to provide insight into the use and relative efficacy of VEN-based combinations among
ND patients (pts) with AML.
Aims: To describe transfusion independence (TI) among ND AML pts treated with VEN-based
combinations vs non-VEN regimens in clinical practice.
Methods: The ARC Initiative is a multicenter chart review study of adult pts with
AML who received VEN ≥April 2016 (VEN cohort) or non-VEN regimens ≥May 2015 (control
cohort). Pts in the VEN cohort were matched 1:1 to pts in the control cohort based
on age category (<60; 60-74; ≥75) and European Leukemia Net (ELN) risk classification.
Outcomes, including TI, hematopoietic cell transplantation (HCT), and best response,
were assessed during first-line therapy over a transfusion observation period defined
as time from initiation of therapy to the earliest amongst progression, initiation
of second-line therapy, admission to hospice, death, or last visit at study site.
TI was defined as no red blood cell (RBC) or platelet (PLT) transfusions during any
consecutive ≥56-day period during the transfusion observation period. Kaplan-Meier
(KM) analyses were conducted to assess the TI rate at 3 months after treatment initiation.
Interim descriptive results are presented from data cutoff of Sept 2021; data will
be updated for the meeting.
Results: A total of 119 VEN and 119 matched control pts with ND AML who had available
information on transfusions were included in this analysis (Table 1). Among pts in
the control cohort, 65.5% received a high intensity regimen (e.g., cytarabine+daunorubicin
[37 pts, 31.1%], CPX-351 [16 pts, 13.4%]) and 34.5% received a low intensity regimen
(decitabine [24 pts, 20.2%], azacitidine [15 pts, 12.6%]). Of pts tested for genetic
mutations, common mutations were TP53 (VEN: 23.1%; control: 15.8%), RUNX1 (VEN: 17.1%;
control: 17.5%), and IDH1/IDH2 (VEN: 12.8%; control: 14.0%). During a median transfusion
observation period of 4.0 months in the VEN cohort and 3.6 months in the control cohort,
87.4% of VEN and 93.3% of control pts received ≥1 RBC or PLT transfusion. Pts in the
VEN cohort received a median of 2.0 RBC and/or PLT transfusions per month and 5.0%
received HCT; pts in the control cohort received a median of 3.5 RBC and/or PLT transfusions
per month and 13.4% received HCT (all p <0.05). Among the 103 pts in the VEN cohort
and 99 pts in the control cohort who had ≥56 days of follow-up, 61.2% of VEN pts and
50.5% of control pts achieved TI during first-line therapy (p=0.17), and 60.6% of
VEN pts and 55.8% of control pts with response data achieved response (p=0.59). The
KM rate of achieving TI at 3 months was 45.0% in the VEN cohort and 35.9% in the control
cohort (log-rank p=0.12). Pts in the VEN and control cohort achieved TI after a median
of 2.3 and 2.8 months, respectively (p=0.08).
Image:
Summary/Conclusion: Interim ARC initiative results show that pts receiving VEN-based
combinations receive statistically fewer transfusions and are trending towards achieving
TI more frequently and reaching TI more quickly after initiation of treatment compared
to matched control pts receiving low and high intensity therapies. Further analyses
are planned to understand the real-world outcomes of ND pts with AML.
P520: PHASE 1/2 STUDY OF SEL24/MEN1703, A FIRST-IN-CLASS DUAL PIM/FLT3 KINASE INHIBITOR,
IN PATIENTS WITH IDH1/2-MUTATED ACUTE MYELOID LEUKEMIA: THE DIAMOND-01 TRIAL.
G. Martinelli1,*, A. Santoro2, C. Gambacorti-Passerini3, S. Vives Polo4, S. R. Solomon5,
S. Mukherjee6, E. Lech-Maranda7, M. Yair Levy8, A. Wierzbowska9, M. Calbacho-Robles10,
G. Marconi11, M. Benedetta Giannini11, I. Cano12, L. Torres Miñana13, E. Acuña-Cruz12,
N. Angelosanto14, A. Galleu15, S. Baldini15, S. Blotta15, F. Ravandi16, P. Montesinos17
1IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori”-IRST S.r.l., Meldola;
2Department of Oncology, IRCCS, Humanitas Research Hospital; 3Department of Hematology,
University of Milano-Bicocca, Milan, Italy; 4Institut Català d’Oncologia-Hospital
Germans Trias i Pujol, Badalona, Spain; 5Northside Hospital Cancer Institute, Atlanta;
6Taussig Cancer Institute, Cleveland Clinic, Cleveland, United States of America;
7Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw,
Poland; 8Texas Oncology-Baylor Charles A. Sammons Cancer Center, Dallas, United States
of America; 9Department of Hematology, Medical University of Lodz, Lodz, Poland; 10Department
of Hematology, Ramón y Cajal University Hospital, Madrid, Spain; 11IRCCS Istituto
Romagnolo per lo Studio dei Tumori “Dino Amadori” - IRST S.r.l., Meldola, Italy; 12Department
of Hematology; 13Department of Hematology, La Fe University Hospital, Valencia, Spain;
1413Clinical Development-Hematology, Ryvu Therapeutics, Krakow, Poland; 15Hematologic
Malignancies - Global Oncology TA, Menarini Group, Florence, Italy; 16Department of
Leukemia, The University of Texas MD Anderson Cancer Center, Houston, United States
of America; 17La Fe University Hospital, Valencia, Spain
Background: Mutations in the FLT3 tyrosine kinase are the most frequent mutations
that occur in adults with acute myeloid leukemia (AML) and can co-occur with mutations
in IDH1 or IDH2 (collectively IDHm) in up to 30% of cases. SEL24/MEN1703 is an orally
available, first-in-class, dual PIM/FLT3 kinase inhibitor. Preliminary results from
the phase 1/2 first-in-human DIAMOND-01 trial (NCT03008187) evaluating single-agent
SEL24/MEN1703 showed activity in adults with relapsed/refractory (R/R) IDHm AML, where
3 of 8 IDHm patients responded.
Aims: Here we report the first safety and efficacy results from an additional expansion
cohort of the DIAMOND-01 trial in 20 patients with R/R IDHm AML.
Methods: Patients with IDHm R/R AML and no standard therapeutic options were eligible.
The recommended dose of 125 mg SEL24/MEN1703 was given orally, once daily for 14 days
over a 21-day cycle until disease progression or unacceptable toxicity. The primary
endpoint was safety, and adverse events (AEs) were graded according to NCI CTCAE v4.03.
The secondary endpoint was antileukemic activity including overall response rate (ORR).
Results: As of 10 January 2022, 14 patients were enrolled in the IDHm cohort. Median
age was 68 years (range 37–79). Four patients had AML secondary to myelodysplastic
syndrome and 7 patients had intermediate cytogenetic risk. Median number of prior
lines of treatment was 2 (range 1–3). Seven patients had IDH2, 1 had IDH1/2, and 4
had IDH1 mutations. Concomitant mutations in FLT3-ITD were detected in 2 patients.
Median duration of treatment was 2 cycles (range 1–8). Safety data (N=12) showed that
serious treatment-emergent AEs (TEAEs; reported in ≥5% patients) were pneumonia (33%),
skin infection, and clostridial gastroenteritis (8% each). These events were all unrelated
to study drug. Drug-related TEAEs were liver injury and hyponatremia (8% each). The
drug-related liver injury occurred in a patient who was concomitantly receiving other
drugs with known hepatotoxic potential. Grade ≥3 TEAEs (≥10% patients) were pneumonia
(33%) and asthenia (17%), both unrelated to study drug. No differentiation syndrome
was observed. Of the 7 patients who completed ≥1 treatment cycle and had ≥1 postbaseline
assessment or clear disease progression, ORR was 28.6%; 1 patient achieved a complete
response with incomplete blood count recovery (CRi) at cycle 3 and underwent hematopoietic
stem cell transplant, 1 patient had a partial response at cycle 4 (confirmed at cycle
7 and still on treatment), 4 had disease progression, and 1 discontinued for an AE
(not drug related). Among the 7 remaining patients, 3 discontinued before completion
of cycle 1 without progression or response, while 4 patients were ongoing and have
not yet had any postbaseline assessment.
Summary/Conclusion: Preliminary results in the IDHm cohort confirm that SEL24/MEN1703,
a first-in-class, orally available, dual PIM/FLT3 inhibitor, has a manageable safety
profile and single-agent activity in patients with R/R IDHm AML. Updated results will
be presented at the congress.
P521: TREATMENT OF BLASTIC PLASMACYTOID DENDRITIC CELL NEOPLASM IN PEDIATRIC PATIENTS
WITH TAGRAXOFUSP, A CD123-TARGETED THERAPY
N. Pemmaraju1,*, B. Cuglievan1, J. Lasky2, A. Kheradpour3, N. Hijiya4, A. S. Stein5,
S. Meshinchi6, C. Mullen7, E. Angelucci8, L. Vinti9, T. I. Mughal10,11, A. Pawlowska12
1Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston;
2Cure 4 The Kids Foundation, Las Vegas; 3Department of Pediatric Hematology and Oncology,
Loma Linda University Children Hospital, Loma Linda; 4Division of Pediatric Oncology,
Hematology, and Stem Cell Transplantation, Columbia University Irving Medical Center,
New York; 5Department of Hematology and Hematopoietic Cell Transplantation, City of
Hope National Medical Center, Duarte; 6Department of Pediatrics, University of Washington
School of Medicine, Seattle; 7Division of Pediatric Hematology/Oncology, Department
of Pediatrics, Golisano Children’s Hospital, University of Rochester, Rochester, United
States of America; 8Hematology and cellular therapy unit, IRCCS Ospedale Policlinico
San Martino, Genova; 9Department of Paediatric Haematology/Oncology, Cell and Gene
Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy; 10Division of Hematology-Oncology,
Tufts University Medical School, Boston; 11Stemline Therapeutics Inc, New York; 12Department
of Pediatrics, City of Hope National Medical Center, Duarte, United States of America
Background: Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and aggressive
hematologic malignancy derived from plasmacytoid dendritic cell precursors that overexpress
CD123, the interleukin-3 receptor alpha. Though primarily a disease of the elderly,
BPDCN has been documented in infants and children in 10–20% of all cases. The rarity
of BPDCN in the pediatric population makes collecting robust safety and efficacy data
challenging and there is a paucity of data published for pediatric patients (pts).
A first-in-class, CD123-directed therapy, tagraxofusp (TAG, SL-401) is the only approved
treatment for patients with BPDCN. A multicohort prospective study (NCT02113982) demonstrated
a tolerable safety profile and a durable overall response rate of 75% (median duration
24.9 months) in first-line (1L) pts. TAG is approved by the US FDA for the treatment
of BPDCN in pts over the age of 2 years with newly diagnosed and relapsed or refractory
(R/R) BPDCN; it is also approved by the EMA for 1L treatment of adult pts. Herein,
we present a case series of pediatric pts with BPDCN treated with TAG, to provide
further insight into the efficacy and safety of this therapy in this understudied
patient population.
Aims: To expand on the body of data regarding TAG therapy in pediatric pts with BPDCN.
Methods: Pediatric case reports of BPDCN were collected from centers across the US
and Europe. TAG was administered according to local institutional guidelines 1L, or
R/R. Data was collated and summarized descriptively. Analyses included tumor response,
survival, and safety (adverse events [AEs] and laboratory values).
Results: Eight pediatric pts with BPDCN were treated with TAG and included in this
analysis. Median age was 15.5 years (range 2 – 21 years), and 7 of the 8 pts were
female. Five pts were treatment naïve, and 3 pts were R/R. Four pts had skin only
disease. Two pts had skin, bone marrow (BM), and lymph node (LN) involvement; 1 of
these also had central nervous system (CNS) involvement. One patient had BM and LN,
but no skin involvement, and 1 patient had BM only disease. A 2-year-old patient received
7 mcg/kg TAG in combination with venetoclax and azacitidine at second relapse; all
other pts received 12 mcg/kg throughout all treatment cycles. The number of TAG cycles
administered ranged from 1 to 4 at the time of data cutoff. A decrease in albumin
occurred in 2 pts, and 2 pts had a rise in transaminases, all of which were manageable.
One patient experienced headaches, hot flashes, fatigue, and mouth sores. One patient
experienced grade 2 capillary leak syndrome in cycle 1, which was treated and resolved,
and the patient went on to receive subsequent cycles. No AEs were seen in 3 pts. Three
1L pts achieved a complete response, including 1 patient with extensive disease (skin,
bone marrow, and CNS) who also received intrathecal therapy. One 1L and 1 R/R patient
achieved stable disease (no progression after 4 TAG cycles each), and 1 patient with
R/R BPDCN progressed while on TAG. Five pts treated with TAG were bridged to a stem
cell transplant (n=3 1L, n=2 R/R; Table 1). Median survival data will be presented
(6 of 8 pts remain alive).
Image:
Summary/Conclusion: These cases expand the knowledge base of BPDCN treatment in pediatric
pts. TAG was well tolerated in all pts, with a manageable safety profile that was
similar to that reported for adults in the pivotal study. Treatment with TAG demonstrated
promising efficacy, with responses that enabled 5 of 8 pediatric pts, including 3
1L and 2 R/R, to be bridged to stem cell transplant.
P522: ABSOLUTE LYMPHOCYTE COUNT IS AN INDEPENDENT SURVIVAL PREDICTOR IN PATIENTS WITH
ACUTE MYELOID LEUKEMIA TREATED WITH INTENSIVE CHEMOTHERAPY
G. Cristiano1,*, J. Nanni1, L. Zannoni1, C. Sartor1, S. Parisi2, S. Paolini2, C. Papayannidis2,
M. Cavo1,2, A. Curti2
1Hematology, Department of Experimental, Diagnostic and Specialty Medicine, University
of Bologna; 2Hematology, IRCCS Azienda ospedaliero-universitaria di Bologna, Istituto
di Ematologia “Seràgnoli”, Bologna, Italy
Background: Absolute lymphocyte count (ALC) is known to be an independent prognostic
factor for overall survival (OS) in patients receiving autologous transplantation
for Hodgkin’s and non-Hodgkin’s lymphomas, multiple myeloma and breast cancer. In
acute myeloid leukemia (AML) patients, enhanced ALC recovery after intensive chemotherapy
(IC) has been associated with superior OS and leukemia-free survival (LFS). However,
few if any data correlated ALC recovery with novel therapies as well as with updated
prognostic factors, such as European Leukemia Net (ELN) risk-classification.
Aims: Our study aims at evaluating the predictive value of ALC recovery in AML patients
treated with IC by analysing its impact through different patient subgroups according
to therapy response, type of chemotherapy regimen and the use of allogeneic stem cell
transplantations (HSCT).
Methods: We evaluated 148 newly diagnosed adult AML patients treated with IC at Bologna
Seràgnoli Hematology Institute since 2007. We defined 4 ALC time-points (TPs): at
15, 21 and 28 days from the start of induction chemotherapy and before consolidation
therapy (CC). ALC cut-off was established at 500/mm3. In addition to the assessment
of the single TP, patients were also grouped in those who obtained ≥500/mm3 ALC in
all 4 TPs and those who had <500/mm3 ALC in at least one TP. Median follow-up was
51,4 months.
Results: When ALC recovery was assessed at the single TP, only ALC-CC recovery showed
a statistically significant association with better OS and LFS. However, significant
outcome differences were observed among CR patients (76,3%), subdivided in those who
achieved ALC in all TPs vs others. A trend toward better correlation of ALC with OS
was observed in patients who received “3 + 7-based” regimen (30,8%;) versus those
who were treated with “fludarabine-based” regimen (69,1%;). Better outcome was observed
in “3 + 7” patients who had ALC recovery in all TPs and at day 15 post-chemotherapy.
Of note, in the “3 + 7” group, FLT3-ITD positive patients, who achieved early ALC
recovery at day 21 had better outcomes. We, then, analysed the impact of post-chemotherapy
ALC recovery in patients who received HSCT (66,3%) vs those who did not (33,6%) (Fig.1).
As expected, transplanted patients had globally better outcome than patients who received
only chemotherapy. In patients who received HSCT the impact of ALC recovery after
chemotherapy was minimal. In contrast, patients who had ALC <500/mm3 ALC in at least
one TP and did not undergo HSCT had the worst outcome. Interestingly, patients who
were not transplanted but had ≥500/mm3 ALC in all 4 TPs had a similar outcome as patients
who received HSCT.
Image:
Summary/Conclusion: Our study indicates ALC recovery after IC as a promising survival
predictor in AML patients, especially in those who do not undergo HSCT. In the path
that aims to investigate the complex interaction between leukemia and the immune system,
our study may provide the clinical rationale for the construction of an immunological
score to be integrated in the current disease-centered risk-classification system.
Future studies in larger cohorts addressing the impact of ALC recovery in association
with the use of novel agents and minimal residual disease status are highly warranted.
P523: MIDOSTAURIN PLUS INTENSIVE CHEMOTHERAPY IN FLT3 MUTATED AML. “REAL LIFE” DATA
VERSUS THE RATIFY STUDY
A. De La Fuente1,1,*, M. Diaz Beya2, P. Beneit3, C. Botella4, A. Fernandez Moreno5,
A. Sampol6, M. Arnan Sangerman7, A. Yeguas Bermejo8, M. D. L. L. Amigo9, J. Labrador10,
A. Garcia Guinon11, A. Garrido12, J. Serrano13, S. Vives Polo14, M. Garcia Fortes15,
M. J. Sayas16, J. M. Bergua17, M. T. Olave18, F. Vall LLovera19, J. Bargay20, M. Pereiro
Sanchez21, R. Garcia Boyero22, A. Diaz Lopez23, M. Tormo24
1Hematology, MD Anderson CC Madrid, Madrid; 2Hematology, H. U. Clinic IDIBAPS, Barcelona;
3Hematology, H. U. San Juan de Alicante; 4Hematology, H. General de Alicante, Alicante;
5Hematology, H. U. Central de Asturias, Oviedo; 6Hematology, H. U. Son Espases, Palma
de Mallorca; 7Hematology, ICO Hospitalet, L’Hospitalet de Llobregat, Barcelona; 8Hematology,
H. U. Salamanca, Salamanca; 9Hematology, H. U. Morales Meseguer, Murcia; 10Hematology,
H. U. de Burgos, Burgos; 11Hematology, H. U. Arnau de Vilanova, Lleida, Lleida; 12Hematology,
Hospital de la Santa Creu i Sant Pau. Instituto de investigación Jose Carreras, Barcelona;
13Hematology, H. U. Reina Sofia, Cordoba; 14Hematology, ICO H. Germans Trias i Pujol,
Barcelona; 15Hematology, H. Virgen de la Victoria, Malaga; 16Hematology, H. U. Doctor
Peset, Valencia; 17Hematology, H. U. san Pedro de Alcantara, Caceres; 18Hematology,
H. U. Lozano Blesa, Zaragoza; 19Hematology, H. U. Mutua Terrassa, Barcelona; 20Hematology,
H. U. Son LLatser, Palma de Mallorca; 21Hematology, C. H. U. Orense, Orense; 22Hematology,
H. G. U. de Castellon, Castellon de la Plana; 23Hematology, Fundacion MD Anderson
Madrid, Madrid; 24Hematology, 20Clinico Universitario Instituto de Investigación Sanitaria
INCLIVA, Valencia, Spain
Background: FLT3 mutation is associated with adverse prognosis in AML (ELN2017, Dohner
et al, Blood 2017) and Intensive chemotherapy (IC) has been the standard of treatment
for FLT3 mutated AML for decades. Midostaurin (Midos) has been approved in combination
with IC for FLT3-mutated AML based on an improvement in overall survival noted in
the RATIFY phase 3 trial (Stone et al, N Engl J Med 2017)
Aims: The aims of this study are to analyze safety and effectiveness of Midos plus
IC in FLT3 AML, to validate RATIFY results in a “real-world” setting and to identify
risk factors.
Methods: We carried out a retrospective multicenter study (MDA-AML-2018-06) in 27
Spanish centers. Inclusion criteria: age >18 years, FLT3-mutated AML diagnosis according
to WHO criteria and start of treatment with midostaurin in combination with IC between
June 2016 and June 2021. We evaluated the response according to 2017 ELN criteria,
toxicity according to CTCAE v4.0 and overall survival (OS) by Kaplan-Meier. Statistical
analysis was performed using SPSS program version 20.0.
Results: A total of 175patients (pt) were included (93 female), median age 53 (18-76),
of them 111were younger than 60yrs and 110pts had ECOG<2. ELN2017 stratification was
favorable in 53pt; Intermediate: 72pt; adverse in 26pt. Median WBC count 44.500/µL
(0.4-395.000). A total of 133pts had FLF3-ITD mutation and of them 74pt with ratio
≥0.5. Safety: Total Midos cycles 548 (median 2), 20 pt (11.5%) suffered QT prolongation,
26pt (14,8%) Midos interruption and 10pt (5.7%) Midos reduction. There were no deaths
related to Midos. Effectiveness: 144 (81.4%) pt achieved CR after Induction1or2, of
them 76pt were consolidated with alloSCT, 24pt received maintenance and 41pt proceed
to W&W. Median OS for the whole population was not reached and the 24months OS was
68%. In our experience ELN2917 classification and ECOG<2 identify groups with differences
in OS (p0.019 and p0.04 respectively). Age <60vs≥60 resulted in no differences for
OS. We observed significant differences for maintenance versus W&W (p=0.001) in favorable
and intermediate risk patients. Comparing maintenance vs alloSCT we observe no differences
in the Intermediate ELN2017 group.
Image:
Summary/Conclusion: Our experience confirms safety and effectiveness of Midos plus
IC as first line in FLT3 AML patients with similar 2yOS as the previous reported in
the RATIFY trial. Interesting that in our experience age 60 and above resulted in
no differences for OS.
P524: PHASE 1B/2 STUDY ON SAFETY, PK, PD, AND PRELIMINARY EFFICACY OF THE SELECTIVE
SYK INHIBITOR LANRAPLENIB IN COMBINATION WITH THE FLT3 INHIBITOR GILTERITINIB, IN
FLT3-MUTATED R/R AML (KB-LANRA 1001)
E. M. Stein1,*, A. Patel2, L. C. Michaelis3, G. Schiller4, R. Swords5, L. A. Carvajal6,
G. Bray6, J. DiMartino6, M. Y. Levy7
1Memorial Sloan Kettering Cancer Center, New York; 2The University of Chicago Medical
Center, Chicago; 3Medical College of Wisconsin, Milwaukee; 4University of California
Los Angeles (UCLA), Los Angeles; 5Oregon Health and Science University, Portland;
6Kronos Bio, Inc., San Mateo; 7Texas Oncology - Baylor Charles A. Sammons Cancer Center,
Dallas, United States of America
Background: Spleen tyrosine kinase (SYK) is a key driver of lymphoid and myeloid cell
signaling pathways and has been implicated in the pathogenesis of acute myeloid leukemias
(AML) defined by dysregulated expression of HOXA9 and MEIS1 transcription factors.
SYK also cooperates with internal tandem duplication (ITD)-mutated FMS-like tyrosine
kinase 3 (FLT3) to drive leukemogenesis. Combined pharmacologic inhibition of SYK
and FLT3 results in robust antileukemic effects in preclinical models of FLT3 ITD-driven
AML. Lanraplenib (LANRA) is an oral, selective SYK inhibitor. Studies demonstrate
dose-dependent reductions in viability of leukemic cells ex vivo from newly diagnosed
FLT3 ITD-mutated AML patients (pts) as well as additive reductions in viability when
combined with the selective FLT3 inhibitor, gilteritinib. Once daily (QD) LANRA was
well-tolerated at doses up to 50mg for 7 days and 30mg for up to 12 weeks in healthy
volunteers and pts with autoimmune disorders.
Aims: The KB-LANRA 1001 trial will investigate the safety, pharmacokinetics (PK) and
pharmacodynamics (PD) of LANRA when combined with gilteritinib in relapsed or refractory
pts with FLT3-mutated AML. We will also evaluate the rate of complete responses (CR)
and CR with partial hematologic recovery (CRh), duration of response (DoR), event-free
(EFS) and overall survival (OS).
Methods: KB-LANRA 1001 is a global, multicenter trial of LANRA in combination with
standard dose gilteritinib (120mg QD) in FLT3-mutated AML pts aged ≥18 with at least
1 prior therapy. Those who have been treated with midostaurin or other multikinase
inhibitors are eligible, though pts who failed to achieve at least a partial response
or who relapsed following prior exposure to gilteritinib or other next-generation
FLT3 inhibitor monotherapy are excluded. Both medications will be administered daily
in sequential, 28-day cycles until progression/relapse, intolerance, or failure to
achieve at least a partial remission after 6 cycles. The Phase 1b component will define
the maximum tolerated and/or recommended Phase 2 dose (MTD/RP2D) of LANRA when added
to gilteritinib in accordance with a 3 + 3 dose escalation design. PK and PD parameters
will be characterized for LANRA monotherapy and when co-administered with gilteritinib.
Four LANRA doses are planned for evaluation (20, 40, 60 and 90mg). All decisions regarding
dose escalation including declaration of the MTD will be made by a dose-escalation
committee based on the prospectively defined definition of dose-limiting toxicities
and other safety metrics. The Phase 2 component will further evaluate safety, PK,
PD and preliminary efficacy of the combination at the LANRA RP2D. For both trial components,
response assessments will be conducted on Day 1 of Cycles 2 and 3 and every 3 cycles
thereafter until 2 consecutive assessments indicate CR or CRh. A 2-stage design will
be employed to test the null hypothesis that the rate of CR/CRh for the combination
is ≤23%, as reported for gilteritinib monotherapy, versus the alternative hypothesis
of CR/CRh ≥46%, among pts treated at the LANRA RP2D. All pts will be followed for
relapse, EFS and OS. Key exploratory endpoints will be assessments of measurable residual
disease among pts who achieve CR/CRh, correlations with selected baseline biomarkers
that may predict efficacy outcomes and assessment of LANRA PD properties (including
target engagement) when administered alone and in combination with gilteritinib.
Results: Trial in progress.
Summary/Conclusion: Accrual is ongoing.
P525: PHASE 3, RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED STUDY OF THE EFFICACY
AND SAFETY OF ENTOSPLETINIB ADDED TO INTENSIVE INDUCTION AND CONSOLIDATION CHEMOTHERAPY
IN NEWLY DIAGNOSED NPM1-MUTATED AML
J. C. Byrd1,*, J. E. Cortes2, M. D. Minden3, T. Oellerich4, E. M. Stein5, J. Elder6,
P. Kumar7, G. Bray7, J. DiMartino7, W. Stock8
1University of Cincinnati, Cincinnati; 2Georgia Cancer Center at Augusta University,
Augusta, United States of America; 3Princess Margaret Cancer Centre, Toronto, Canada;
4Frankfurt Cancer Institute, Frankfurt am Main, Germany; 5Memorial Sloan Kettering
Cancer Center, New York; 6PharPoint Research, Inc., Durham; 7Kronos Bio, Inc., San
Mateo; 8University of Chicago Medicine, Chicago, United States of America
Background: Spleen tyrosine kinase (SYK) is a component of both lymphoid and myeloid
cell signaling pathways and is implicated in the pathogenesis of a subset of acute
myeloid leukemia (AML) defined by dysregulated expression of HOXA9 and MEIS1 transcription
factors. Entospletinib (ENTO) is an oral, selective SYK inhibitor that is acceptably
tolerated when administered with intensive induction and consolidation in newly diagnosed
AML patients (pts). In a Phase 2 study, following induction with cytarabine and daunorubicin
(7 + 3) plus ENTO, higher rates of complete response (CR) or CR with incomplete hematologic
recovery (CRi) occurred in pts with rearrangements of the KMT2A gene (MLL-r) and mutations
of the nucleophosmin 1 (NPM1) gene, both of which are associated with aberrant expression
of HOXA9 and MEIS1, compared to pts without these mutations. In an exploratory analysis,
pts with HOXA9/MEIS1 expression levels above the median experienced superior overall
survival (OS) compared to pts with expression levels below the median.
Aims: We hypothesize that the addition of ENTO to intensive induction/consolidation
in newly diagnosed pts with NPM1-mutated AML will improve the CR rate without evidence
of measurable residual disease (MRD-negative CR) post-induction and duration of event-free
survival (EFS).
Methods: AGILITY is a global, multi-center, double-blind, placebo-controlled trial
of ENTO in combination with cytarabine plus daunorubicin or idarubicin induction (7 + 3)
and age-adjusted high-dose cytarabine (HiDAC) consolidation in newly diagnosed AML
pts aged 18–74 who are candidates for intensive induction and harbor a documented
NPM1 mutation. Pts with co-mutated FLT3 are excluded. Pts will be stratified based
on age (<60 vs ≥60 years) and anthracycline administered during induction (daunorubicin
vs idarubicin). Approximately 180 pts will be randomized to receive 7 + 3 induction
and HiDAC consolidation with ENTO (400mg orally twice daily) or with placebo. Pts
with <5% leukemic blasts after induction cycle 1 will proceed to the first cycle of
consolidation; pts with ≥5% residual blasts will undergo a second induction cycle.
Pts who do not achieve CR after the last cycle of induction plus ENTO or placebo will
be designated as induction treatment failures (ITF) for purposes of EFS estimation.
Pts who achieve or remain in CR after 2 chemotherapy cycles (either 2 induction cycles
or 1 induction and 1 consolidation cycle) will be evaluated for MRD in bone marrow
based on enumeration of mutant NPM1 alleles using a molecular assay. Pts may receive
≤3 cycles of consolidation with HiDAC and ENTO or placebo per their original randomized
treatment assignment beyond chemotherapy cycle 2. All pts will be followed for relapse
and survival. The primary endpoint is the rate of MRD-negative CR (<0.01% mutant NPM1
alleles). A key secondary endpoint is EFS, defined as time from randomization to earliest
occurrence of ITF, relapse from CR, or death from any cause. OS will be evaluated.
Key exploratory endpoints will be the correlation between recurring genomic mutations
and response or progression and longitudinal assessment of peripheral blood for detection
of NPM1-m alleles among pts who achieve MRD-negative CR post-induction.
Results: Trial in progress.
Summary/Conclusion: Accrual is ongoing.
P526: REAL-WORLD COMPARISON OF DIFFERENT TREATMENT MODALITIES FOR FAVORABLE RISK ACUTE
MYELOID LEUKEMIA: STANDARD DOSE ANTHRACYCLINE VS HIGH DOSE ANTHRACYCLINE VS ADDITION
OF GEMTUZUMAB OZOGAMICIN
J. Mort1, B. Brewer2, B. Mautner1, S. Dibenedetto1, E. Pierce1, E. Morse1, L. Wells1,
F. Ghamsari1, A. Morris1, S. Clark3, J. Reid3, I. Patel4, C. Jackson5, M. Perciavalle6,
B. Yelvington6, R. Miller6, K. Abernathy6, H. Wolfe7, S. Locke7, J. Zeidner3, D. Reed5,
V. Duong4, B. Dholaria6, T. LeBlanc7, M. Keng1, B. Horton2, F. El Chaer1,*
1Hematology and Oncology; 2Public Health Sciences, Division of Translational Research
and Applied Statistics, University of Virginia, Charlottesville; 3Hematology and Oncology,
University of North Carolina, Chapel Hill; 4Hematology and Oncology, University of
Maryland Medical Center, Baltimore; 5Hematology and Oncology, Wake Forest Baptist
Medical Comprehensive Cancer Center, Winston-Salem; 6Hematology and Oncology, Vanderbilt
University Medical Center, Nashville; 7Hematology and Oncology, Duke Cancer Institute,
Durham, United States of America
Background: The standard remission induction regimen for medically fit patients with
acute myeloid leukemia (AML) consists of a backbone of cytarabine & an anthracycline
(“7 + 3” therapy). However, the choice & dose of the anthracycline varies between
institutions & practitioner preference, particularly in AML with favorable risk cytogenetics.
Gemtuzumab ozogamicin (GO) may improve outcomes in this patient population but is
associated with increased toxicities.
Aims: We aimed to compare the outcomes of 3 cohorts of newly diagnosed AML with favorable
cytogenetics who received induction 7 + 3 induction therapy: 1. Standard dose (45
or 60 mg/m2 daunorubicin) (SD); 2. Higher dose (90 mg/m2 daunorubicin or 12 mg/m2
idarubicin) (HD) of anthracycline, 3. 7 + 3 with fractionated GO.
Methods: We performed a multicenter retrospective analysis of medically fit patients
≥ 18 yo with favorable risk AML determined using ELN 2017 guidelines from 2015-2020
treated across 6 US academic institutions. Categorical variables were summarized using
means and standard deviations. The former was compared using Fisher’s exact test;
the latter was compared using either the Wilcoxon text or the Kruskal-Wallis as appropriate.
One way ANOVA test was used to compare continuous variables. Kaplan-Meier curves were
generated to provide visual summaries of survival and time-to-event data.
Results: Our study included 189 treated patients who met eligibility criteria with
a median follow up of 2 years. Patients’ characteristics and outcomes are summarized
in Table 1. Sixty-nine (37%) received induction with SD, 100 (53%) received HD, and
20 (11%) received GO. Patients in the SD and HD group were younger (median age 50
and 52 yo, respectively) and the majority (>94%) had de novo AML. The rates of composite
complete remission in the SD, HD, and GO groups were 66.7%, 70% and 75%, respectively
(p=0.761). RFS and OS were similar between all groups (p=0.4 and 0.3, respectively).
The rate and duration of cytopenias at day 40 after induction, major bleeding, infections,
admission to the intensive care unit, new onset cardiomyopathy (ejection fraction<50%),
liver dysfunction and acute renal injury were similar among all groups. No patients
developed veno-occlusive disease.
Table 1:
Participant Characteristics and Outcomes by Treatment Regimen
GON = 20
HDN = 100
SDN = 69
P-value
Age in years (standard deviation)
57.75 (9.32)
49.68 (13.67)
51.55 (13.83)
0.038
de novo AML, N (%)
19 (95.0)
94 (94.0)
65 (94.2)
1.000
Disease status after induction
0.812
CR, N (%)
14 (70.0)
66 (66.0)
40 (58.0)
CRi, N (%)
0 (0.0)
2 (2.0)
2 (2.9)
Persistent, N (%)
1 (5.0)
4 (4.0)
2 (2.9)
Composite CR, N (%)
15 (75.0)
70 (70.0)
46 (66.7)
0.761
Admission to ICU, N (%)
4 (20.0)
17 (17.0)
11 (15.9)
0.969
Infections, N (%)
17 (85.0)
74 (74.0)
62 (89.9)
0.082
Major bleeding, N (%)
3 (15.0)
6 (6.0)
4 (5.8)
0.299
VOD, N (%)
0 (0.0)
0 (0.0)
0 (0.0)
Transaminitis, N (%)
4 (20.0)
19 (19.0)
19 (27.5)
0.538
Acute renal injury, N (%)
3 (15.0)
19 (19.0)
15 (21.7)
0.962
Cardiomyopathy, N (%)
4 (20.0)
8 (8.0)
5 (7.2)
0.523
Cytopenias at D40 after induction, N (%)
0 (0.0)
7 (7.0)
6 (8.7)
0.542
Image:
Summary/Conclusion: Our analysis demonstrated no differences in RFS and OS in patients
with AML treated with either GO, HD or SD. In addition, the rates of complications
secondary to chemotherapy were similar among all groups. However, a measurable residual
disease analysis is ongoing for further characterization of disease response.
P527: PHASE 1 TRIAL OF PEGCRISANTASPASE IN COMBINATION WITH VENETOCLAX IN ADULTS WITH
RELAPSED OR REFRACTORY ACUTE MYELOID LEUKEMIA (R/R AML) - SAFETY, EFFICACY AND PK/PD
IN THE FIRST TWO COHORTS
Y. Liu1, O. M. Bah1, D. Bollino1, K. Caprinolo1, J. Zarrabi1, S. Philip1, R. G. Lapidus1,
E. T. Strovel2, Z. N. Singh3, M. E. Kallen3, Y. Ning3, R. Koka3, S. Niyongere1, V.
H. Duong1, M. R. Baer1, M. Graveno1, A. Emadi1,*
1University of Maryland Greenebaum Comprehensive Cancer Center; 2Pediatrics; 3Pathology,
University of Maryland School of Medicine, Baltimore, United States of America
Background: Glutamine depletion induced by Erwinia asparaginases including pegcrisantaspase
(PegC) downregulates the mTOR/p70S6K pathway and inhibits 4EBP1 phosphorylation. We
recently found that the combination of PegC with the Bcl-2 inhibitor venetoclax (Ven)
has strong in vitro and in vivo activity in poor-prognosis AML, mediated by enhancement
of eIF4E-4EBP1 interaction on the cap-binding complex and decreasing protein synthesis.
We hypothesized that Ven-PegC treatment will be safe and effective in the treatment
of R/R AML. Here we report the results from the first two cohorts of a phase 1 trial
of Ven-PegC in adults with R/R AML.
Aims: The primary objective is to estimate the maximum tolerated dose and to determine
the regimen-limiting toxicities (RLT) of Ven-PegC. The exploratory endpoints include
plasma amino acids levels 1- and 2-weeks post-PegC administration.
Methods: This is an open-label phase 1b clinical trial (NCT04666649) of daily oral
Ven in combination with biweekly out-patient IV PegC in 28-day cycles that will be
administered to 4 cohorts (Table 1A) based on a standard 3 + 3 design.
Results: Eight patients (pts) were enrolled into the first two cohorts; 7 received
≥ 1 dose of PegC and one dropped out before receiving study drugs. Pts’ characteristics
are shown in Table 1B. Median age was 66 (59-76) years; two (29%) were female; six
(86%) had adverse-risk AML by ELN classification; and four (57%) had complex karyotypes.
Pharmacokinetic (PK) and pharmacodynamic (PD) data are available for 5 pts; all pts
achieved a nadir plasma asparaginase activity > 0.1 IU/mL during cycle 1 (3/3 in cohort
1 within 3 weeks and 2/2 in cohort 2 within 10 days). Mean plasma amino acid levels
(µmol/L) at baseline were: asparagine (Asn) 29 (17-38), glutamine (Gln) 421 (384-465),
glutamate (Glu) 59 (22-103), serine (Ser) 66 (49-80) and glycine (Gly) 203 (166-207).
All pts achieved an Asn level of 0 by day 7; Asn remained undetectable throughout
the study. Plasma Gln (µmol/L) reduction from baseline occurred in 4/5 (80%) pts within
cycle 1: pt4 423→273 (Day15), pt5 446→86 (Day28), pt6 467→216 (Day22), pt7 388→156
(Day8), pt10 420→395 (Day10). Plasma Glu levels increased in all pts, indicating conversion
of Gln to Glu by PegC. Interestingly, plasma levels of Ser increased 1.5-2-fold at
the nadir of plasma Gln (Figure 1A-C, maximum amino acid changes after Ven-PegC).
Seven pts from cohorts 1 (4 pts) and 2 (3 pts) who received at least one dose of Ven-PegC
were included in analysis of toxicity and efficacy, with data cutoff February 26,
2022 (Table 1B). No RLT has been observed. In cohort 1, pt2 died before completing
cycle 1 due to septic shock and delay in seeking medical care, pt4 died of AML progression.
In cohort 2, pt7 had progressive disease after cycle 1, pt9 opted for comfort care
before evaluation of response, and pt10 died of AML progression,
In cohort 1, pt6 achieved measurable residual disease (MRD)-negative complete remission
with partial hematologic recovery (CRh) and proceeded to allogeneic hematopoietic
stem cell transplant (allo-HSCT) after 3 cycles of Ven-PegC, and bone marrow blasts
in pt5 decreased from 78% to 9% after cycle 1. These two pts had the lowest plasma
Gln levels (103 [pt6, cycle2 day29] and 86 [pt5, cycle1 day28]).
Image:
Summary/Conclusion: No RLT has been observed following Ven-PegC in cohorts 1 or 2.
The Ven-PegC combination at the lowest dose of PegC administered, produced an MRD
negative CRh in one patient and a transient substantial reduction in bone marrow blast
count in another, both associated with decreases in plasma Gln levels. The study is
ongoing.
P528: A MIRNA SIGNATURE RELATED TO STEMNESS IDENTIFIES HIGH-RISK PATIENTS IN PEDIATRIC
ACUTE MYELOID LEUKEMIA
E. Esperanza Cebollada1,2,*, S. Gomez Gonzalez3, N. Vega García1,2, C. Vicente Garcés1,2,
M. Torrebadell1,2,4, S. Rives2,4,5, J. L. Dapena2,5, A. Català2,4,5, M. Camós1,2,4
1Hematology Laboratory, Hospital Sant Joan de Déu; 2Leukaemia and other Pediatric
Hemopathies. Developmental Tumors Biology Group; 3Developmental Tumor Biology Laboratory,
Institut de Recerca Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona;
4Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto
de Salud Carlos III, Madrid; 5Pediatric Hematology and Oncology Department, Hospital
Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
Background: MicroRNAs (miRNAs) regulate gene expression and may function as tumor
suppressors or cancer promoters. MicroRNAs regulating hematopoietic stem cells (HSC)
could be prognostic biomarkers in leukemia, particularly in KMT2A-rearranged (KMT2A+)
patients, in whom few mutations have been described.
Aims: To analyze the role of miRNAs involved in HSC self-renewal and stemness pathways
(NOTCH, WNT/beta-catenin, HOX, and FLT3) in a pediatric acute lymphoblastic leukemia
(ALL) and myeloid acute leukemia (AML) cohort of patients.
Methods: We analyzed by quantitative PCR the expression of 89 selected miRNAs. To
calculate miRNAs ∆∆CT expression differences between paired subgroups we used the
lmFit function of the Limma R package, and the false discovery rate method to adjust
the p-values with a 0.05 cut-off.
Results: We studied 110 patients (38% female, median age 6.1 years (range 0-17.4)),
including 70 ALL, 39 AML, and one undifferentiated leukemia. Our series was primed
with KMT2A+ cases of all lineages (n=28). Globally, in the unsupervised hierarchical
cluster analysis (HCA), patients were separated by lineage. Based on the sub-branching,
we tried to correlate the clusters with clinic-biological variables. We found no association
with age, sex, central nervous system infiltration, or leukocyte count. However, Kaplan-Meier
analysis revealed that some clusters had significantly different event-free survival
(EFS) (p=0.014): poor (clusters P1 and P4), good (P2), and intermediate (P3). Of note,
clusters with good and poor survival branched off again according to lineage (P1 included
most of AML cases and P4 most of ALL patients). We then studied survival according
to lineage. Within ALL patients, we saw a group with a non-significant worse outcome
(P4ALL), as compared with the remaining patients (P2ALL and P3ALL). In contrast, in
AML patients, the HCA distinguished a group of 14 patients (36%, P1AML) with significantly
worse outcome: OS at 5 years of 64.3%±12.8 vs. the remaining AML patients, all showing
excellent survival (grouped in P2AML, 90.5%±6.4), p=0.028. Notably, differences in
EFS at 5 years were even higher: 43%±13.2 (P1AML) vs. 86.5%±7.2 (P2AML), p=0.003.
P1AML included cases from all molecular categories (RUNX1::RUNX1T1, CBFB::MYH11, KMT2A+,
and cases without recurrent genetic alterations). Of note, no PML::RARA cases fell
in this cluster. We identified 24 miRNAs (23 underexpressed and 1 overexpressed) differentially
expressed in P1AML patients. The 24 miRNAs signature included underexpressed miRNAs
known to act as tumor suppressors regulating cell proliferation or apoptosis (hsa-miR-223-3p,
hsa-miR-193a-3p, hsa-miR-181a-5p, hsa-miR-181b-5p, hsa-miR-181c-5p, hsa-miR-708-5p,
hsa-miR-34b-5p, and hsa-miR-22-3p). The only miRNA overexpressed, hsa-miR-9-5p, promotes
proliferation and inhibits apoptosis, functioning as oncomiR in KMT2A-rearranged AML
cases. The remaining miRNAs (hsa-miR-199b-5p, hsa-miR-222-5p, hsa-miR-373-3p, hsa-miR-195-5p,
hsa-miR-151a-5p and hsa-miR-1290, hsa-miR-20b-5p and hsa-miR-17-5p, hsa-miR-24-3p,
hsa-miR-128-3p, hsa-miR-21-5p, hsa-miR-331-5p, hsa-miR-30b-5p, hsa-let-7g, and hsa-let-7i-5p),
among their pleiotropic functions, regulate cellular stemness and proliferation. Our
results suggests that within each molecular category, a miRNA signature could distinguish
those patients with activated stemness and proliferation pathways and a more aggressive
leukemia.
Image:
Summary/Conclusion: We obtained a distinctive signature of 24 miRNAs capable of distinguishing
pediatric AML patients with excellent or poor outcome.
P529: FIRST EUROPEAN REAL-WORLD EVIDENCE PROSPECTIVE REGISTRY OF FIRST-LINE ADULT
PATIENTS WITH BLASTIC PLASMACYTOID DENDRITIC CELL NEOPLASM TREATED WITH FIRST-IN-CLASS
CD123-TARGETED THERAPY TAGRAXOFUSP
U. Platzbecker1,*, E. Angelucci2, P. Montesinos3, R. M. Lemoli4, A. Spyridonidis5,
J. Casariego6, T. I. Mughal7,8, M. Mohty9
1Medical Clinic and Policlinic of Hematology, Cell Therapy and Hemostaseology, University
Hospital Leipzig, Leipzig, Germany; 2Hematology and Cellular Therapy Unit, IRCCS Ospedale
Policlinico San Martino, Genoa, Italy; 3Department of Hematology, Hospital Universitari
i Politècnic La Fe, València, Spain; 4Clinic of Hematology, Department of Internal
Medicine, IRCCS San Martino Hospital, University of Genoa, Genoa, Italy; 5Department
of Internal Medicine, BMT Unit, University of Patras, Patras, Greece; 6Aldebaran Research
& Development SL, Madrid, Spain; 7Division of Hematology-Oncology, Tufts University
School of Medicine, Boston; 8Stemline Therapeutics, New York, United States of America;
9Department of Hematology and Cellular Therapy, Hôpital Saint-Antoine, Sorbonne University,
Paris, France
Background: Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare hematologic
malignancy derived from plasmacytoid dendritic cells that overexpress interleukin-3
(IL-3) receptor alpha (CD123). It is a highly aggressive disease characterized by
skin and bone marrow involvement and is associated with a poor prognosis (median overall
survival 8–14 months). Tagraxofusp (TAG, SL-401) is a first-in-class CD123-targeted
therapy comprising human IL-3 fused to a truncated diphtheria toxin payload. It is
currently the only approved first-line (1L) treatment for adult patients (pts) with
BPDCN in Europe. As of July 2021, 65 pts with BPDCN have received TAG in a clinical
setting outside the US as part of an expanded access program. Collecting real-world
clinical data from TAG is important to increase understanding of clinical outcomes,
especially in pts commonly underrepresented in clinical trials. We describe the first
real-world evidence prospective registry in pts with BPDCN receiving TAG. It has been
designed to meet requirements outlined in the marketing authorization of the European
Commission for further evaluation of the efficacy and safety of TAG in pts with 1L
or relapsed/refractory BPDCN.
Aims: To investigate effectiveness and safety of TAG in pts with 1L BPDCN.
Methods: This is a noninterventional, single-arm, post-authorization study (EudraCT:
2021-001684-24) in adult pts with BPDCN prescribed TAG under real-world routine clinical
practice conditions (12 mcg/kg intravenously [IV] once daily on days 1–5 of a 21-day
cycle, per the summary of product characteristics recommendation). A minimum of 80
pts will be included in approximately 40–55 sites in Europe, estimated over 4 years.
Eligible pts have a diagnosis of BPDCN and are to be treated (or have recently started
treatment) with TAG monotherapy as 1L treatment, per physician’s decision.
Primary endpoints are rate of complete response (CR), defined as CR + clinical CR
(CR with residual skin abnormality not indicative of active disease) after 3 months
of treatment, and safety of TAG including the incidence and severity of capillary
leak syndrome (CLS). Secondary endpoints include number of pts bridging to SCT, progression-free
survival, overall survival, best overall response, duration of response, TAG dose
interruptions/administration of IV albumin supplementation in pts with CLS or CLS
symptoms, as well as safety and incidence and severity of adverse events of special
interest (which include CLS and hepatic, renal, and cardiac events).
Quarterly data collection is anticipated, as well as collection at screening, enrollment,
early discontinuation, and study end (Figure). Safety data collection will end 18
months after the last enrolled pt’s first visit (LPFV). Interim analyses will be performed
annually; an effectiveness interim analysis is scheduled at 20 months post-LPFV. Analyses
will be performed using descriptive statistics. Survival data will be summarized using
the Kaplan-Meier method. Subgroup analyses for effectiveness and safety may be performed,
on the basis of gender, age, or baseline Eastern Cooperative Oncology Group performance
status. Enrollment is planned to start by June 2022.
Taking the registry as the basis, upon study finalization a complementary comparative
analysis of effectiveness and safety data from the TAG registry with a retrospective
clinical cohort will be undertaken using appropriate methodology, such as propensity
score matching or inverse probability treatment weighting.
Results: This is a TiP and there are no results at this time.
Image:
Summary/Conclusion: This is a TiP and there are no results at this time.
P530: TAGRAXOFUSP IN BLASTIC PLASMACYTOID DENDRITIC CELL NEOPLASM WITH/WITHOUT CENTRAL
NERVOUS SYSTEM INVOLVEMENT AND INTRATHECAL CHEMOTHERAPY AS PRIMARY TREATMENT OR PROPHYLAXIS:
AN ITALIAN EXPERIENCE.
G. Rivoli1,*, G. Beltrami1, A. Raiola1, A. Dominietto1, M. Riggi2, E. Angelucci1
1Hematology and cellular therapy unit, IRCCS Ospedale Policlinico San Martino, Genova,
Italy; 2Stemline Therapeutics Switzerland GmbH, Zug, Switzerland
Background: The precise frequency of central nervous system involvement (CNS+) in
patients (pts) with blastic plasmacytoid dendritic cell neoplasm (BPDCN) is currently
unknown. Recent small series of clinical experience suggest the incidence to be about
10% at baseline and 30% at first relapse. CNS is a ”sanctuary” for any systemic treatment
and can lead to disease recurrence even after the achievement of a complete response
(CR) at the medullary and skin level. Thus, it is essential to ensure clearance (or
prophylaxis) of the disease including the meningeal compartment. Intrathecal (IT)
chemotherapy is a valid complementary tool aimed at eradication of the disease, with
modest systemic side effects; it is currently indicated as both prophylaxis and primary
treatment. Combining IT chemotherapy with systemic treatment has made it possible
to obtain long remissions in pts with BPDCN even in the presence of CNS+. Tagraxofusp
(TAG), a first-in-class CD123-targeted therapy, was approved by the European Medicines
Agency in 2021 for treatment of BPDCN in first-line adult pts.
Aims: Data from 5 pts with BPDCN with/without CNS+ treated with TAG were retrospectively
collected in our center in Geneva. The aim was to evaluate if CNS treatment/prophylaxis
impacts prognosis and efficacy of systemic treatment with TAG.
Methods: A diagnosis of BPDCN was confirmed by hematopathology with biomarkers, including
CD123, CD4, and CD56, and presence of CNS involvement by the characteristic morphology
and “8-color flow cytometry” for CD123. Pts received TAG intravenous infusions at
12 mcg/kg once daily on days 1–5 of a 21-day cycle. TAG was repeated for 1–4 cycles,
depending on the level of response and potential bridge to hematopoietic stem cell
transplant (HSCT). Hospitalization was mandatory for the first cycle; the following
cycles were administered in an outpatient setting if no severe complications occurred
during the first cycle. IT chemotherapy (methotrexate 12.5 mg, dexamethasone 4 mg,
and cytarabine 50 mg) was administered at each cycle at the same doses until negative
cerebrospinal fluid (CSF) was observed in CNS+ pts. After 4 treatment cycles, response
was evaluated, including CSF examination.
Results: TAG was administered in 5 first-line pts with BPDCN; 3 were CNS+ (1 clinically
evident case and 2 with cytofluorometric positivity only). All pts received IT chemotherapy,
both as a primary-intention treatment in the 3 CNS+ pts, and as prophylaxis in the
2 CNS-negative pts. Table 1 reports the main pt characteristics. Pts 2, 3, and 5 with
CNS+ achieved clearance of the disease and none reported CNS relapse. In pts receiving
CNS prophylaxis, pt 1 received TAG plus HSCT and is in CR after 29 months; pt 4 received
high-dose chemotherapy, achieved a good partial remission after TAG, and remains alive
after 4 months with HSCT shortly planned. No pts developed CNS adverse events following
IT chemotherapy.
Image:
Summary/Conclusion: These preliminary results confirm the feasibility of IT chemotherapy
with systemic TAG. Baseline disease and CNS involvement did not appear to predispose
pts to different efficacy results or treatment-related adverse events. IT chemotherapy
effectively cleared and controlled CNS involvement. IT prophylaxis should be considered
in pts with BPDCN who receive CD123-targeted therapies.
P531: AN OBSERVATIONAL, MULTICENTER, RETROSPECTIVE ANALYSIS OF PATIENTS WITH BLASTIC
PLASMACYTOID DENDRITIC CELL NEOPLASM TREATED WITH TAGRAXOFUSP IN THE EUROPEAN EXPANDED
ACCESS PROGRAM
M. Herling1,*, E. Deconinck2, M. Anant3, D. Manteigas4, M. Riggi3, E. Angelucci5
1Department of Hematology, Cell Therapy and Hemostaseology, University Hospital Leipzig,
Leipzig, Germany; 2Regional University Hospital of Besançon, Besançon, France; 3Stemline
Therapeutics Switzerland GmbH, Zug, Switzerland; 4Cmed, West Sussex, United Kingdom;
5Hematology and Cellular Therapy Unit, IRCCS Ospedale Policlinico San Martino, Genoa,
Italy
Background: Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare aggressive
hematologic malignancy, with poor prognosis (median age 67 yr). It is characterized
by clonal expansion of plasmacytoid dendritic tumor cells expressing specific markers
including the interleukin-3 receptor alpha (CD123). Primary sites are skin and bone
marrow, followed by peripheral blood, lymph nodes, viscera, and central nervous system.
Tagraxofusp (TAG), a CD123-targeted therapy, was approved by the European Medicines
Agency on 07.01.2021. In 08.2019, a Global Expanded Access Program (EAP) was implemented
to provide access to patients (pts) prior to regulatory authorization of TAG in real-world
practice.
Aims: We conducted a European multicenter non-interventional, retrospective analysis
of BPDCN pts treated with TAG. Main objectives were rates of complete response, and
incidence and severity of capillary leak syndrome (CLS). Secondary outcomes included
rate of pts bridged to stem cell transplantation, progression-free survival, and overall
survival, safety of TAG measured by the incidence and severity of adverse events (AEs),
and number of TAG doses administered in each cycle.
Methods: The main inclusion criterion was diagnosis of BPDCN, confirmed by hematopathology
with established marker panels (including CD123). The physician who signed the supply
form of TAG in each participating center informed the pts on their treatment. Training
of the physicians, nurses, and pharmacists was mandatory before delivering the treatment.
The analysis included all pts enrolled in the European EAP from 08.2019 to 12.2021.
Pts received TAG intravenous infusions, at 12 mcg/kg once daily on days 1–5 (up to
day 10 allowed) of a 21-day cycle. Hospitalization was required only for the first
cycle (subsequent cycles were allowed to be administered in an outpatient setting).
Results: Overall, 76 adult (median age 64 yr, range 21–85 yr) and 4 pediatric pts
(1, 4, 14, and 16 yr) were included across 57 European centers: Germany 18 centers,
France 17, Italy 9, Switzerland 5, United Kingdom 4, Spain 3, and Austria 1. Most
pts were male (78%), representing real-world distribution. Sixty-three pts received
first-line TAG and 17 pts as second or further line of treatment. The median number
of cycles (based on the number of treatments delivered for the TAG EAP supply of each
21-day cycle) was 2.5 (range 1–8) in first line and 2.6 (range 1–13) cycles in second-line
and further pts, respectively. No deaths due to CLS were reported. The analysis is
still ongoing at the time of abstract finalization; complementary data on safety,
efficacy, number of pts transplanted, and time-related parameters will be reported
at the meeting.
Summary/Conclusion: The is the largest retrospective analysis of real-world clinical
practice outside of a clinical trial in BPDCN pts treated with TAG. The EAP was carefully
followed in all 57 centers with initial training provided to the multidisciplinary
teams before treatment initiation. This is thought to have positively affected prevention
and management of CLS and other grade 3–4 AEs; no death related to CLS occurred. The
preliminary results confirm the feasibility and safety of TAG, allowing the administration
also in elderly pts, with manageable safety. In fact, AEs mainly occurred in cycle
1 in an inpatient setting, with similar rates and severity in older vs younger pts.
Baseline characteristics did not appear to predispose pts to different treatment-related
AEs.
P532: ACUTE MYELOID LEUKEMIA (AML): UNICENTRIC REPORT ON 1029 PATIENTS DIAGNOSED IN
TERTIAL REFERAL CENTER
A. Fricke1,*, K. Nachtkamp1, K. Döhner2, B. Hildebrandt3, B. Betz3, M. Rudelius4,
M. Seidl5, C. Zahner1, G. Kobbe1, A. Kündgen1, P. S. Jäger1, B.-N. Baermann1, U. Germing1
1Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf,
Düsseldorf; 2Department of Internal Medicine III, University Hospital Ulm, Ulm; 3Institute
of Human Genetics, University Düsseldorf, Düsseldorf; 4Pathological Institute, Ludwig-Maximilians-University
Hospital Munich, Munich; 5Pathological Institute, University Hospital Düsseldorf,
Düsseldorf, Germany
Background: Since 2016 patients with AML can be diagnosed according to the WHO classification
proposals. The ELN genetic risk classification is a tool for treatment planning.
Aims: In order to validate the WHO and the ELN classification, we collected data from
1029 patients diagnosed and / or treated in our institution from 2005 to 2021.
Methods: Patients were followed up until December 2021. Diagnoses were based on cytomorphology,
histomorphology, flow cytometry, karyotyping, and molecular genetics performed at
the University of Düsseldorf and partly at the University of Ulm. Treatment intensity
was defined as follows: no treatment at all, best supportive care (BSC) (including
low dose Ara-C and Hydroxyurea), Hypomethylating agents (HMA), induction and consolidation,
or allogeneic hematopoietic stem cell transplantation (HSCT).
Results: Median age at diagnosis was 64 years (ys) (19 – 94). 46% were females. There
were 270 patients with recurrent genetic abnormalities (26.5%), 346 with AML-MRC (34%),
121 with tAML (11.9%), 243 with AML-NOS (23.9%), 3 with myeloid sarcoma (0.3%), 4
with blastic plasmacytoid dendritic neoplasm (0.4%), 20 with biphenotypic AML (2%),
and in 22 patients the classification was not possible.
17.7% of patients belong to the good risk category of ELN classification, 21.6% to
the intermediate risk category and 30% to the adverse risk group. In 30.7%, the classification
was not possible due to missing data.
274 patients were treated with HSCT as most intensive therapy (28%), 348 patients
with induction and consolidation (35.6%). 209 patients received HMA (21.4%). 127 patients
were treated with BSC (13%) and in 20 patients (2%) no therapy was administered. In
51 patients, AML was diagnosed at our laboratory, but treatment took place outside
of our department.
The median survival time of the entire group was 18 months with 39% of the patients
alive by the end of 2021. Median survival was 21 months in females as compared to
16 months in males (p = 0.04). Patients aged over 65 ys had a median survival of 7
months as compared to 134 months in patients aged less than 65 ys (p < 0.0005). There
was no difference in overall survival in patients younger than 50 ys of age.
Patients with recurrent genetic abnormalities did not reach median survival whereas
median survival of patients with AML-MRC, tAML, and NOS was 11, 12, and 14 month (p
< 0.0005). The latter three categories did not differ in terms of median survival
(p = 0.23). Within the group of AML with recurrent genetic abnormalities, only the
patients with t(6;9) and inv(3) reached median survival whereas all other categories
had a favorable outcome.
Patients of the good risk ELN group did not reach the median survival time whereas
median survival of the intermediate and adverse risk groups was 27 and 12 months (p
< 0.0005). Patients who underwent HSCT did not reach median survival. Patients treated
with induction and consolidation had a median survival time of 123 months compared
to 8 months median survival in patients who got HMA (p < 0.0005).
In a multivariate analysis including intensity of therapy, age at diagnosis, and ELN
group, only intensity of treatment and ELN group were independently influencing overall
survival.
Summary/Conclusion: The WHO and the ELN classification could be assessed correctly
in about 70% of patients only, primarily due to the missing examination of somatic
mutations. The ELN classification together with the treatment intensity have major
impact on outcome. Patients who neither receive induction and consolidation nor HSCT
are facing an unfavorable outcome, even if they are treated with HMA.
P533: SINGLE-CENTER PHASE 2 STUDY OF PD-1 INHIBITOR COMBINED WITH DNA DEMETHYLATION
AGENT + CAG REGIMEN IN PATIENTS WITH HIGH-RISK ACUTE MYELOID LEUKEMIA: INTERIM ANALYSIS
X.-N. Gao1,*, Y.-F. Su1, Y. Jing2, J. Wang1, L. Xu1, L.-L. Zhang2, A. Wang1, Y.-Z.
Wang1, Y.-F. Li2, D.-H. Liu1
1Senior Department of Hematology, the Fifth Medical Center; 2Department of Hematology,
the First Medical Center, Chinese PLA General Hospital, Beijing, China
Background: The expression of PD-L1 is increased in acute myeloid leukemia (AML) cells.
However, blocking the immune checkpoint alone has limited efficacy as a single agent
in highly proliferative leukemia cells. Here we report the results of an interim analysis
of an ongoing single-arm phase 2 trial (no. NCT04541277) of PD-1 inhibitor combined
with DNA demethylation agent + CAG regimen in patients with high-risk AML.
Aims: To assess safety and response to Tislelizumab + Azacytidine/Decitabine + CAG
regimen after minimum 1 cycle of therapy in patients who failed at least one prior
induction course of therapy or developed relapse (Cohort 1), and patients with persistent
minimal residual disease (MRD)-positivity after consolidation treatment (Cohort 2).
Methods: Patients must have failed at least one prior induction course of therapy
or have a relapse or have a persistent MRD-positive disease after consolidation treatment.
Other eligibility criteria included in ECOG performance status ≤ 2, normal organ function,
and no autoimmune diseases requiring systemic immunosuppression. The patients received
Azacytidine 75 mg/m2 subcutaneously daily, day 1-7 or Decitabine 20 mg/m2 intravenously
daily, day 1-5 plus CAG regimen (cytarabine 100 mg intravenously every 12 hours, day
1-5; aclarubicin 20 mg intravenously daily, day 1-5; and concurrent use of G-CSF 5 mg/kg/day
subcutaneously) with Tislelizumab 200 mg beginning on the next day after chemotherapy
was stopped and every 4 weeks thereafter. This study was approved by the Ethics Committee
of the Chinese PLA General Hospital, and signed informed consents were obtained from
all patients.
Results:
Efficacy: Fifteen patients has been enrolled (Table 1). Fourteen (93.3%) are evaluable
for response, 7 (46.7%) completed 2 cycles and 8 (53.3%) completed 1 cycle: Of the
12 patients in the previous treatment failure cohort, 7 achieved complete remission
(CR)/CR with incomplete hematologic recovery (CRi) (1/7) (Table 1B), 3 partial remission
(PR), and 2 non-remission. Of the 2 patients in the persistent MRD-positivity after
consolidation treatment cohort, 1 achieved MRD negativity and 1 had no response. The
8-week mortality were 18.8%: 2 patients died of rapidly progressive disease concomitant
with lung infection at 34 days and 56 days after treatment, respectively. One died
of relapse of disease at +4 months after received HLA-haploidentical peripheral blood
stem cell transplantation (PBSCT). One died of acute graft-versus-host disease of
the gastrointestinal tract at +35 days after received a HLA-haploidentical PBSCT in
NR status at transplant. With a median follow-up of 5.4 months (1.1 -12.7), the median
overall survival (OS) is not reach with an estimated 1-yr OS of 59.6% (95%CI, 17.8%-85.6%).
The median OS for responders (CR/CRi+PR) is not reach vs. 1.9 months for non-responders
(p=0.083). The median event-free survival (EFS) is 6.5 months with an estimated 1-yr
EFS of 58.3% (95%CI, 7.7%-89.3%).
Toxicity: Grade 2 immune-related adverse events (IRAEs) were observed in 2 (13.3%)
patients, included in 1 patient with grade 2 skin rash which resolved with steroids,
and the other with grade 2 peripheral sensory neuropathy. All patients developed treatment-related
grade 3 or 4 hematologic toxicities. Two patients died of lung infection at 34 days
and 56 days after treatment in the setting of PD.
Image:
Summary/Conclusion: Tislelizumab + Azacytidine/Decitabine + CAG regimen is safe and
effective for high-risk AML patients. IRAEs are mild, low-grade. Special attention
needs to be paid to patients who have an increased risk of developing graft-versus-host
disease.
P534: AZACITIDINE PLUS VENETOCLAX FOR THE TREATMENT OF RELAPSED AND FIRST-LINE AML
PATIENTS
S. Garciaz1,*, M.-A. Hospital1, A.-S. Alary2, C. Saillard1, Y. Hicheri1, B. Mohty1,
J. Rey1, E. D’incan1, A. Charbonnier1, V. Ferdinand1, V. Maisano1, L. Lombardi1, A.
Ittel3, M.-J. Mozziconacci3, V. Gelsi-Boyer3, N. Vey1
1Hematology; 2Molecular biology; 3Biopathology, Institut Paoli-Calmettes, MARSEILLE,
France
Background: Venetoclax (VEN) belongs to a novel BH3-mimetic class of small molecules
that selectively targets BCL-2, activating the apoptosis effectors BAX and BAK to
drive mitochondrial outer membrane permeabilization, cytochrome c release and cell
death. Combination of VEN and the hypomethylating agent azacitidine (AZA) has deeply
changed the paradigm of treatment of newly diagnosed (ND) AML patients ineligible
for high dose chemotherapy because of older age or comorbidities. There is scarce
evidence for the utilization of VEN-AZA for relapsed or refractory (R/R) AML, a category
of patients classically associated with an extremely poor outcome.
Aims: The objective of our study was to describe a R/R AML cohorts of AML patients
treated with VEN-AZA in our institution and to compare the clinical and molecular
characteristics predicting response in R/R AML versus ND AML
Methods: This retrospective study included consecutive patients treated with VEN-AZA
for R/R AML and ND AML. Patients received AZA at standard dose of 75 mg/m2 QD for
seven days and VEN was administrated either at 400 mg or at 100 mg when associated
with strong CYP3P450 A3 inhibitors after three days ramp up. Response was determined
using the ELN 2017 criteria. The ORR was defined as the combination of complete response
(CR), CR with incomplete hematologic recovery (CRi), and morphologic leukemia-free
state (MLFS).
Results: We compared the outcome of 39 R/R AML and 38 concomitant ND AML patients
treated in our institution between Jan. 2020 and Dec. 2021. The median age was 69
(22-86) and 73 (61-81) in the R/R and ND groups, respectively. Thirty-five percent
of patients had MRC-AML. Adverse cytogenetics was found in 36% of patients in the
R/R group and 59% of patients in the ND group. Most frequent mutations were ASXL1,
RUNX1, TET2, IDH1/2 and TP53 found in 33%, 33%, 28%, 24% and 22% of patients, respectively.
Overall response rate was lower in R/R AML (37% versus 56%) including 13% CR, 8% CRi,
3% PR and 13% MLFS in the R/R AML group and 32% CR, 13% CRi and 13% MLFS in the ND
AML group. Adverse cytogenetics was associated with treatment failure only in the
R/R group (Relative Risk=0.10, p=0.005). ASXL1, IDH1/2 and SFSR2 mutations were associated
with a trend in a higher response rate in the R/R group. Median overall survival (OS)
were 5.9 months in the R/R group and 9.4 months in the ND group. In the R/R group,
median OS were 2.2 months in the adverse cytogenetics group versus 8.7 months in the
intermediate cytogenetics group (p=0.02). Median leukemia-free survival of responding
patient was not different between the two groups (9 months), indicating that VEN-AZA
can be efficient as a salvage treatment for selected R/R AML patients.
Summary/Conclusion: We described one of the largest series of R/R AML patients treated
wiht VEN-AZA. By a doing a direct comparison between R/R AML and ND AML treated concomitantly,
we found that adverse cytogenetics was associated with treatment failure only in the
R/R group suggesting that this subgroup of patients should not be treated with VEN-AZA.
Further analyses including more patients are needed to determine which subgroup may
benefit from the VEN-AZA as a salvage treatment.
P535: UPDATES FROM ITALIAN MULTICENTER REAL-LIFE EXPERIENCE ON CPX-351 THERAPY IN
YOUNG PATIENTS (<60 YEARS OLD).
B. Garibaldi1,*, M. Franciosa2, F. Pilo3, D. Menotti4, V. Cardinali5, L. Brunetti4,
E. A. Martino6, E. Vigna6, I. Tanasi7, A. Duminuco1, C. Maugeri8, M. S. Parisi8, P.
F. Fiumara8, E. Mauro8, M. Gentile6, M. P. Martelli5, D. Capelli4, C. Romani3, S.
Galimberti2, G. A. Palumbo1,8, F. Di Raimondo8, C. Vetro8
1Postgraduate School of Hematology, University of Catania, Catania; 2Division of Haematology,
A.O.U. Pisana, U.O. Ematologia, Pisa; 3U.O.C. Ematologia e Trapianto di cellule staminali
emopoietiche, A.O. Brotzu, Cagliari; 4SOD clinica ematologica, Ospedale Umberto I
di Ancona, Ancona; 5Division of Haematology, A.O. S. Maria della Misericordia, Perugia;
6Division of Haematology, A.O. di Cosenza, Cosenza; 7Division of Haematology, A.O.U.
Policlinico ‘G.B. Rossi’, Verona; 8Division of Haematology, A.O.U. Policlinico “G.Rodolico”
- S.Marco, Catania, Italy
Background: CPX-351 has been indicated as a valid treatment approach in acute myeloid
leukemia (AML) with myelodysplasia-related changes (AML-MRC) and therapy-related AML
(t-AML). Data on young patients are limited.
Aims: Explore CPX-351 efficacy in younger patients (pts) (<60 years old) in real-life.
Methods: Since September 2019 we treated 32 pts from 7 Italian centres with CPX-351.
Median age was 55 (range 32-59) and ECOG range 0-2. Our cohort consisted in 6 pts
with t-AML and 26 pts with MRC-AML (10 morphologic, 11 secondary to MDS, 3 secondary
to MDS/MPN, 6 with MRC cytogenetics features including 2 secondary to MDS and 2 morphologic
AML-MRC). 2 pts (both with AML-MRC) harboured FLT3-ITD and 1 patient FLT3-TKD, 4 pts
IDH1 and 3 pts TP53 mutations. NPM1 was negative in all pts. 10 pts had complex karyotype
(CK) and 5 pts received prior treatment with hypomethylating agents for MDS.
Results: All pts underwent induction therapy with intravenous administration of CPX-351
at day 1, 3 and 5 except 1 patient who received two doses due to pneumonia. 2 pts
(6%) died during induction. Most frequent complication was febrile neutropenia (46%).
At disease re-evaluation, we obtained 16 (50%) Complete Remission (CR), 5 (16%) CR
with incomplete hematological recovery (CRi), 1 (3%) Morphological Leukemia Free State
(MLFS), 4 (12,5%) Partial Remission (after reinduction converted to 2 CR, 1 CRi, while
1 patient was refractory) and 4 (12,5%) refractory pts who switched to salvage therapies
except for one who died due to disease progression. Overall response rate (ORR) was
68% after induction and 78% after reinduction. All 25 responding pts underwent consolidation,
except one that proceeded directly to allogenic stem cell transplantation (HSCT).
16 out 25 responding pts (64%) underwent HSCT. CR rate in patient with CK was 60%.
At induction, median days of severe neutropenia (defined as neutrophils ≤ 500/uL)
and thrombocytopenia (defined as platelets ≤20.000/uL) were respectively 26 and
22. Regarding subgroup analysis by driving mutations, the 4 IDH1 pts obtained a CR
and 3 of them received HSCT. 2 out of 3 TP53 pts were refractory and the other one
obtained a CR followed by HSCT. About the 2 FLT3-ITD pts, one died during induction
and the other one obtained CR after reinduction and then underwent HSCT. After a median
follow-up of 19 months (mo), 6 pts out of the 25 who reached CR/CRi (24%) relapsed
and 20 out of 32 (63%) pts are alive. Median relapse-free survival (RFS) for responding
pts was not reached. RFS was inferior for t-AML compared to AML-MRC (7 mo vs not reached
median, p=0.032). RFS was negatively influenced by CK (median RFS 7 mo for CK vs not
reached if no CK, p=0.009) and performance status (PS) (median RFS not reached for
PS 0 vs 15 months for PS 1 vs 7 mo for PS 2, p=0.017). Median overall survival (OS)
was 20 mo. Pts responding to induction did not reach median OS, while refractory pts
had a median OS of 9 months. Noteworthy, OS was not influenced by cytogenetic risk,
subtype of AML (if MRC or t-AML) or mutational status. Pts undergoing HSCT at first
CR, did not reach median OS, while not transplanted pts had a median OS of 17 mo.
COX multivariate analysis showed that reaching CR was the only significative parameter
for OS (HR 8, 95% CI 1.18-54).
Summary/Conclusion: CPX-351 is active in young pts with t-AML and AML-MRC with an
ORR higher than that reported in the pivotal study (78% vs 47%) and high rate of bridge
to transplant at first remission (56%) also considering the different age range. CK
and PS 1-2 negatively impacted RFS. OS was influenced by response to induction and
HSCT.
P536: VALUE OF MEASURABLE RESIDUAL DISEASE BY MULTIPARAMETRIC FLOW CYTOMETRY IN NON-PROMYELOCYTIC
ACUTE MYELOID LEUKEMIA. REAL WORLD EVIDENCE
A. D. Gimenez Conca1,*, J. Gonzalez2, M. M. Rivas3, A. Navickas4, I. Fernandez5, I.
Rey6, H. Dick7, S. Cranco8, M. Moirano9, L. Ferrari5, M. Clavijo10, A. Suero11, R.
Ramirez12, A. L. Basquiera13, N. Carnelutto14, M. L. Rapan15, A. Jorge Alberto1, C.
Belli16
1Hospital Italiano de Buenos Aires; 2Hospital Durand, Buenos Aires; 3Hospital Austral,
Pilar; 4Hospital El Cruce, Florencio Varela; 5Fundaleu; 6Hospital Ramos Mejia, Buenos
Aires; 7Hospital Italiano de La Plata, La Plata; 8Instituto Alexander Fleming, Buenos
Aires; 9Hospital San Martin, La Plata; 10Hospital Aleman; 11Hospital Cesar Milstein,
Buenos Aires; 12Instituto Conciencia, Neuquen; 13Hospital Privado, Cordoba; 14Hospital
de Clínicas; 15Sanatorio Sagrado Corazon; 16Academia Nacional de Medicina, Buenos
Aires, Argentina
Background: Measurable residual disease (MRD) allows recognition of a group of patients
with acute myeloid leukemia (AML) with a higher risk of relapse. There are, however,
remaining open questions, since clinical studies applied variable techniques, cutoff
values and time points. Multiparameter flow cytometry (MFC) is the most accessible
method in Argentina, since real time PCR assessment is not available for the majority
of the institutions. We designed, therefore, this real-life study using decentralized
assessment of MRD by MFC to solve the role of positive MRD at different times during
treatment with standard chemotherapy.
Aims: Our aim was to assess the impact of positive MRD at the end of induction and
after first consolidation (C1) as a prognostic factor for relapse-free survival (RFS)
and overall survival (OS) in patients with non-promyelocytic AML.
Methods: The registry of the Argentinian Group of Acute Leukemia (GALA) from the Argentine
Society of Hematology (SAH) includes data from 20 centers. We retrospectively selected
an analytical cohort with all patients with AML, treated with an intensive regimen
and in complete remission (CR) after induction, from January 2010 to March 2021. The
OS and RFS were estimated with Kaplan-Meier method and its comparison was evaluated
by log-rank, censoring patients at the time of bone marrow transplantation (BMT).
The cut-off point for MRD analysis was 0.1% evaluated at the end of induction and
after C1. A subanalysis of patients with unknown and intermediate cytogenetic risk
(CRM modified according to available molecular data of FLT3, NPM1 and CEBPA) was performed.
Results: Of the 1,160 patients enrolled in the registry, 493 met the inclusion criteria
and 20 were excluded due to lack of MRD data at the end of induction. Among the 473
patients analyzed: 238 were women, the median age was 49.5 years [IQR 36.6-59.7]),
184 (38.9%) were intermediate risk and 51 (10.8%) with not evaluable cytogenetic and
molecular data. With a median follow-up of 18.9 months, 158 (33.4%) relapsed and 130
(27.5%) died.
The median (Me) OS for patients with post induction negative MRD (MRDneg) was 61.2
(CI95% not evaluable) versus 25.9 months (CI95% 2.5-49.2) with positive MRDpos, p
=0.03. The RFS were 20.4 (CI95 8-32.8) and 12.4 (CI95% 6.5-18.3), p=0.06, for MRDneg
and MRDpos respectively.
Regarding the evaluation of MRD post C1, the Me OS was not reached for patients with
MRDneg, while it was 22.1 months (CI95% 9.3-34.8) among patients with MRDpos, p=0.03.
Their respective RFS were 30.3 (CI95% not evaluable) and 14.4 months (CI95% 0-30.7),
p=0.03, respectively.
In the subanalysis carried out in the group with intermediate or unknown cytogenetic
risk post C1, the Me OS was not reached in MRDneg versus 17.5 months (CI95% 9.3-25.7)
in MRDpos, p=0.007. In addition, a median RFS of 20.4 months (CI95% 0.7-40.1) vs 7.4
(CI95% 3.9-10.9), p=0.003, among those with MRDneg and MRDpos, respectively.
Summary/Conclusion: The assessment of MRD by MFC showed predictive power at the different
moments of follow-up evaluated. It was more evident after consolidation, with statistically
significant impact in OS and RFS. The post-consolidation MRD subanalysis in the group
with IR or non-evaluable cytogenetics showed promising results. This real-practice
data is consistent with those published recently. Post-C1 MRD could be a useful tool
in clinical practice deciding the best post-induction strategy in intermediate-risk
patients. A further detailed study is required to confirm our preliminary results.
P537: SURVIVAL AFTER ALLOGENEIC TRANSPLANTATION FOR ACUTE MYELOID LEUKEMIA IN ADULTS
IN DENMARK FROM 2000 TO 2020: A POPULATION-BASED COHORT STUDY
L. K. Gjærde1,2,*, L. H. Jakobsen3,4, C. Juhl-Christensen5, G. Olesen5, I. Petruskevicius5,
M. T. Severinsen3, C. W. Marcher6, K. Theilgaard-Mönch1, N. S. Andersen1, L. S. Friis1,
B. Kornblit1, S. L. Petersen1, I. Schjødt1, H. Sengeløv1,2
1Department of Hematology, Rigshospitalet; 2Department of Clinical Medicine, University
of Copenhagen, Copenhagen; 3Department of Hematology, Aalborg University Hospital;
4Department of Mathematical Sciences, Aalborg University, Aalborg; 5Department of
Hematology, Aarhus University Hospital, Aarhus; 6Department of Hematology, Odense
University Hospital, Odense, Denmark
Background: Allogeneic hematopoietic cell transplantation (HCT) is a potentially curative
treatment of acute myeloid leukemia (AML), but survival is challenged by the risk
of relapse and treatment-related/non-relapse mortality (NRM). In the recent two decades,
older patients (>50 years of age) and patients with comorbidities have been able to
receive HCT due to the introduction of non-myeloablative conditioning in Denmark in
2001.
Aims: We aimed to investigate trends in overall survival (OS) and other HCT-related
outcomes in a population-based cohort study of all adults (≥18 years) who received
a first HCT for AML in Denmark between 2000–2020.
Methods: Indications for HCT comprised intermediate- or adverse-risk (including secondary/treatment-related)
AML in 1st complete remission (CR) and all patients in ≥2nd CR. The total effect of
calendar year of HCT on transplant outcomes was tested in a Cox model for OS and Fine-Gray
competing risks models for NRM, relapse, grade II–IV acute graft-versus-host disease
(GvHD) and chronic GvHD. Time-specific OS estimates and cumulative incidences of HCT
outcomes were derived using the pseudo-observation framework. Cure fractions (the
fraction of patients who experienced no excess mortality compared to the Danish general
population matched on calendar year, age and sex) were estimated in a mixture cure
model.
Results: From 2000 to 2020, 659 adults received a first HCT for AML (95% de novo AML;
5% secondary/therapy-related AML) in Denmark. Median (min–max) age at HCT was 56 (19–74)
years, going from 44 years in 2000–2005 to 59 years in 2016–2020. 63% of patients
were transplanted in 1st CR, 35% in ≥2nd CR, and 2% after primary induction failure.
Non-myeloablative conditioning was performed in 60% of patients, going from 26% in
2000–2005 to 65% in 2016–2020. Median follow-up was 7.5 years. Main causes of death
during follow-up were relapse (56%), infection (11%), and organ failure (10%). OS
at 2- and 5-years for all patients was 66% (95% CI: 63–70%) and 57% (95% CI: 53–61%),
respectively. OS differed over time (p = 0.02), decreasing from 2000 to 2010 from
when it increased until 2020 (Figure Panel A). This change was mainly driven by corresponding
changes in NRM over time (p < 0.001, Figure Panel B) rather than significant increases
in the relapse rate over time (p = 0.16, Figure Panel C). Adjusting for age did not
influence the trends notably. For acute GvHD, the risk decreased over time (p = 0.004),
ranging from a 1-year cumulative incidence of 42% (95% CI: 26–62%) in 2000 to 26%
(95% CI: 19–34%) in 2020, whereas the risk of chronic GvHD remained stable (p = 0.49),
with a 2-year cumulative incidence of 49% (95% CI: 45–53%) in the full cohort. Cure
fractions differed over time (p = 0.05) decreasing from 63% (95% CI: 41–78%) in 2000
to 46% (95% CI: 38–54%) in 2010, and increasing again to 68% (95% CI: 52–80%) in 2020.
Image:
Summary/Conclusion: Survival after HCT for AML in adults in Denmark decreased in the
first decade after introducing non-myeloablative conditioning regimens for older or
comorbid patients, mainly because of higher NRM, but survival improved again in the
later decade. The risk of acute, but not chronic, GvHD decreased over time. The latest
improvements in peri-HCT supportive care and post-HCT maintenance treatments may further
improve survival in the coming decade.
P538: TREATMENT PATTERNS AND CLINICAL OUTCOMES IN ACUTE PROMYELOCYTIC LEUKEMIA: REAL-WORLD
DATA
V. B. Goli1,*, H. Jain1, L. Nayak1, J. Thorat1, B. Bagal1, A. Rajendra1, S. P G2,
D. Shetty3, H. Jain3, P. Tembhare2, N. Patkar2, M. Sengar1
1Medical Oncology; 2Pathology; 3Cytogenetics, TATA MEMORIAL HOSPITAL, MUMBAI, India
Background: All-trans retinoid acid (ATRA) and arsenic trioxide (ATO) have changed
the treatment paradigm of acute promyelocytic leukemia (APML). Current therapeutic
strategies are based on the risk stratification at diagnosis and molecular response
at the end of consolidation.We report our experience of treatment patterns and clinical
outcomes of newly diagnosed APML patients.
Aims: The primary objective of our study was to evaluate event-free survival in all
risk categories of APML.Secondary objectives include disease-free survival, overall
survival, rates of complete remission (CR) at the end of induction, rates of molecular
CR at the end of consolidation.
Methods: We included treatment-naïve APML patients age>14 years who were treated between
May 2014 to May 2018.The diagnosis of APML was established by peripheral blood/bone
marrow morphology and flow cytometry (FCM) and confirmed by fluorescent in situ hybridization
(FISH) and reverse-transcriptase polymerase chain reaction (RT-PCR) for PML-RARA.The
details of patients were retrieved from electronic medical records.Patients with low/intermediate
risk were treated with ATO-ATRA based induction and consolidation,where as patients
with high risk were treated with varying combination of ATO/ATRA/chemotherapy with
or without maintenance.
Results: We registered 149 patients during the study period.The median age was 37
years(range 15-72 years, male 56.6%, female-37%).93 patients (62.4%) were stratified
as low risk and 56 patients (37.5%) as high risk based on Sanz’s score.The induction
therapy for high risk APML was single agent ATO (42.8%),ATO/ATRA/anthracycline (26.7%),ATO/anthracycline
(25%),and ATO/ATRA (5%).The consolidation for high risk APML includes ATRA/daunorubicin
(50%),ATO/ATRA (26.8%),HIDAC (4%) and APL 2000 in one patient.All low-risk APML patients
received ATO-ATRA in induction and consolidation.Anthracycline was added to three
patients in induction to decease white cell count.The proportion of deaths that occurred
in first week include 6.5% in low risk and 9% in high risk group.The median follow
up was 42 months.The hematological CR at the end of induction was 85%.(128/149 patients,expired
during induction-14,lost to follow up-7).The rate of end of consolidation PCR negativity
was 83%.Differentiation syndrome occurred in 75% of patients.For the overall patient
population, the 3-year probability for OS, EFS, DFS was 88.1% (95% C.I, 82.9 93.6),
75.9% (95% C.I, 68.9-83.5), 81.4% (95% C.I, 75-88.4) respectively. The 3-year survival
probability for OS, EFS, and DFS for low risk APML was 90% (95% C.I, 84.1-96.4), 80.1%
(95% C.I, 72-89.1), 84.5% (95% C.I, 76.9-92.7) respectively whereas for high risk
APML was 84.7% (95% C.I, 75.3-95.2), 68.6% (95% C.I, 56.9-82.9), 76.1% (95% C.I-65-89)
respectively.The total number of relapses were 11.The median time to relapse for low
risk and high risk was 23months(18-32months) and 20months(range 5-39months) respectively.
Image:
Summary/Conclusion: Treatment with ATO and ATRA based regimen are effective in real
world setting.The emphasis should be to reduce the early death rate due to bleeding
and infectious complications.
P539: PROGNOSTIC RELEVANCE OF MINIMAL RESIDUAL DISEASE IN THERAPY RELATED AND SECONDARY
ACUTE MYELOID LEUKEMIA RECEIVING CPX-351 OR FLUDARABINE-BASED INDUCTION.
F. Guolo1,2,*, P. Minetto1, C. Riva1,2, F. Parodi2, M. Miglino1,2, E. Tedone3, N.
Colombo3, E. Carminati4, C. Nurra4, A. Cagnetta1, M. Cea1,2, R. M. Lemoli1,2
1Clinic of Hematology, Department of Internal Medicine (DiMI), IRCCS Ospedale Policlinico
San Martino; 2University of Genoa; 3Flow Cytometry Unit, Department of Pathological
Anatomy; 4Molecular Biology Unit, Department of Pathological Anatomy, IRCCS Ospedale
Policlinico San Martino, GENOVA, Italy
Background: Minimal residual disease (MRD) assessment retains high prognostic value
in Acute Myeloid Leukemia patients (AML) undergoing intensive induction therapy. Widely
available MRD techniques include multicolor flow cytometry (MFC), RT-PCR for recurrent
genetic lesion and, for patients lacking specific markers, RT-PCR for the pan- leukemic
marker WT1. However, most of the data on the prognostic value of MRD come from trials
including younger patients treated with conventional 3 + 7 regimen. Furthermore, AML
arising from a previous myelodisplastic syndrome (s-AML) and therapy-related AML (t-AML)
are usually under-represented in trial involving younger patients. Few data are, therefore,
available on the kinetics and the prognostic value of MRD in elderly s- AML and t-AML
patients; especially in the context of more modern frontline treatment such as CPX-351.
Aims: We evaluated MRD in a cohort of elderly s-AML or t-AML patients receiving induction
therapy either with an age-adjusted fludarabine-containing regimen (FLAI3) or CPX-351,
in order to compare the probability of achieving MRD negativity, to assess the prognostic
value of MRD and to define the best time-points for MRD assessments.
Methods: A total of 151 elderly (>60 year, median age 68, range 60-77) patients were
analyzed in this study. Patients were treated between January 2005 and January 2020
in our Center, either with CPX-351 (n=50) or fludarabine- high dose cytarabine-idarubicin
(FLAI), with (n=72) or without (n =29) gemtuzumab-ozogamicin (GO). MRD was analyzed
in all patients achieving hematological complete remission (CR) with both MFC and
WT1 expression levels. All patients were affected by s-AML or t-AML, defined according
to the WHO 2016 criteria.
Results: After induction, CR was achieved in 95 patients (59%). CR rate was 40/50
in patients treated with CPX-351 (80)% significantly higher when compared to patients
receiving FLAI (55/101, 54.5%, p<0.05). The addition of GO to FLAI did not increase
CR rate. Among CR patients, a total of 51 (53.7%) and 53 patients (55.8%) achieved
MRD negativity, with MFC or WT1-based methods, respectively. MFC MRD negativity probability
was higher among patients receiving CPX-351 as induction therapy (MFC MRD negativity
rate of 26/40, 65% and 25/55, 45% in CR patients who received CPX-351 or FLAI, respectively,
p<0.05). Adding GO to FLAI did not improve MRD negativity probability. The most informative
timepoint was after first cycle. WT1-based MRD assessment led to similar results.
In multivariate analysis, MRD showed significant prognostic value for Overall Survival
(OS) in all treatment group (2-year OS of 34% and 77% in patients with or without
residual MFC MRD after induction, respectively, p<0.05). Similarly, consolidation
with allogeneic stem cell transplantation (HSCT) was correlated with higher OS. Notably,
12/40 (30%) CR patients treated with CPX 351 underwent HSCT.
Summary/Conclusion: In conclusion MRD assessment retains a strong prognostic value
also in s-AML and t-AML patients. The evaluation of MRD with both methods lead to
similar conclusions and allowed us to obtain MRD data from virtually all AML patients
treated in the selected time period. CPX-351 treatment resulted in higher CR rate
with deeper MRD responses, if compared to FLAI3 and allowed a significant number of
elderly AML patients to proceed to HSCT.
P540: PEVONEDISTAT, AZACITIDINE AND VENETOCLAX FOR PATIENTS WITH RELAPSED/REFRACTORY
ACUTE MYELOID LEUKEMIA– A PHASE I STUDY
G. S. Guru Murthy1,*, S. Kaufmann2, A. Saliba2, A. Szabo3, L. Michaelis1, S. Abedin1,
L. Runaas1, K. Carlson1, S. Maldonado-Schmidt4, A. Hinman4, A. Thomas4, A. Baim4,
M. Litzow2, E. Atallah1
1Hematology-Oncology, Medical College of Wisconsin, Milwaukee; 2Hematology-Oncology,
Mayo Clinic, Rochester; 3Biostatistics; 4Clinical trials, Hematology-Oncology, Medical
College of Wisconsin, Milwaukee, United States of America
Background: Outcomes of patients with relapsed/refractory acute myeloid leukemia (RR-AML)
have remained poor. Venetoclax therapy in RR-AML is associated with lower complete
remission (CR) as compared to newly diagnosed AML. Preclinical studies suggest overexpression
of anti-apoptotic MCL-1 as a mechanism of BCL-2 inhibitor resistance (Konopleva et
al. Cancer Cell 2006). Pevonedistat is a first in class inhibitor of Nedd8 activating
enzyme that induces pro-apoptotic NOXA leading to neutralization of MCL-1 and apoptosis.
In preclinical AML models, combination of pevonedistat and venetoclax showed synergistic
effect (Knorr KL et al. Cell Death Differ. 2015).
Aims: To assess the safety and outcomes of adding pevonedistat to azacitidine and
venetoclax in patients with RR-AML.
Methods: This is a phase I multicenter study (NCT04172844) with 3 + 3 design to determine
the safety and recommended phase 2 dose (RP2D) of pevonedistat, venetoclax and azacitidine
in RR-AML. Patients aged 18 years or above with morphologically documented RR-AML,
ECOG performance status 0-2 and adequate organ function were eligible. Exclusion criteria
were isolated extramedullary relapse, hematopoietic stem cell transplantation (HSCT)
within 100 days of enrollment, and active acute graft versus host disease. Previous
therapy with hypomethylating agent (HMA) or venetoclax was not an exclusion. Treatment
included azacitidine (75 mg/m2 daily x 7 days), venetoclax (400 mg daily x 28 days),
and pevonedistat in escalating doses (10-20 mg/m2 IV days 1,3,5 of each cycle) in
28-day cycles. Pevonedistat was given at 10 mg/m2 dose in cohort 1, 15 mg/m2 in cohort
2 and 20 mg/m2 in cohort 3.
Results: Sixteen patients with RR-AML participated in the study (15 evaluable for
response). Median age was 73 years (61-91), 37.6% had secondary/therapy related AML,
56.3% received prior venetoclax/HMA and 18.1% had relapse after prior allogeneic HSCT.
Most common grade 3 or higher adverse events included neutropenia (44%), thrombocytopenia
(38%), febrile neutropenia (25%), anemia (25%), and sepsis (19%). There was 1 dose
limiting toxicity (DLT) in cohort 1 (atrial fibrillation), but no subsequent DLT despite
planned dose escalation. Pevonedistat 20 mg/m2 was established as the RP2D. The rate
of CR/CRi/morphological leukemia free state (MLFS) was 40% (CR/CRi 33%) for the overall
cohort and 85.7% in venetoclax/HMA naïve RR-AML with a median time to response of
1 cycle. Among patients achieving CR/CRi, 60% attained minimal residual disease negativity
by flow cytometry.
Correlative studies with BH3 mimetic profiling showed variable baseline sensitivity
to BH3 mimetics and sequential western blot assays showed upregulation of PUMA (for
two patients in CR) and NOXA (for one patient with MLFS). Higher levels of DNMT1 were
seen in patients with CR or MLFS. One patient with CR also had evidence of leukemic
stem cell/progenitor cell sensitivity to pevonedistat at 50-100 nM.
Summary/Conclusion: Combining pevonedistat with venetoclax and azacitidine is safe
and tolerable in patients with RR-AML. Dose escalation yielded encouraging efficacy
with pevonedistat 20 mg/m2 as the RP2D.
P541: OUTCOMES AND MANAGEMENT OF PATIENTS WITH NEWLY DIAGNOSED ACUTE MYELOID LEUKEMIA
PRESENTING WITH HYPERLEUKOCYTOSIS
F. Haddad1,*, K. Sasaki1, T. Abuasab1, S. Venugopal1, D. Rivera Delgado1, A. Bazinet1,
R. Babakhanlou1, K. Kim1, J. Senapati1, F. Ong1, S. Desikan1, N. Short1, N. Pemmaraju1,
G. Borthakur1, C. DiNardo1, N. Daver1, E. Jabbour1, G. Garcia-Manero1, F. Ravandi1,
T. Kadia1
1Leukemia, The University of Texas MD Anderson Cancer Center, Houston, United States
of America
Background: Patients with acute myeloid leukemia (AML) presenting with hyperleukocytosis
have higher mortality rates and inferior outcomes. Analyzing the factors associated
with mortality and survival in AML patients with a white blood cell count (WBC) ≥100
x 109/L can guide management and improve early mortality.
Aims: To analyze the outcomes of patients with AML presenting with hyperleukocytosis
and establish a treatment algorithm for the management of those patients.
Methods: In a retrospective analysis, we screened patients who presented to our institution
for newly diagnosed AML over the period of 10 years and identified those with WBC
≥100 x 109/L. Logistic regression models estimated odds ratios (OR) for 4-week mortality
and Cox proportional hazard models estimated hazard ratios (HR) for overall survival
(OS).
Results: We identified 129 patients with newly diagnosed AML and hyperleukocytosis.
Median age was 65 years (range, 23-86 years) and 66% of patients had ECOG performance
status (PS) <2. Median WBC was 146 x 109/L (range, 100-687) and 78 patients had clinical
leukostasis (CL) including renal failure in 31 patients (24%), new onset hypoxia in
29 patients (23%), headache in 19 patients (15%), chest pain in 9 patients (7%) and
neurological symptoms in 3 patients (2%). FLT3 and RAS pathway mutations were found
in 63% and 27% of patients, respectively. 29 of 129 patients (22%) had poor-risk cytogenetics.
Compared with patients without evidence of CL, those with CL were less likely to have
good PS (ECOG PS <2, 58% vs 82%; P = 0.006), had higher 4-week mortality (16% vs 2%;
P = 0.015) and 8-week mortality (19% vs 6%; P = 0.038).
Cytoreduction consisted of hydroxyurea in 124 patients (96%), cytarabine in 69 patients
(54%) and leukapheresis in 31 patients (24%). Patients who underwent leukapheresis
were less likely to receive cytarabine compared with those who did not (35% vs 59%;
P = 0.024); and tended to have more CL compared with patients who did not (74% vs
56%; P = 0.093). 30 patients had tumor lysis syndrome (TLS). TLS risk did not increase
with WBC and was not associated with the cytoreductive modality used. 11 patients
had intracranial hemorrhage (ICH): 9 patients (82%) with WBC ≥150 x 109/L and 18%
with WBC <150 x 109/L (P = 0.048). No association was observed between the incidence
of ICH and the cytoreductive therapy.
4-week and 8-week mortality rates were 10% and 14%, respectively. After a median follow-up
of 49.4 months (95% CI, 26.2-72.6), median OS was 14.3 months (95% CI, 7-21.6), with
2-year OS of 40%. Median OS was 12.3 months (95% CI, 7.4-17.2) compared with 29 months
(95% CI, 2.1-55.6) in patients with or without CL, respectively (P = 0.007). Median
OS was 9.9 months (95% CI, 7.5-12.2) and 21.3 months (95% CI, 10.7-31.8) among those
who did or did not undergo leukapheresis, respectively (P=0.003). Median OS was 42
months (95% CI, 14.2-69.8) in patients younger than 65 years compared with 8 months
(95% CI, 6-10) in those 65 years and older.
Image:
Summary/Conclusion: Older age, poor-risk cytogenetics, TLS and disseminated intravascular
coagulation (DIC) were associated with early mortality and inferior OS in patients
with hyperleukocytosis. Careful monitoring of those patients with prompt cytoreduction
and management of complications may help improve their outcomes.
P542: REAL WORLD OUTCOMES FOR PATIENTS WITH ACUTE PROMYELOCYTIC LEUKEMIA IN BRITISH
COLUMBIA (BC): EXCELLENT OUTCOMES WITH LOW EARLY DEATH RATE AND HIGH OVERALL SURVIVAL
IN A POPULATION BASED STUDY
R. Henderson1,*, Y. Eissa1, R. Stubbins1, Y. Abou Mourad1, S. Chung1, D. Forrest1,
K. Hay1, F. Kuchenbauer1, S. Nantel1, T. Nevill1, M. Power1, J. Rodrigo1, C. Roy1,
D. Sanford1, K. Song1, H. Sutherland1, C. Toze1, J. White1, S. Narayanan1
1Leukaemia/Bone Marrow Transplant Programme, Vancouver General Hospital, Vancouver,
Canada
Background: Acute promyelocytic leukaemia (APL) therapy has significantly improved
with the use of All trans retinoic acid (ATRA) and Arsenic Trioxide (ATO) for low
risk disease (Lo-Coco 2013). Treatment of high risk disease remains more challenging
with a higher risk of early mortality & greater concern of relapse. All patients (pts)
with APL in British Columbia, Canada are treated by the Leukemia/BMT Program at Vancouver
General Hospital, Vancouver. Since 2014, all pts have received ATRA/ATO based therapy
based on the Le-Coco study. Pts with high risk disease (WCC > 10 x 109/L at presentation)
also receive a 3 day course of doxorubicin (60mg/m2) for induction only.
Aims: We sought to review treatment outcomes since initiating these treatment protocols
with emphasis on drug toxicities, dose modifications & management of high risk & elderly
patients in order to describe real world outcomes.
Methods: Ethics was approved by the board of the BCCA and UBC. Data was collected
by chart reviews, hospital databases & collated on a central file. Demographics, risk
stratification, treatment received, adverse events & survival was collected & analysis
performed on Microsoft excel v16.57 & SPSS Statistics v28.0.1.1. Kaplan Meier curves
were performed for PFS & OS, student t and chi squared tests were performed to assess
differences between subgroups.
Results: We identified 67 pts with APL during the study period. 40% were male 60%
female, the median age was 61 yrs. (21-83yo). 36(54%) pts were > 60 yrs at diagnosis.
The median duration of follow up was 2.9 yrs. All had molecular confirmation of disease.
16(24%) pts had high risk disease at diagnosis. At time of censor overall survival
was 97% and PFS 94%. 55(82%) pts had a documented toxicity. Common toxicities included
differentiation syndrome (DS) in 27(40%) pts, hepatotoxicity 17 (25%), QTc prolongation
13(19%) & neurological effects 9 (13%). Infections were documented in 40(59%) pts
including bacterial infections in 25 patients (37%), culture negative febrile neutropenia
in 12 (20%), viral infections in 5 (7%) & fungal infections in 1 (1.5%). Clinically
significant bleeding was documented in 10 (15%) of patients, 5 of which were CNS/retinal
bleeds.
DS was more common in high risk disease though this difference was not statistically
significant (p=0.365). 8(30%) pts with DS needed ICU admission. There were 2 early
deaths, in the cohort, both in pts > 75 yo with high risk disease due to multi-organ
failure and bleeding within 5 days of presentation.
Dose modifications were assessed in 64 pts (2 deaths & one lost to follow up). 37
(58%) were described as having had a dose modification. 13 (20%) had a reduction in
treatment dose, 16(25%) had doses/days of drug omitted. 6(9%) had reductions in dose
and omissions. The most common reasons for dose modifications were DS 12(19%), QTc
prolongation 10 (15%) & hepatotoxicity 8 (12%). Pts over 60yo were more likely to
have a dose modification than pts below 60yo (73% vs 43% p = 0.014).
65 pts diagnosed achieved morphological remission & 64 a molecular remission at the
end of planned therapy. 2 relapses occurred in our cohort, 1 had a concomitant cytogenetic
abnormality incorporating a Tp53 deletion. The second initially received ATRA only
induction due to age & comorbidity.
Summary/Conclusion: This real world, population based data for patients with APL in
BC confirms high response rates, survival and relatively low early death rate and
acceptable toxicity even in older patients with high risk disease and comorbidities.
In our cohort appropriate dose modifications did not impact remission or relapse rates.
P543: TARGETING SAMHD1 WITH HYDROXYUREA IN FIRST-LINE CYTARABINE-BASED THERAPY OF
NEWLY DIAGNOSED ACUTE MYELOID LEUKAEMIA: RESULTS FROM THE HEAT-AML TRIAL
M. Jädersten1,*, I. Lilienthal2, N. Tsesmetzis2, M. Lourda2, S. Bengtzén3, A. Bohlin3,
C. Arnroth4, T. Erkers4, B. Seashore-Ludlow4, G. Giraud5, G. S. Barkhordar6, S. Tao7,
L. Fogelstrand8, L. Saft4, P. Östling4, R. Schinazi7, B. Kim7, T. Schaller9, G. Juliusson10,
S. Deneberg1, S. Lehmann11, G. Rassidakis4, M. Höglund11, J.-I. Henter2, N. Herold2
1Department of Hematology, Karolinska University Hospital; 2Childhood Cancer Research
Unit; 3Centre for hematology and regenerative medicine; 4Department of Oncology-Pathology,
Karolinska Institutet, Stockholm; 5Department of Immunology, Uppsala University, Uppsala;
6Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg,
Sweden; 7Department of Pediatrics, Emory University, Atlanta, United States of America;
8Department of Laboratory Medicine, Sahlgrenska Academy at University of Gothenburg,
Gothenburg, Sweden; 9Department of Infectious Diseases, University Hospital Heidelberg,
Heidelberg, Germany; 10Department of Hematology, Skåne University Hospital, Lund;
11Department of Medical Sciences, Uppsala University, Uppsala, Sweden
Background: Treatment of newly diagnosed acute myeloid leukaemia (AML) is based on
combination chemotherapy with cytarabine and anthracyclines. Five-year overall survival
is below 30%, which has partly been attributed to cytarabine resistance. Preclinical
data suggest that addition of hydroxyurea potentiates cytarabine efficacy by increasing
ara-CTP levels through targeted inhibition of SAMHD1.
Aims: To evaluate feasibility, safety, and efficacy of adding hydroxyurea to standard
AML-directed therapy according to national guidelines.
To perform translational studies including SAMHD1-staining on bone marrow sections,
pharmacokinetics and drug-sensitivity analysis on leukemic cells ex vivo.
Methods: This phase-1 trial (EudraCT-number: 2018-004050-16) was run at two sites
(Karolinska University Hospital and Uppsala University Hospital, Sweden). Eligibility
criteria included age >18 years, newly diagnosed non-promyelocytic AML, and fitness
for intensive chemotherapy. Patients with CBF-AML eligible for treatment with gemtuzumab-ozogamicin
were excluded. Treatment comprised 2 to 4 cycles of ara-C 1000 mg/m2 i.v. b.i.d. on
day 1-5 during all 4 cycles and daunorubicin 60 mg/m2 i.v. q.d. on day 1-3 during
cycles 1 and 2, and on day 1-2 during cycle 3. Patients with FLT3-mutated AML received
midostaurin 50 mg b.i.d. on day 8-21 of each cycle. Risk-adapted allo-HSCT was performed
at the discretion of the treating haematologist. The dose of hydroxyurea was escalated
in a 3 + 3 design: 500 + 500 mg (level 1), 1000 + 500 mg (level 2), and 1000 + 1000 mg
(level 3), each dose being given 1 hour prior to start of the ara-C infusion b.i.d.
on day 1-5.
Here we report the results of the first 9 patients in the run-in phase 1 part of the
study. The phase 2 part will include an additional 60 patients. Recruitment is ongoing,
utilizing the highest dose of hydroxyurea.
Expression of SAMHD1 was assessed using a double-immunostaining method (SAMHD1/CD68),
an autostainer system (BenchMark Ultra, Ventana, Rotkreuz, Switzerland) and previously
validated protocols20. CD68+/SAMHD1+ histiocytes (macrophages) served as internal
controls in all bone marrow biopsies assessed.
Drug sensitivity analysis was performed on AML mononuclear cells utilized a high-throughput
system evaluating >500 cytotoxic agents including combinations of ara-C and hydroxyurea
in different doses.
Results: All nine patients (100%) achieved complete remission, and all eight (100%)
with validated MRD measurements (flow-cytometry or RT-qPCR) had an MRD level <0.1%
after two cycles of chemotherapy. Six of nine patients underwent hematopoietic stem
cell transplantation. With a median follow-up of 13.2 months, no relapse has been
observed.
No unexpected toxicities were observed. Pharmacokinetic analyses showed a significant
increase in ara-CTP levels (1.5-fold; P=0.04) in the 6 patients receiving single doses
of 1000 mg hydroxyurea. Drug-sensitivity analysis indicated an additive effect of
ara-C and hydroxyurea on leukemic cells ex vivo. There was no apparent correlation
between expression of SAMHD1-expression and efficacy; all patients had deep responses.
Image:
Summary/Conclusion: The high rate of complete remission and MRD negativity together
with the pharmacokinetic and ex vivo evidence suggest that the efficacy of cytarabine-based
AML treatment can be enhanced by addition of hydroxyurea as a targeted inhibitor of
SAMHD1. Importantly, orally administered hydroxyurea may provide a safe, inexpensive,
and broadly accessible strategy to improve outcome in AML. These results will have
to be validated in a larger patient cohort.
P544: MOLECULAR PREDICTORS OF RESPONSE AND SURVIVAL IN TREATMENT-NAÏVE PATIENTS WITH
ACUTE MYELOID LEUKEMIA FOLLOWING VENETOCLAX AND HYPOMETHYLATING AGENTS
I. Johnson1,*, K. McCullough2, F. Farrukh3, A. Al-Kali3, H. Alkhateeb3, K. Begna3,
A. Mangaonkar3, M. Litzow3, W. Hogan3, M. Shah3, M. Patnaik3, A. Pardanani3, A. Tefferi3,
N. Gangat3
1Department of Medicine; 2Divison of Hematology; 3Mayo Clinic, Rochester, United States
of America
Background: Venetoclax (Ven) in combination with hypomethylating agents (HMA) is FDA-approved
for elderly/unfit AML patients. Limited data exists on molecular predictors of response
and survival. In a prior study, response was superior (CR/CRi >80%) with NPM1, IDH1/2,
DNMT3A mutations, inferior with TP53, RUNX1, FLT3/ITD, RAS and prolonged survival
with NPM1 and IDH2 mutations (2 yr OS 71.8%/79.5%) (DiNardo, Blood, 2020).
Aims: Our objective was to determine the impact of mutations on response and survival
in treatment-naïve AML patients receiving Ven+HMA.
Methods: Treatment-naïve AML patients receiving Ven+HMA outside a clinical trial at
the Mayo Clinic were included. Molecular studies were performed by next-generation
sequencing. Response was assessed according to the 2017 European LeukemiaNet (ELN)
criteria. Standard statistical analyses were performed using JMP Pro (Version 16.0.0).
Results: i) Patient characteristics
103 AML patients (median age 74 years, 67% male, 62% de novo) received upfront Ven+HMA.
ELN cytogenetic risk included favorable (6%, n=6), intermediate (50%, n=52) or adverse
(44%, n=45). Mutations involved TP53 in 25 patients (25%), TET2 in 24 (23%), IDH1/IDH2
in 20 (19%), RUNX1 in 19 (19%), ASXL1 in 18 (18%),), SRSF2 in 18 (18%), K/NRAS in
15 (15%), NPM1 in 13 (13%), DNMT3A in 13 (13%), FLT3-ITD in 10 (10%) patients.
ii) Predictors of response
40 (39%) patients achieved complete remission (CR), 20 (19%) CR with incomplete hematological
recovery (CRi), resulting in CR/CRi in 60 (58%) patients.
In univariate analysis, presence of ASXL1 mutation was associated with favorable response
(CR/CRi 83% vs 53%, p=0.01), secondary AML (CR/CRi 49% vs 65%, p=0.09), adverse karyotype
(49% vs 67%, p=0.11), presence of TP53 (32% vs 67%, p=0.002) and FLT3-ITD mutations
(30% vs 61%; p=0.06) predicted inferior response. In multivariable analysis, including
the aforementioned variables, presence of ASXL1 mutation (83% vs 53%; OR 4.5) and
absence of TP53 (67% vs 32%; OR 3.3) and FLT3-ITD mutations predicted favorable response
(61% vs 30%; OR 6.4). Moreover, in ASXL1 mutated patients, CR/CRi was not impacted
by presence of TP53 mutation (100% vs 81%) whereas in ASXL1 unmutated patients, presence
of TP53 mutation predicted inferior response (26% vs 63%; p=0.001). Presence of NPM1
(CR/CRi; 69% vs 57%, p=0.41), IDH1/2 (70% vs 55%; p=0.23), DNMT3A (54% vs 59%; p=0.73),
RUNX1 (58% vs 58%; p=0.1), RAS (60% vs 58%, p=0.88) did not impact response.
iii) Predictors of survival
After a median follow up of 6.6 months, 68 patients died and 9 underwent allogeneic
stem cell transplant. Median overall survival (mOS) was 8.5 months and longer in transplanted
patients (not reached vs 8.4 months, p=0.08).
Age-adjusted analysis in 94 patients not transplanted, identified CR/CRi (p<0.0001),
NPM1 (p=0.009), IDH1/2 mutations (p=0.02) as favorable, and TP53 (p=0.01), ASXL1 mutations
(p=0.17) adverse karyotype (p=0.05) unfavorable risk factors for survival. Despite
higher CR/CRi, ASXL1 mutated patients had shortened mOS due to higher relapse.
Multivariable analysis confirmed the negative survival impact of not achieving CR/CRi,
ASXL1 mutation and adverse karyotype. Accordingly, a three-tiered model was generated
by using the three variables (Figure 1).
Image:
Summary/Conclusion: Our observations differ from those of DiNardo, et al. (Blood,
2020). The current study identifies presence of ASXL1 mutation and absence of TP53
and FLT3-ITD mutations as predictors of favorable response. Additionally, we propose
a novel three-tiered survival model based on response, ASXL1 mutation and karyotype.
P545: CHARACTERISTICS AND OUTCOME OF PATIENTS WITH ACUTE MYELOID LEUKEMIA AND TRISOMY
19
S. Kayser1,2,*, D. Martínez-Cuadrón3,4, R. Rodriguez-Veiga5, M. Hänel6, M. Tormo7,
K. Schäfer-Eckart8, C. Botella9, F. Stölzel10, T. Bernal del Castillo11, U. Keller12,
C. Rodriguez-Medina13, G. Held14, M.-L. Amigo15, C. Schliemann16, M. Colorado17, M.
Kaufmann18, M. Barrios Garcia19, S. W. Krause20, M. Görner21, E. Jost22, B. Steffen23,
A. D. Ho24, C. Baldus25, H. Serve26, U. Platzbecker1, C. Müller-Tidow24, C. Thiede27,
M. Bornhäuser28, P. Montesinos4,5, C. Röllig10, R. F. Schlenk24,29,30
1Medical Clinic and Policlinic I, Hematology and Cellular Therapy, University Hospital
Leipzig, Leipzig; 2National Center of Tumor Diseases, German Cancer Research Center
(DKFZ), Heidelberg, Germany; 3Hematology Department, 3HemaHospital Universitari i
Politècnic, La Fe, Valencia; 4CIBERONC, Instituto Carlos III, Madrid; 5Hematology
Department, Hospital Universitari i Politècnic, La Fe, Valencia, Spain; 6Klinikum
Chemnitz, Chemnitz, Germany; 7Hematology Department, Hospital Clínico Universitario,
INCLIVA Research Institute, University of Valencia, Valencia, Spain; 8Hospital Nord,
Nürnberg, Germany; 9Hospital General, Alicante, Spain; 10Department of Medicine I,
University Hospital Carl-Gustav-Carus, Dresden, Germany; 11Hospital Central de Asturias,
Asturias, Spain; 12Department of Hematology, Oncology and Tumor Immunology, Charité-University
Medical Center, Campus Benjamin Franklin, Berlin, Germany; 13Hematology Department,
Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria,
Spain; 14Westpfalz Klinikum, Kaiserslautern, Germany; 15Hospital General Universitario
Morales Meseguer, Murcia, Spain; 16University Hospital Muenster, Münster, Germany;
1716Hospital Universitario Marqués de Valdecilla, Santander, Spain; 18Robert Bosch
Hospital Stuttgart, Stuttgart, Germany; 19Department of Hematology, 18DepartmentHospital
Regional Universitario de Málaga, Malaga, Spain; 20Department of Internal Medicine
5 – Hematology/Oncology, 19Department of Internal Medicine 5 – HeUniversity Hospital
of Erlangen, Erlangen; 21Klinik für Hämatologie, Onkologie und Palliativmedizin, Klinikum
Bielefeld Mitte, Bielefeld; 22University Hospital Aachen, Aachen; 23Department of
Internal Medicine II, Department oUniversity Hospital of Frankfurt Main, Frankfurt
am Main; 24Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg;
25Department of Internal Medicine II, University Hospital of, Kiel; 26Department of
Internal Medicine II, 22Department of InterUniversity Hospital of Frankfurt Main,
Frankfurt am Main; 27Department of Medicine I, 9DepartmUniversity Hospital Carl-Gustav-Carus;
28Department of Medicine I, 9DepUniversity Hospital Carl-Gustav-Carus, Dresden; 29NCT
Trial Center, National Center of Tumor Diseases, German Cancer Research Center (DKFZ);
30Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg
University Hospital, Heidelberg, Germany
Background: Trisomy 19 is a recurrent but rare cytogenetic abnormality reported in
patients with acute myeloid leukemia (AML). The prognostic significance of this abnormality
in AML patients is not clear. Prognosis of AML patients with trisomy 19 seems to be
poor as compared to that of patients with intermediate-risk cytogenetics. Allogeneic
hematopoietic stem cell transplantation (allo-HCT) may improve survival if applied
early in first complete remission (CR).
Aims: To characterize AML patients with trisomy 19 and compare outcomes according
to different treatment strategies.
Methods: We retrospectively studied 97 AML patients with trisomy 19 (median age at
diagnosis, 57 years; range, 17-83 years) treated between 2001 and 2019 within 2 study
groups (SAL & PETHEMA). Standard statistical methods were applied.
Results: Median white blood cell count at diagnosis was 6.7/nl (range, 0.1-151/nl)
and platelets 48.5/nl (range, 4-307/nl). Type of AML was de novo in 66 (68%), secondary
after myelodysplastic syndrome/ myeloproliferative neoplasm in 16 (16%), and therapy-related
in 9 (9%) patients (missing, n=6; 6%). Thirty-five (36%) patients were female. Cytogenetic
analysis revealed trisomy 19 as the sole abnormality in 10 (12.5%), additional abnormalities
in a non-complex karyotype in 8 (8%) and a complex karyotype in 79 (81.5%) patients.
Most frequent additional cytogenetic abnormality was trisomy 8 (n=46); in 17 patients
karyotypes were characterized by trisomies only. A total of 65 patients (67%) had
NPM1 and FLT3-ITD mutation testing. Of those, only 3 (5%) and 1 (2%) harbored NPM1
and FLT3-ITD mutations, respectively. Only 4 (8%) of 51 patients were CEBPA mutated.
Ninety-two patients (95%) were treated intensively and 4 (4%) received non-intensive
therapy (missing, n=1, 1%). In intensively treated patients early death rate was 10%
(n=9); CR was achieved in 52% (n=48) and 35% (n=35) patients were refractory. Factors
associated with response to intensive induction therapy were trisomy 19 as sole abnormality
or within a karyotype characterized by trisomies only (OR, 5.64; 95%-CI, 1.71-18.63;
P=0.005) and age at diagnosis (10 years difference OR, 0.57; 95%-CI, 0.40-0.80; P=0.001).
One of 4 patients treated non-intensively achieved a CR. An allo-HCT was performed
in 34 (35%) patients, of whom 19 patients were transplanted in first CR after induction
therapy. Type of donor was matched-related in 12 and matched-unrelated in 22 patients.
Median follow-up of the whole cohort was 6.4 years (95%-CI, 2.91-8.97 years). Five-year
overall (OS) and relapse-free survival rates were 20% (95%-CI, 13-31%) and 26% (95%-CI,
16-43%). OS rates were significantly higher in intensively treated patients with trisomy
19 as sole abnormality or within a karyotype characterized by trisomies only (P=0.05).
An Andersen-Gill model including allo-HCT as a time dependent covariable on OS revealed
as significant parameters trisomy 19 as sole abnormality or within a karyotype characterized
by trisomies only (HR, 0.47; 95%-CI, 0.25-0.89; P=0.021) and age at diagnosis (10
years difference; HR, 1.29; 95%-CI, 1.10-1.52; P=0.002), whereas allo-HCT had no beneficial
impact (HR, 1.45; 95%-CI, 0.81-2.59; P=0.21).
Summary/Conclusion: Patients with trisomy 19 are very heterogeneous in particular
with respect to cytogenetic and molecular abnormalities. In our cohort, patients with
trisomy 19 as sole abnormality or within a karyotype characterized by trisomies only
had a high CR rate and better clinical outcome. In the cohort of intensively treated
patients, allo-HCT did not improve OS.
P546: EFFECTS OF CHEMOTHERAPY DOSE REDUCTIONS IN OVERWEIGHT AND OBESE PATIENTS WITH
ACUTE MYELOID LEUKEMIA – A DANISH NATIONWIDE COHORT STUDY
D. Kristensen1,2,*, L. B. Nielsen1,2, L. H. Jakobsen1,3, T.-C. C. Kristensen1, T.
C. El-Galaly1,2, A. S. Roug1,2,4, M. T. Severinsen1,2
1Department of Hematology, Clinical Cancer Research Center, Aalborg University Hospital;
2Department of Clinical Medicine; 3Department of Mathematical Sciences, Aalborg University,
Aalborg; 4Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
Background: The majority of chemotherapeutic agents are dosed according to a body
weight derived variable. Studies in solid cancers have shown that overweight patients
frequently receive dose reduction (DR) of chemotherapy, despite no evidence corroborates
increased toxicity of full dosing. Rather, DR to ≤95% of actual weight-based dose,
has been shown to result in shortened overall survival (OS). Consequently, the American
Society of Clinical Oncology does not recommend up-front dose reduction based on body
mass index (BMI) or body surface area (BSA) in overweight patients. Current evidence
regarding DR and outcome among overweight patients with acute myeloid leukemia (AML)
receiving induction chemotherapy (IC) is limited.
Aims: The purpose of this study was to investigate the association between DR and
outcome in overweight patients with AML.
Methods: We utilized the Danish National Acute Leukemia Registry to conduct a retrospective
cohort study. Overweight (BMI≥25) AML patients aged 18-75 years and treated with IC
between 2000-2012 were included. We defined DR as ≤95% of actual BSA-based chemotherapy
dose. Relative risks (RR) for DR, complete remission (CR) rates, and 30- and 90-day
mortality were modeled, and OS and relapse-free-survival (RFS) were calculated and
compared using the 5-year restricted mean survival time difference (Δ5y-RMST).
Results: The study population included 536 overweight AML-patients of whom 54 patients
(10.1%) were categorized as DR (mean reduction 11.2%). Risk factors for DR in univariate
analysis were increasing BMI (30-34.9: RR, 2.52 [95% CI, 1.32-4.71]; ≥35: RR, 4.66
[95% CI, 2.37-8.91]), increasing BSA (2.0-2.2: RR 4.61 [95% CI, 1.96-12.6]; ≥2.2:
RR 15.21 [95% CI, 6.75-40.67]), therapy-related AML (RR 2.85 [95% CI, 1.12-7.24])
and favorable risk cytogenetics (RR 2.20 [95% CI, 1.02-4.33]). No significant differences
were observed for rates of CR, 30- and 90-day mortality between patients receiving
DR and non-DR IC. Dose reduction did not affect median RFS (DR, 14.5 [95% CI, 9.0
to 41.7] months; non-DR, 15.0 [12.3 to 19.3]) with an adjusted Δ5y-RMST of 0.2 (-8.4
to 8.8) months nor median OS (DR, 17.0 [11.9-45.5] months; non-DR, 17.5 [14.8-20.5])
with an adjusted Δ5y-RMST of 0.8 (-5.7 to 7.3) months (figure panel A+C). We constructed
a case-matched cohort matched on age, sex, AML subtype and BMI (figure panel B+D)
and performed a sensitivity analysis using ≤90% cut-off to define DR which led to
the same conclusions.
Image:
Summary/Conclusion: This study demonstrates that ~10% of Danish overweight AML-patients
treated with IC are dose reduced ≥5% compared to full BSA-based doses. Risk factors
were increasing BMI and BSA in addition to therapy-related AML and favorable cytogenetic
risk. Importantly, our results suggest that IC dose reduction does not adversely impact
AML outcomes including 30- and 90-day mortality, rates of CR, RFS and OS. However,
we encourage future prospective clinical studies to address this question with specific
and uniform standards or protocol specifications for dose reduction or dose capping
in overweight and obese patients with AML.
P547: CHARACTERISTICS AND OUTCOMES OF PATIENTS WITH ACUTE MYELOID LEUKAEMIA TREATED
WITH VENETOCLAX COMBINATION THERAPY: REAL-WORLD EXPERIENCE IN BOTH FRONTLINE AND RELAPSED/REFRACTORY
SETTINGS
H. P. J. Lam1,2,*, S. Leong1,2, Z. Kirkham1,2, A. Wilson1, R. Burt1, R. Sellar1, A.
Fielding1, R. Gupta1, E. Payne1, M. Mansour1, P. Kottaridis1, A. Khwaja1, J. O’Nions1,2
1Department of Haematology; 2NIHR UCL Clinical Research Facility, University College
London Hospitals NHS Foundation Trust, London, United Kingdom
Background: Venetoclax (Ven) in combination with hypomethylating agents, such as azacitidine
(Aza) and low dose cytarabine (LDAC) has been shown to be effective therapy in acute
myeloid leukaemia (AML) and has become standard of care for newly-diagnosed patients
unfit for intensive chemotherapy (DiNardo et al., 2020; Wei et al., 2019; Pollyea
et al., 2020). Efficacy has also been shown in the relapsed/refractory (R/R) setting
in more limited data sets (Báez-Gutiérrez et al., 2021; Pollyea et al., 2020, Stahl
et al., 2020; DiNardo et al., 2019). Ven combination therapy has become widely used
in newly-diagnosed patients in the UK since its approval during the COVID-19 pandemic
as an alternative to intensive chemotherapy and subsequently for patients unfit for
intensive therapy.
Aims: We describe the characteristics and outcomes of patients with AML or high risk
myelodysplastic syndrome (HR-MDS) receiving Ven combinations in frontline and R/R
settings to provide real-world insight into their use in UK clinical practice.
Methods: A retrospective analysis was performed of all patients with AML or HR-MDS
who received Ven combination therapy at University College London Hospital between
April 2020 and September 2021. Patient demographics, treatment history and bone marrow
results were obtained from electronic health care and laboratory records. Disease
stratification and response assessments were made as per European LeukemiaNet (ELN)
criteria (Döhner et al., 2017).
Results: At the time of analysis, 95 patients received Ven combinations (61 as frontline
treatment and 34 for R/R AML), with a median follow up of 14 months. The majority
of patients in both groups had adverse risk ELN classification (70.5% of frontline
patients, 64.7% of R/R) and received Ven-Aza (100% frontline and 91.1% R/R) (Table
1). The median ages were 72 and 59 years respectively. The incidence of composite
CR/CRi was 70.5% in the frontline setting, with median duration of response (DoR)
of 8.3 months and overall survival (OS) of 7.1 months. In R/R AML, the CR/CRi rate
was 64.7%, median DoR 10.5 months and median OS 9.8 months. Four out of the 43 patients
who achieved CR/CRi (9.3%) following frontline treatment and 9 of the 22 R/R (40.9%)
patients proceeded to allogeneic stem cell transplant (alloSCT) post induction. The
median survival for all patients who underwent alloSCT is not reached in this analysis.
The highest CR/CRi rates were observed in intermediate risk patients (90.9% in frontline
treatment, 71.4% in R/R), with lower rates in both favourable (80% and 66.7%) and
adverse risk patients (65.1% and 59.1% respectively). The presence of NPM1 and IDH1/2
mutations were associated with high CR/CRi rates in both the frontline (85.7% and
84.6% respectively) and R/R groups (100% and 81.8%), with below average response rates
seen in TP53 mutated AML (62% in frontline, 40% in R/R). Notable responses were seen
in patients with RUNX1 mutations in both settings (77.8% frontline, 66.6% R/R).
Image:
Summary/Conclusion: Our data describes real world effectiveness for venetoclax combinations
as both frontline and salvage therapy in UK clinical practice, similar to that seen
in clinical trials. This further contributes to our understanding of these therapies,
in particular their use as a viable treatment option in R/R patients and as a bridge
to alloSCT, and highlights the importance of further characterisation of genetic predictors
of response to inform treatment decisions in real-world practice.
P548: BEMCENTINIB COMBINED WITH LOW-DOSE CYTARABINE IS EFFICACIOUS AND WELL TOLERATED
IN RELAPSED AML PATIENTS UNFIT FOR INTENSIVE CHEMOTHERAPY. UPDATES FROM THE ONGOING
PHASE II TRIAL (NCT02488408)
S. Loges1,2,3,*, M. Heuser4, J. Chromik5, G. Sutamtewagul6, S. Kapp-Schwoerer7, M.
Crugnola8, N. Di Renzo9, R. Lemoli10, D. Mattei11, I. Ben-Batalla1,2,3, J. Waizenegger1,2,3,
L.-M. Rieckmann1,2,3, M. Janning1,2,3, C. D. Imbusch2, N. Beumer1,2,12, D. Micklem13,
C. Gorcea-Carson14, G. Lawson14, J. Nautiyal14, S. Deharo14, W. Fiedler15, Y. Alvarado-Valero16,
B. Gjertsen17
1DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim; 2Division
of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg;
3Department of Personalized Oncology, University Hospital Mannheim and Medical Faculty
Mannheim, University of Heidelberg, Mannheim; 4Hematology, Haemostasis, Oncology and
Stem Cell Transplantation, Hannover Medical School, Hannover; 5University Hospital
Frankfurt, Frankfurt, Germany; 6University of Iowa Hospitals and Clinics, Iowa City,
United States of America; 7University Hospital of Ulm, Ulm, Germany; 8University of
Parma, Parma; 9Haematology and SCT Unit, Vito Fazzi Hospital, Lecce; 10University
of Genoa, Genoa; 11ASOS. Croce e Carle, Cuneo, Italy; 12Faculty of Biosciences, Heidelberg
University, Heidelberg, Germany; 13BerGenBio ASA, Bergen, Norway; 14BerGenBio Ltd,
Oxford, United Kingdom; 15University Medical Center Hamburg, Hamburg, Germany; 16The
University of Texas M.D. Anderson Cancer Center, Houston, United States of America;
17Haukeland University Hospital, Bergen, Norway
Background: The new standard of care (SOC) in newly-diagnosed AML patients (pts) unfit
for intensive chemotherapy (IC) due to age or co-morbidities yields favourable efficacy.
However, beyond 1st line, these pts have limited treatment options, with a dismal
mOS of 2.4 months at relapse, highlighting a significant unmet need for new treatments
in this pt population. Bemcentinib (BEM) is a first-in-class, orally bioavailable,
highly selective AXL-inhibitor. AXL (a receptor tyrosine kinase) represents an important
target in AML, as its expression on AML cells and stem cell compartment is associated
with poor prognosis, resistance to chemotherapy and decreased antitumor immune response.
Aims: The ongoing BGBC003 PhII trial studied the safety and efficacy of the BEM+LDAC
combination in relapsed (REL) AML pts unfit for IC; in addition, a comprehensive translational
biomarker analysis was pursued. Here, we present preliminary efficacy and safety data
and include initial translational multiomics data from bone marrow mononuclear cells
(BMMNC).
Methods: Pts received BEM at 200 mg/OD with 3 loading doses at 400 mg/OD and LDAC
at SOC schedule. Study endpoints included overall survival (OS), objective response
(OR), clinical benefit rate (CBR) (OR+unchanged [UC]) and exploratory biomarker analyses.
Longitudinal BMMNC samples (n=32) from 13 pts underwent scRNA-seq and CITEseq (Chromium
10x genomics; TotalSeq, Biolegend). Pts were stratified by best response: CR, CRi,
PR for Responders; UC, PD for Non-Responders. Cell type annotations were inferred
from expression of known markers at RNA and protein level and for identification of
malignant cells copy number variation was included.
Results: As of 13 Dec 2021, 22 REL AML pts (10/22 in 1st relapse) were treated with
BEM+LDAC. Median age was 75.5yrs [66-86], median prior lines of therapy 1 [1-8] and
adverse cytogenetic risk in 7/22 (32%). Only 19/22 pts were evaluable for efficacy.
The overall REL population (n=19) demonstrated a composite CR (CRc=CR+CRi) of 26%
(5/19) and CBR of 79% (14/19); AML pts in 1st relapse (10/19) showed a CRc of 30%
(3/10) and CBR of 70% (7/10) whereas REL pts with time on treatment (ToT) > 3 months
(11/19) achieved CRc of 45% (5/11) and CBR 91% (9/11); median ToT of 12.9 months for
CR/CRi pts; 1 pt remains on treatment. Median OS was 6.2 months in the overall REL
pts (n=19), 7.4 months in the AML pts in 1st relapse (10/19) and 11.3 months in REL
pts with ToT >3 months. Late onset responses suggest an immunological mechanism of
action (iMOA) and may contribute to a longer ToT and survival.
CITEseq analysis identified differences in the immune compartment, underscoring an
iMOA associated with response to BEM+LDAC. Furthermore, increased gene set enrichment
of TNFα signalling via NfkB at screening in malignant blasts of responders emphasizes
the influence of the TNFα signalling pathway as an interesting field of further research.
The safety profile of BEM+LDAC in the REL AML pts (n=19) is comparable with the known
safety profile of LDAC. TRAEs of ≥G3 observed in ≥10% of pts were anaemia (33% BEM+LDAC;
22% BEM), and ECG QT prolonged (11% BEM+LDAC; 11% Bem). No G5 TRAEs reported.
Summary/Conclusion: BEM+LDAC is efficacious and well tolerated in REL AML pts unfit
for IC, with promising survival benefit compared to historical data. Translational
research showed activation of CD8+T cells and B/Plasma cells in response to treatment,
indicating that BEM elicits activation of two major adaptive immune cell populations
responsible for anti-AML immune responses. BEM+LDAC warrants further evaluation in
this population.
P549: ANALYSIS OF THE CLINICAL SIGNIFICANCE AND PROGNOSTIC IMPACT OF TET2 SINGLE NUCLEOTIDE
POLYMORPHISM I1762V IN PATIENTS WITH ACUTE MYELOID LEUKEMIA
Y. Li1, X. Lyu1,*
1Central Lab, Henan Cancer Hospital, Zhengzhou, China
Background: Acute myeloid leukemia (AML) is a malignancy that originates from myeloid
hematopoietic stem/progenitor cells in the bone marrow. The currently recognized cause
of AML is chromosomal abnormalities or genetic mutations in myeloid hematopoietic
stem/progenitor cells induced by various physicochemical factors such as benzene,
pesticides, ionizing radiation, and chemotherapeutic drugs. With the development of
high-throughput sequencing technology, a variety of acquired somatic mutations are
used for the diagnosis, treatment and prognostic assessment of AML. Also, genetic
susceptibility due to many single nucleotide polymorphisms (SNPs) is thought to be
important in the development of AML. The TET2 gene encodes a methylcytosine dioxygenase
that catalyzes the conversion of methylcytosine to 5-hydroxymethylcytosine and contributes
to the epigenetic regulation of myelopoiesis. Mutations in the TET2 gene lead to dysmethylation
and myeloid transformation. The TET2 SNP I1762V (rs2454206AG/GG, c.5284A>G, p.Ile1762Val)
has an allele frequency of approximately 21% in the Chinese population. Some studies
have shown that pediatric AML patients carrying mutations at this locus have a better
prognosis. In this study, we investigated the clinical significance of the TET2 SNP
locus I1762V in AML patients and its impact on prognosis through a comprehensive analysis
of high-throughput sequencing results from 413 AML patients.
Aims: This study aimed to explore the clinical significance and effect on the prognosis
of TET2 Single nucleotide polymorphism I1762V in patients with acute myeloid leukemia
(AML).
Methods: Target sequencing on 58 hematological tumor-related genes in bone marrow
samples of 413 AML patients was performed using the high-throughput sequencing method.
TET2 I1762V and other somatic mutations were annotated and compared with patients’
clinical information and prognosis.
Results: I1762V was detected in 154 patients with AML, which was significantly different
from the normal population in NyuWa Chinese Population Variant Database (χ2=72.4,
P<0.001). I1762V was not related to sex, age, and karyotype of AML patients (P>0.05).
The proportion of NPM1 gene mutations and KIT gene mutations in patients with I1762V
was significantly higher than the others(P < 0.001). NPM1 mutations and KIT mutations
were mutually exclusive. The survival analysis results showed that the OS and PFS
of AML patients with I1762V were significantly higher than those of wild type patients
(HR=0.55, P < 0.05), while the OS and PFS in AML patients with DNMT3A mutation (with
or without I1762V mutation) were lower than those of wild type patients (HR=1.79,
P < 0.05).
Summary/Conclusion: TET2 SNP I1762V is an factor affecting the prognosis of AML patients,
which can be used to guide the treatment and evaluate the prognosis of AML. TET2 SNP
I1762V influenced the concomitant or reciprocal mutations in AML patients; I1762V
was closely associated with the prognosis of AML patients. The findings demonstrate
that the TET2 SNP locus I1762V is an important guide for the diagnosis, treatment
and prognostic assessment of AML patients.
P550: A PHASE 3, RANDOMIZED TRIAL OF MAGROLIMAB IN COMBINATION WITH VENETOCLAX AND
AZACITIDINE IN PREVIOUSLY UNTREATED PATIENTS WITH ACUTE MYELOID LEUKEMIA INELIGIBLE
FOR INTENSIVE CHEMOTHERAPY (ENHANCE-3)
N. G. Daver1,*, K. Liu2, S. Werneke2, E. Rustia2, G. Ramsingh2, P. Vyas3
1The University of Texas MD Anderson Cancer Center, Houston; 2Gilead Sciences, Inc.,
Foster City, United States of America; 3Department of Medicine, University of Oxford
Radcliff, Oxford, United Kingdom
Background: Acute myeloid leukemia (AML) is an aggressive clonal hematopoietic malignancy
of myeloid cells associated with limited outcomes for patients who are ineligible
for intensive chemotherapy due to age or comorbidities. Magrolimab (Hu5F9-G4) is an
antibody blocking CD47, a “don’t eat me” signal on cancer cells, resulting in tumor
phagocytosis by macrophages. A triplet regimen of magrolimab + azacitidine + venetoclax
has shown promising activity in patients with AML.
Aims: To evaluate the efficacy, safety, and tolerability of magrolimab + azacitidine
+ venetoclax in previously untreated patients with AML who are ineligible for intensive
chemotherapy.
Methods: This is a phase 3, randomized, double-blind, placebo-controlled, multicenter
study. Approximately 432 patients will be randomized 1:1 to receive magrolimab + azacitidine
+ venetoclax (experimental arm) or placebo + azacitidine + venetoclax (control arm)
(Figure). Randomization will be stratified by age, cytogenetic risk group, and geographic
region. Patients are eligible if they are ≥75 years of age or 18 to 74 years of age
with specific comorbidities. Receipt of prior antileukemic therapy for AML is not
allowed. Magrolimab will be administered intravenously with an initial 1 mg/kg priming
dose on days 1 and 4 to mitigate on target anemia. The dose will then be escalated
to 15 mg/kg on day 8, then to 30 mg/kg on days 11 and 15, then weekly for 5 doses,
followed by every other week beginning 1 week after the fifth weekly 30 mg/kg dose.
Placebo will be dosed at the same frequency as magrolimab. Venetoclax will be administered
orally at 100 mg on day 1, 200 mg on day 2, and 400 mg daily starting on day 3 and
thereafter. Azacitidine will be given intravenously or subcutaneously at 75 mg/m2
on days 1 to 7, or days 1 to 5 and 8 to 9 of 28-day cycles. Patients will remain on
study until disease progression, relapse, loss of clinical benefit, or unacceptable
toxicities, or until other discontinuation criteria are met. The dual primary endpoints
are complete remission (CR) rate within 6 treatment cycles and overall survival. Secondary
endpoints include duration of CR, transfusion independence, and event-free survival.
Results: Trial in progress.
Image:
Summary/Conclusion: Patient enrollment is ongoing. Clinical trial information: NCT05079230.
P551: A PHASE 3, RANDOMIZED, OPEN-LABEL STUDY EVALUATING THE SAFETY AND EFFICACY OF
MAGROLIMAB IN COMBINATION WITH AZACITIDINE IN PREVIOUSLY UNTREATED PATIENTS WITH TP53-MUTANT
ACUTE MYELOID LEUKEMIA
N. G. Daver1,*, P. Vyas2, M. P. Chao3, G. Xing3, C. Renard3, G. Ramsingh3, D. A. Sallman4,
A. H. Wei5
1The University of Texas MD Anderson Cancer Center, Houston, United States of America;
2Weatherall Institute of Molecular Medicine, MRC Molecular Hematology Unit, University
of Oxford, Oxford, United Kingdom; 3Gilead Sciences, Inc., Foster City; 4Moffitt Cancer
Center, Tampa, United States of America; 5Department of Haematology, Alfred Hospital
and Monash University, Melbourne, Australia
Background: Magrolimab (Hu5F9-G4) is an antibody blocking CD47, a macrophage immune
checkpoint and “don’t eat me” signal on cancer cells, that eliminates leukemia stem
cells by inducing tumor phagocytosis. Hypomethylating agents synergize with magrolimab
by inducing “eat me” signals on leukemic blasts, thereby enhancing phagocytosis. Magrolimab
with azacitidine (AZA) has shown encouraging activity in frontline acute myeloid leukemia
(AML) unfit for intensive chemotherapy and myelodysplastic syndrome. In TP53-mutant
(TP53m) AML, it demonstrated an objective response rate of 69%, complete response
(CR) rate of 45%, and median overall survival (OS) of 12.9 months. The TP53 gene mutation
is observed in approximately 10-15% of newly diagnosed AML and is associated with
poor survival. The published first-line median OS in TP53m AML is 4-7 months, regardless
of whether patients are treated with intensive chemotherapy or non-intensive approaches,
such as a hypomethylating agent with venetoclax (VEN). AML patients harboring the
TP53 gene mutation represent a significant unmet medical need.
Aims: To evaluate the efficacy, safety, and tolerability of magrolimab+AZA vs physician’s
choice of VEN+AZA or “7 + 3” chemotherapy in patients with previously untreated TP53m
AML.
Methods: This is a phase 3, randomized, open-label, multicenter study. Approximately
346 patients will be randomized (1:1) to magrolimab+AZA (experimental arm) or physician’s
choice of VEN+AZA or 7 + 3 chemotherapy, based on patient fitness. Randomization will
be stratified by appropriateness for non-intensive vs intensive therapy, geographic
region (US vs non-US sites), and age (<75 vs ≥75 years). Patients are eligible if
they are ≥18 years old with histologically confirmed AML with no prior antileukemic
therapy, with at least one TP53 gene mutation that is not benign or likely benign
(confirmed by central laboratory) or biallelic 17p deletions (based on a locally evaluated
karyotype/fluorescence in situ hybridization report). During the first 28-day cycle,
patients randomized to magrolimab+AZA will receive magrolimab intravenously (IV) at
a priming dose of 1 mg/kg on days 1 and 4, 15 mg/kg on day 8, and 30 mg/kg on days
11, 15, and then weekly for 5 doses, followed by every other week beginning 1 week
after the fifth weekly 30 mg/kg dose. For patients randomized to the VEN+AZA arm,
VEN and AZA will be administered according to labeled indications. Patients treated
with 7 + 3 chemotherapy will receive 1-2 induction cycle(s) with IV daunorubicin or
idarubicin on days 1-3 and cytarabine 100 mg/m2 or 200 mg/m2 on days 1-7 followed
by up to 4 consolidation cycles with high-dose cytarabine (3000 mg/m2). Patients receiving
magrolimab+AZA or VEN+AZA will remain on treatment until disease progression, relapse,
loss of clinical benefit, unacceptable toxicities, or stem cell transplant. Patients
can undergo stem cell transplantation per investigator decision. The primary endpoint
is OS in patients appropriate for non-intensive therapy, and the key secondary endpoint
is OS in all patients.
Results: Trial in progress.
Summary/Conclusion: Patient enrollment is ongoing. Clinical trial information: NCT04778397.
P552: TREATMENT OUTCOMES IN NEWLY DIAGNOSED, UNTREATED PATIENTS WITH TP53-MUTATED
ACUTE MYELOID LEUKEMIA: A SYSTEMATIC LITERATURE REVIEW AND META-ANALYSIS
N. G. Daver1,*, S. Iqbal2, C. Renard2, R. J. Chan2, K. Hasegawa2, H. Hu2, P. Tse3,
J. Yan3, M. J. Zoratti3, F. Xie3, G. Ramsingh2
1The University of Texas MD Anderson Cancer Center, Houston; 2Gilead Sciences, Inc.,
Foster City, United States of America; 3McMaster University, Hamilton, Canada
Background:
TP53 mutations are present in 10% to 15% of patients with acute myeloid leukemia (AML)
and are associated with resistance to therapy and poor outcomes. Currently available
frontline therapies for TP53-mutated AML include intensive chemotherapy (IC), hypomethylating
agents (HMAs), and venetoclax combined with HMA (VEN+HMA).
Aims: To describe response and survival outcomes associated with IC, HMAs, and VEN+HMA
in newly diagnosed, untreated patients with TP53-mutated AML.
Methods: EMBASE and MEDLINE were systematically searched to identify prospective and
retrospective studies that reported complete remission (CR), CR with incomplete hematologic
recovery (CRi), median overall survival (OS), event-free survival (EFS), or duration
of response (DOR) outcomes in patients with TP53-mutated AML treated with IC, HMAs,
or VEN + HMA. Screening and data extraction were conducted independently and in duplicate
by 2 reviewers. Response outcomes (CR and CRi) were pooled using random-effects models,
and the median of medians method was used for time-related outcomes (OS, EFS, DOR).
Results: From the 3006 abstracts identified (May 20, 2021), 17 publications (12 studies:
6 randomized controlled trials, 2 single-arm trials, and 4 retrospective studies)
satisfied the inclusion criteria (Table). The percentage of patients achieving CR
was highest with IC (43%; 95% CI, 30%-56%; N=133), followed by VEN+HMA (33%; 95% CI,
22%-47%; N=54;) and HMA (21%; 95% CI, 7%-49%; N=14). The CRi rate was highest with
VEN+HMA (20%; 95% CI, 12%-33%; N=54), followed by IC (6%; 95% CI, 1%-20%; N=35). No
studies reported CRi outcomes for HMA alone. The rates of CR/CRi were 49% (95% CI,
37%-60%; N=121) for VEN+HMA, 46% (95% CI, 39%-53%; N=200) for IC, and 13% (95% CI,
2%-48%; N=28) for HMA alone. The median OS was similarly low across the 3 treatments:
6.5 months for IC (95% CI, 5.1-8.5 months; N=155), 6.2 months for VEN+HMA (95% CI,
5.2-7.2 months; N=73), and 6.1 months for HMA (95% CI, 4.9-7.2 months; N=34). The
median EFS was 3.7 months (95% CI, 1.6-5.7 months; N=133) with IC. The median DOR
was 3.5 months (N=35) with IC and 5.0 months (95% CI, 3.5-6.5 months; N=54) with VEN+HMA.
No studies reported EFS for HMA alone or VEN+HMA or DOR for HMA alone.
Table.
Outcomes in newly diagnosed, untreated adult patients with TP53-mutated AML treated
with IC, HMA, and VEN+HMA
ICstudies
Total, N
Outcome(95% CI)
HMAstudies
Total, N
Outcome(95% CI)
VEN+HMAstudies
Total, N
Outcome(95% CI)
CR rate
2
133
0.43(0.30-0.56)
1
14
0.21(0.07-0.49)
2
54
0.33(0.22-0.47)
CRi rate
1
35
0.06(0.01-0.2)
0
-
-
2
54
0.20(0.12-0.33)
CR/CRi rate
4
200
0.46(0.39-0.53)
2
28
0.13(0.02-0.48)
4
121
0.49(0.37-0.60)
Median OS, months
3
155
6.5(5.1-8.5)
2
34
6.1(4.9-7.2)
2
73
6.2(5.2-7.2)
Median EFS, months
2
133
3.7(1.6-5.7)
0
-
-
0
-
-
Median DOR, months
1
35
3.5(not reported)
0
-
-
2
54
5.0(3.5-6.5)
Summary/Conclusion: CR, CR/CRi, DOR, and median OS were modest across all treatments
for newly diagnosed, untreated patients with TP53-mutated AML. Limitations of this
analysis include low study and patient numbers for some interventions, the lack of
direct trial comparisons, and that CR/CRi outcomes were not reported separately for
DOR. The findings of this review highlight the significant unmet need for this very
difficult-to-treat population.
P553: A PHASE 2, OPEN-LABEL, MULTIARM, MULTICENTER STUDY TO EVALUATE MAGROLIMAB COMBINED
WITH ANTILEUKEMIA THERAPIES FOR FIRST-LINE, RELAPSED/REFRACTORY, OR MAINTENANCE TREATMENT
OF ACUTE MYELOID LEUKEMIA
P. Vyas1,*, N. G. Daver2, M. P. Chao3, G. Xing3, C. Renard3, G. Ramsingh3, A. H. Wei4,
D. A. Sallman5
1Weatherall Institute of Molecular Medicine, MRC Molecular Hematology Unit, University
of Oxford, MRC Molecular Haematology Unit and Oxford Biomedical Research Centre, University
of Oxford and Oxford University Hospitals, Oxford, United Kingdom; 2The University
of Texas MD Anderson Cancer Center, Houston; 3Gilead Sciences, Inc., Foster City,
United States of America; 4Department of Haematology, Alfred Hospital and Monash University,
Melbourne, Australia; 5Moffitt Cancer Center, Tampa, United States of America
Background: Magrolimab (Hu5F9-G4) is an antibody blocking CD47, a macrophage immune
checkpoint and “don’t eat me” signal on cancer cells. Magrolimab induces tumor phagocytosis
and eliminates leukemia stem cells. Chemotherapy and hypomethylating agents synergize
with magrolimab by inducing “eat me” signals on leukemic blasts, thereby enhancing
phagocytosis. Magrolimab+azacitidine (AZA) has shown encouraging clinical efficacy
with an objective response rate (ORR) of 63% and a complete remission (CR) or CR with
incomplete hematologic recovery (CRi) rate of 54% in first-line acute myeloid leukemia
(AML). Newly diagnosed AML patients (pts) who are ineligible for intensive chemotherapy
(IC) are incurable despite the progress made with AZA+venetoclax (VEN; median overall
survival [OS] 14-18 months), while AML pts with relapsed/refractory (R/R) disease
after intensive regimens have a dismal prognosis. Furthermore, for pts in remission,
maintenance therapy with oral AZA has improved OS and prevented relapse, although
relapse rates remain high. Magrolimab combinations are being evaluated in each of
these settings to improve the standard of care.
Aims: To evaluate the safety, efficacy, and tolerability of magrolimab combined with
antileukemia therapies in first-line pts ineligible for IC (cohort 1), R/R pts after
IC (cohort 2), and pts in CR/CRi with presence of minimal residual disease (MRD) after
IC (cohort 3).
Methods: This is an open-label, multiarm, multicenter study that includes 3 safety
run-ins with corresponding phase 2 cohorts. Pts in cohort 1 must be ≥75 years or 18-74
years with comorbidities that preclude IC. Pts in cohort 2 must have R/R AML after
initial IC. Pts in cohort 3 must have achieved a CR/CRi with MRD positivity, as assessed
by flow cytometry, after IC, and not be candidates for hematopoietic stem cell transplant.
Each cohort will initially enroll 6 pts for 1 cycle (28 days) to assess dose-limiting
toxicities and determine the recommended phase 2 dose (RP2D). Pts in cohort 1 will
receive a combination of magrolimab, VEN, and AZA. Pts in cohort 2 will receive magrolimab
in combination with mitoxantrone, etoposide, and cytarabine (MEC). Pts in cohort 3
will receive magrolimab with oral CC-486. All will receive magrolimab on the same
schedule. In cycle 1, magrolimab will be administered intravenously (IV) as priming
and ramp-up doses at 1 mg/kg on days 1 and 4, 15 mg/kg on day 8, 30 mg/kg on days
11, 15, and then weekly for 5 doses, followed by every other week beginning 1 week
after the fifth weekly 30 mg/kg dose. For cohort 1, AZA (75 mg/m2) will be administered
IV/subcutaneously on days 1-7 or 1-5, 8, and 9 of each cycle. VEN, MEC, and CC-486
will be administered according to labeled indications. After completion of the safety
run-in, additional pts will be enrolled in cohort 1 (n=40), cohort 2 (n=30), and cohort
3 (n=40) for phase 2. In phase 2, pts in cohorts 1 and 3 will receive study treatment
at RP2D until disease progression, unacceptable toxicity, or loss of clinical benefit.
Pts in cohort 2 will receive magrolimab with MEC for up to 3 cycles followed by magrolimab
alone for a total of 12 magrolimab cycles. In cohorts 1 and 2, the primary efficacy
endpoint is the CR rate. Secondary endpoints of interest include OS, ORR, and MRD-negative
CR/CRi rates. The primary efficacy endpoint in cohort 3 is the MRD-negative CR rate.
Results: Trial in progress.
Summary/Conclusion: Patient enrollment is ongoing. Clinical trial information: NCT04778410.
P554: GILTERITINIB VERSUS SALVAGE CHEMOTHERAPY FOR RELAPSED/REFRACTORY FLT3-MUTATED
ACUTE MYELOID LEUKEMIA: A PHASE 3, RANDOMIZED, MULTICENTER, OPEN-LABEL TRIAL IN ASIA
J. Wang1,*, B. Jiang2, J. Li3, L. Liu4, X. Du5, H. Jiang6, J. Hu7, M. Yuan8, T. Sakatani9,
T. Kadokura9, M. Takeuchi9, S. Izuka9, L. Girshova10, J. Tan11, S. Bondarenko12, L.
L. L. Wong13, A. Khuhapinant14, E. Martynova15, N. Hasabou16, R. Tiu16
1State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood
Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College,
Tianjin; 2Department of Hematology, Peking University International Hospital; 3Department
of Hematology, Peking Union Medical College Hospital, Beijing; 4Department of Hematology,
Shanghai Tongren Hospital, Shanghai; 5Department of Hematology, Guangdong Provincial
People’s Hospital, Guangzhou; 6Department of Hematology, Peking University Peoples
Hospital, Beijing; 7Fujian Institute of Hematology, Fujian Medical University Union
Hospital, Fuzhou, Fujian; 8Astellas Pharma, Inc., Beijing, China; 9Astellas Pharma,
Inc., Tokyo, Japan; 10Almazov National Medical Research Centre, St. Petersburg, Russia;
11Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Malaysia
Sarawak, Sarawak, Malaysia; Department of Hematology, Ampang Hospital, Selangor, Malaysia;
12Clinical Research Institution of Pediatric Hematology and Transplantation under
the Name of Raisa Gorbacheva, State Medical University, St. Petersburg, Russia; 13Hematology
Unit, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia; 14Division of Hematology,
Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University,
Bangkok, Thailand; 15Krasnoyarsk Regional Clinical Hospital, Krai, Russia; 16Astellas
Pharma, Inc., Northbrook, United States of America
Background: Due to poor prognosis, treatment options for patients (pts) with relapsed/refractory
to therapy (R/R) FLT3-mutated (FLT3
mut+) acute myeloid leukemia (AML) are needed globally. The phase 3 ADMIRAL trial
showed superior survival benefit and favorable safety for pts with R/R FLT3
mut+ AML receiving gilteritinib vs salvage chemotherapy (SC); randomized, controlled
trials of treatments in a mostly Asian population are lacking.
Aims: We evaluated the efficacy and safety/tolerability of gilteritinib vs SC in Asian
pts with R/R FLT3
mut+ AML after first-line therapy.
Methods: In this phase 3, open-label COMMODORE (NCT03182244) trial, adult pts in China,
Russia, Singapore, Thailand, and Malaysia with R/R FLT3
mut+ AML were randomized 1:1 to gilteritinib 120 mg orally per day or SC (low-dose
cytarabine; mitoxantrone/etoposide/intermediate-dose cytarabine; or fludarabine/high-dose
cytarabine/granulocyte colony-stimulating factor) over continuous 28-day cycles. Pts
had ECOG score ≤2; pts with acute promyelocytic leukemia, BCR-ABL–positive leukemia,
active central nervous system disease, or secondary AML were excluded. Primary endpoint
was overall survival (OS); key secondary efficacy endpoints were event-free survival
(EFS) and complete remission (CR). Other endpoints were duration of remission, composite
CR (CRc), and safety/tolerability. OS and EFS were analyzed with stratified Cox proportional
hazard models; response rates were analyzed with the Cochran-Mantel-Haenszel test.
Subgroup analyses were planned. Interim analysis results are presented.
Results: As of June 30, 2020, 234 pts were randomized (gilteritinib, n=116; SC, n=118).
Median age was 51.5 and 49.5 years in the gilteritinib and SC groups, respectively;
most pts had not received prior FLT3 inhibitors (87.9% and 93.2%). Baseline FLT3 mutations
in the gilteritinib vs SC groups were: FLT3-ITD (91.4% vs 83.1%), FLT3-TKD (6.0% vs
11.9%), and both FLT3-ITD and FLT3-TKD (2.6% vs 5.1%). Median OS follow-up duration
was 11.1 mo for gilteritinib and 6.9 mo for SC. Median OS was longer with gilteritinib
(9.0 mo) vs SC (4.7 mo; HR 0.549 [95% CI: 0.379, 0.795]; P=0.00126); 1-year survival
rate was 33.3% and 23.2%, respectively. OS benefit was seen with gilteritinib vs SC
across most subgroups (Figure). Pts on gilteritinib had longer EFS than pts on SC
(median EFS 2.8 vs 0.6 mo; HR 0.551 [95% CI: 0.395, 0.769]; P=0.00004). More pts had
CR on gilteritinib (16.4%) vs SC (10.2%; P=0.17690); CRc rates were 50.0% and 20.3%
(P<0.00001). Grade ≥3 adverse events (AEs) with gilteritinib (97.3%) vs SC (94.2%)
were comparable; serious AE rates were higher for gilteritinib (73.5%) vs SC (61.5%).
Adjusted for treatment exposure, AE rates were lower with gilteritinib (grade ≥3,
55.56 events/pt-year [E/PY]; serious, 6.19 E/PY) than SC (grade ≥3, 164.00 E/PY; serious,
12.40 E/PY). Most common AEs for gilteritinib were anemia (76.1%), thrombocytopenia
(46.9%), pyrexia (41.6%), and increased blood lactate dehydrogenase (41.6%); for SC,
most common AEs were anemia (64.4%), decreased white blood cell count (41.3%), and
thrombocytopenia (38.5%). AEs leading to death occurred in 22 (19.5%) and 15 (14.4%)
pts receiving gilteritinib or SC, respectively.
Image:
Summary/Conclusion: Gilteritinib significantly prolonged OS and EFS vs SC in pts with
R/R FLT3
mut+ AML in Asia. Safety/tolerability adjusted for treatment exposure was favorable
for gilteritinib vs SC. COMMODORE results further validate/affirm the clinical efficacy/safety
data from ADMIRAL, reinforcing the significant benefit of gilteritinib in R/R FLT3
mut+ AML.
P555: A PHASE 1B/2 STUDY OF THE CD123-TARGETING ANTIBODY-DRUG CONJUGATE PIVEKIMAB
SUNIRINE (IMGN632) IN COMBINATION WITH VENETOCLAX (VEN) AND AZACITIDINE (AZA) FOR
PATIENTS WITH CD123-POSITIVE AML
N. G. Daver1,*, P. Montesinos2, A. Aribi3, G. Martinelli4, J. Altman5, G. Roboz6,
E. S. Wang7, P. W. Burke8, D. Jeyakumar9, R. B. Walter10, D. J. DeAngelo11, H. P.
Erba12, A. Advani13, L. Gastaud14, X. Thomas15, E. Todisco16, N. Pemmaraju1, L. Mendez17,
A. de la Fuente18, G. Gaidano19, A. Curti20, N. Boissel21, C. Recher22, C. Schliemann23,
P. Vyas24, C. M. Sloss25, J. Wang25, K. A. Malcolm25, P. A. Zweidler-McKay25, K. L.
Sweet26
1Department of Leukemia, The University of Texas MD Anderson, Houston, United States
of America; 2Hospital Universitari i Politècnic La Fe, Valencia, Spain; 3Gehr Family
Center for Leukemia Research, City of Hope, Duarte, United States of America; 4Department
of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e
A. Seràgnoli”, Bologna, Italy; 5Division of Hematology and Oncology, Northwestern
University Feinberg School of Medicine, Chicago; 6Division of Hematology & Medical
Oncology, Weill Cornell Medicine/New York Presbyterian Hospital, New York; 7Roswell
Park Comprehensive Cancer Center, Buffalo; 8Department of Internal Medicine, Division
of Hematology/Oncology, University of Michigan, Ann Arbor; 9University of California
Irvine, Chao Family Comprehensive Cancer Center, Orange; 10Fred Hutchinson Cancer
Research Center, Seattle; 11Department of Medical Oncology, Harvard Medical School,
Dana-Farber Cancer Institute, Boston; 12Division of Hematologic Malignancies and Cellular
Therapy, Duke University, Durham; 13Taussig Cancer Institute, Cleveland Clinic, Cleveland,
United States of America; 14Medical Oncology Department, Antoine Lacassagne Hospital,
Nice; 15Department of Hematology, Hospices Civils de Lyon, Lyon-Sud Hospital, Lyon,
France; 16Division of Onco-Hematology, European Institute of Oncology IRCCS, Milano,
Italy; 17Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United
States of America; 18MD Anderson Cancer Center Madrid, Madrid, Spain; 19Division of
Hematology, Department of Translational Medicine, Università del Piemonte Orientale,
Novara; 20IRCCS Azienda ospedaliero-universitaria di Bologna, Istituto di Ematologia
“Seràgnoli”, Bologna, Italy; 21Université de Paris, Service Hématologie, Hôspital
Saint-Louis, Paris; 22Service d’Hématologie, CHU de Toulouse - Institut Universitaire
du Cancer Toulouse Oncopole, Toulouse, France; 23Department of Medicine A, Hematology,
Oncology and Pneumology, University Hospital Muenster, Muenster, Germany; 24MRC Molecular
Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford,
Oxford NIHR Biomedical Research Centre, and Oxford University Hospitals NHS Trust,
Oxford, United Kingdom; 25ImmunoGen, Inc., Waltham; 26Department of Malignant Hematology,
H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States of America
Background: Overexpression of CD123 occurs in multiple hematological malignancies,
including acute myeloid leukemia (AML), blastic plasmacytoid dendritic cell neoplasm
(BPDCN), acute lymphoblastic leukemia (ALL) and others. With limited expression on
normal hematopoietic progenitor cells, the CD123 antigen is an attractive target for
new therapeutics. IMGN632 is a CD123-targeting antibody-drug conjugate (ADC) comprising
a novel anti-CD123 antibody coupled, via a peptide linker, to a unique DNA-alkylating
cytotoxic payload of the IGN (indolinobenzodiazepine pseudodimer) class. In preclinical
models of AML, IMGN632 exhibited potent anti-leukemia activity, with a wide therapeutic
index. Confirming preclinical expectations, encouraging single-agent activity has
emerged for IMGN632 in the ongoing Phase I trial in patients with CD123-positive BPDCN
and AML (Daver. Blood (2019) 134 (Supplement_1):734.).
Preclinical data have demonstrated increased activity with the addition of IMGN632
to AZA alone and to AZA+VEN in multiple AML xenograft and PDX models, leading to improved
survival in these models (Kuruvilla. Blood (2019) 134 (Supplement_1):1375.). We have
reported compelling activity (ORR 59% and CCR rate 38%) of the IMGN632 triplet in
relapsed or refractory AML patients (Daver. ASH 2021 Abstract #372). The safety profile
of the triplet included rates of cytopenias similar to those observed with AZA+VEN;
additional low-grade adverse events included infusion-related reactions, dyspnea,
fatigue, gastrointestinal toxicities, electrolyte imbalances, and pneumonia.
Aims: This Phase 1b/2 study is designed to determine the safety, tolerability, and
preliminary anti-leukemia activity of IMGN632 when administered in combination with
AZA and VEN to patients with relapsed and frontline CD123-positive AML.
Methods: Dose escalation for the IMGN632+AZA+VEN triplet is complete. The Recommended
Phase 2 Dose levels are: IMGN632 45 mcg/kg given on day 7 with 14 days of VEN, and
either AZA 50 OR 75 mg/m2 for 7 days of a 28-day cycle.
Results: N/A
Summary/Conclusion: Phase 2 expansion cohorts for patients with untreated/frontline
and relapsed AML are enrolling to further characterize the safety profile and assess
the antileukemic activity. NCT04086264
P556: CHANGES IN HEALTH-RELATED QUALITY OF LIFE IN PATIENTS WITH NEWLY-DIAGNOSED ACUTE
MYELOID LEUKEMIA RECEIVING IVOSIDENIB + AZACITIDINE OR PLACEBO + AZACITIDINE
A. Schuh1,*, S. de Botton2, C. Recher3, S. Vives Polo4, E. Zarzycka5, J. Wang6, G.
Bertani7, M. Heuser8, R. Calado9, S.-P. Yeh10, J. Hui11, S. Pandya11, D. Gianolio11,
C. Chamberlain11, H. Dohner12, P. Montesinos13
1Princess Margaret Cancer Centre, Toronto, Canada; 2Institut Gustave Roussy, Villejuif;
3Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France; 4Hospital
Universitario Germans Trias i Pujol-ICO Badalona, Josep Carreras Research Institute,
Badalona, Spain; 5Department of Hematology and Transplantology, Medical University
of Gdansk, Gdansk, Poland; 6Institute of Hematology & Hospital of Blood Disease, Tianjin,
China; 7ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; 8Hannover Medical
School, Hannover, Germany; 9Ribeirão Preto School of Medicine, University of São Paulo,
São Paulo, Brazil; 10China Medical University, Taichung, Taiwan; 11Servier Pharmaceuticals
LLC, Boston, United States of America; 12Ulm University Hospital, Ulm, Germany; 13Hospital
Universitari i Politècnic La Fe, València, Spain
Background: Ivosidenib (IVO) is a potent, targeted inhibitor of mutant isocitrate
dehydrogenase 1 (mIDH1) that is approved for acute myeloid leukemia (AML). IVO plus
azacitidine (AZA) demonstrated clinical benefit compared with placebo and AZA in the
AGILE study (NCT03173248).
Aims: To assess the impact of IVO+AZA versus placebo+AZA on health-related quality
of life (HRQoL) using data from the AGILE study.
Methods: In the double-blind, placebo-controlled phase 3 AGILE study, patients (pts)
were randomized 1:1 to IVO 500 mg QD + AZA 75 mg/m2 SC or IV for 7 days in 28-day
cycles, or placebo+AZA. HRQoL was a secondary endpoint assessed using two validated
questionnaires: the European Organisation of Research and Treatment of Cancer Core
Quality of Life Questionnaire (EORTC QLQ-C30) and the EuroQol 5-dimension 5-level
questionnaire (EQ-5D-5L). Questionnaires were administered pre-dose on cycle (C) 1
Day (D) 1, on C1D15, C2D1, C2D15, and on D1 of every odd cycle thereafter until the
end of treatment. Score change from baseline across visits for all subscales of EORTC
QLQ-C30 was analyzed with mixed models. A 10-point threshold in EORTC QLQ-C30 subscale
score was used to evaluate clinically meaningful changes from baseline or differences
between arms. Two-sided nominal p-values are reported. All patients gave written informed
consent.
Results: At baseline, 69 and 68 pts out of 72 receiving IVO+AZA completed the EORTC
QLQ-C30 and EQ-5D-5L, respectively, and 66 pts out of 74 receiving placebo+AZA completed
both. Mean baseline HRQoL scores were similar between treatment arms (EORTC QLQ-C30
global health status [GHS/QoL] score was 56.3 and 53.2 in the IVO+AZA and placebo+AZA
arms, respectively). There was an initial decline in HRQoL (EORTC QLQ-C30 GHS/QoL)
in both arms for ~4 months, consistent with time to response, and which was generally
not clinically meaningful (Table). IVO+AZA was associated with preserved or improved
HRQoL compared to baseline for most subscales of the EORTC QLQ-C30 from C5 to C19
(after which no placebo+AZA HRQoL data were available), and at most timepoints for
EQ-5D-5L VAS scores and index values. EORTC QLQ-C30 subscales with clinically meaningful
improvements from baseline at most timepoints from C5 to C19 in the IVO+AZA arm included
GHS/QoL (Table), fatigue, pain and appetite loss. In contrast, there were few clinically
meaningful improvements from baseline in placebo+AZA pts. GHS/QoL scores were significantly
improved (p≤0.05) for IVO+AZA versus placebo+AZA at C2D1, C2D15, C7 and C9 (Table),
and differences between treatment arms were clinically meaningful at C2D1, C2D15,
C7, C9, C13, C15 and C19 (Table). Likewise, improvements in EORTC QLQ-C30 fatigue,
appetite loss, nausea and vomiting, diarrhea, cognitive functioning and social functioning
favored IVO+AZA over placebo+AZA at multiple timepoints.
Image:
Summary/Conclusion: Data from the AGILE study show that patients with mIDH1 AML receiving
treatment with IVO+AZA tended to report maintenance or improved HRQoL from cycle 5
through to cycle 19 compared with placebo+AZA.
P557: HEMATOLOGIC IMPROVEMENTS WITH IVOSIDENIB + AZACITIDINE COMPARED WITH PLACEBO
+ AZACITIDINE IN PATIENTS WITH NEWLY-DIAGNOSED ACUTE MYELOID LEUKEMIA
H. Dohner1,*, P. Montesinos2, S. Vives Polo3, E. Zarzycka4, J. Wang5, G. Bertani6,
M. Heuser7, R. Calado8, A. Schuh9, S.-P. Yeh10, A. de la Fuente11, C. Cerchione12,
S. Daigle13, J. Hui13, S. Pandya13, D. Gianolio13, C. Recher14, S. de Botton15
1Ulm University Hospital, Ulm, Germany; 2Hospital Universitari i Politècnic La Fe,
València; 3Hospital Universitario Germans Trias i Pujol-ICO Badalona, Josep Carreras
Research Institute, Badalona, Spain; 4Department of Hematology and Transplantology,
Medical University of Gdansk, Gdansk, Poland; 5Institute of Hematology & Hospital
of Blood Disease, Tianjin, China; 6ASST Grande Ospedale Metropolitano Niguarda, Milan,
Italy; 7Hannover Medical School, Hannover, Germany; 8Ribeirão Preto School of Medicine,
University of São Paulo, São Paulo, Brazil; 9Princess Margaret Cancer Centre, Toronto,
Canada; 10China Medical University, Taichung, Taiwan; 11MD Anderson Cancer Center
Madrid, Madrid, Spain; 12Hematology Unit, Istituto Scientifico Romagnolo per lo Studio
e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy; 13Servier Pharmaceuticals, Boston,
United States of America; 14Institut Universitaire du Cancer de Toulouse Oncopole,
Toulouse; 15Institut Gustave Roussy, Villejuif, France
Background: Ivosidenib (IVO) is a potent oral targeted inhibitor of mutant isocitrate
dehydrogenase 1 (mIDH1). IVO plus azacitidine (AZA) significantly improved event-free
survival (EFS), overall survival and complete remission + partial hematologic recovery
rates compared with placebo + AZA, in patients (pts) with newly diagnosed IDH1-mutant
acute myeloid leukemia (AML) in the Phase 3 AGILE trial (NCT03173248).
Aims: To report blood count recovery results from the AGILE trial.
Methods: Pts were randomized 1:1 to IVO 500 mg QD + AZA 75 mg/m2 SC or IV for 7 days
in 28-day cycles (n=72), or placebo+AZA (n=74). Red blood cell (RBC)/platelet transfusion
history were assessed at screening and follow-up. Bone marrow (BM) and peripheral
blood samples were obtained at screening, and during weeks 9, 17, 25, 33, 41, 53,
and every 24 weeks thereafter, and at end of treatment and during EFS follow up. Samples
were analyzed at each local site according to ICSH guidelines. All patients gave written
informed consent.
Results: In the IVO+AZA and placebo+AZA arms, 4.2% and 5.5% of pts, respectively,
received concomitant granulocyte colony-stimulating factor. Hemoglobin levels steadily
increased from baseline at a similar rate in both treatment arms. Mean platelet count
recovered from baseline values in the IVO+AZA and placebo+AZA arms (71.0 and 92.6
x 109/L, respectively) as early as week 9 of treatment (171.1 and 155.1 x 109/L, respectively)
and continued to steadily increase thereafter in the treated population. In pts receiving
IVO+AZA, mean neutrophil counts rapidly increased from baseline (0.99 x 109/L) to
week 2 (2.05 x 109/L) and week 5 (4.07 x 109/L), and then generally stabilized to
within the normal range to study end (last available cycle value; ~2.0 x 109/L). Mean
neutrophil counts initially declined with placebo+AZA before slowly recovering to
near-normal levels after 36-40 weeks. The increased blood counts were accompanied
by a rapid decrease in the mean BM blast percentage from 54.8% at baseline to 12.0%
and 7.2% at week 9 and 17, respectively, in IVO+AZA treated patients and were maintained
for 149 weeks. The decline in BM blasts was slower in the placebo+AZA arm (53.7%,
34.6% and 19.6% at baseline, week 9 and week 17, respectively). Among patients who
were RBC/platelet transfusion-dependent at baseline (~54.0% in both groups), 46.2%
in the IVO+AZA group achieved RBC/platelet transfusion independence compared with
17.5% in the placebo+AZA arm (1-sided p=0.0032). Additionally, fewer adverse events
of febrile neutropenia (28.2% vs 34.2%) and infections (28.2% vs 49.3%) were reported
in the IVO+AZA arm compared to the placebo+AZA arm.
Summary/Conclusion: IVO+AZA demonstrated a significant clinical benefit compared with
placebo+AZA and this sub-analysis demonstrated a rapidly improved recovery of blood
counts and a reduced dependence on RBC and/or platelet transfusion. Moreover, rates
of febrile neutropenia and infections were reduced with IVO+AZA.
P558: GERMAN AMLCG-SURVIVORSHIP STUDY – SOMATIC LONG-TERM CONSEQUENCE OF AML AND ITS
THERAPY: FROM HEART TO KIDNEY.
A. S. Moret1,*, E. Telzerow1, C. Sauerland2, M. Rothenberg-Thurley1, F. H. A. Mumm1,
J. Braess3, B. Wörmann4, U. Krug5, W. E. Berdel6, W. Hiddemann1, K. Spiekermann1,
P. Heußner7, K. Kraywinkel8, D. Görlich2, K. H. Metzeler9
1Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University
Hospital, LMU Munich, Munich; 2Institute of Biostatistics and Clinical Research, University
of Münster, Münster; 3Department of Oncology and Hematology, Hospital Barmherzige
Brüder, Regensburg; 4Charité University Hospital Berlin, Berlin; 5Department of Medicine
3, Hospital Leverkusen, Leverkusen; 6Department of Medicine A, University of Münster,
Münster; 7Hospital Garmisch-Partenkirchen, Garmisch-Partenkirchen; 8Department of
Epidemiology and Health Monitoring, Robert Koch Institute, Berlin; 9Department of
Hematology, Cell Therapy and Hemostaseology, University Hospital Leipzig, Leipzig,
Germany
Background: As outcomes of patients with acute myeloid leukemia (AML) have improved,
the fraction of patients surviving long-term are increasing. Information on somatic
and psycho-social health consequences of AML and its treatment is sparse.
Aims: Our aim was to perform a multi-dimensional analysis of health outcomes in AML
long-term survivors (AML-LTS). This report focuses on somatic morbidity; overall and
health-related quality of life are reported separately (Telzerow et al.).
Methods: We conducted a cross-sectional study including AML survivors who had been
enrolled in clinical trials or the patient registry of the AML-CG study group and
were alive ≥5 years after initial diagnosis. Data concerning somatic health status
were collected through patient questionnaires, assessment by the patients’ physicians,
and medical and laboratory reports. An age- and sex-matched control cohort was derived
from German population-based health surveys (Robert Koch Institute, DEGS1 survey;
n=6013).
Results: Data on somatic health status was available for 355 survivors, aged 28-93
years, who participated 5 to 19 years after their AML diagnosis. Thirty-eight percent
of survivors were treated with chemotherapy with or without an autologous transplant,
whereas 62% had undergone allogeneic transplantation (alloSCT).
We compared the prevalence of somatic diseases between AML-LTS and the general German
population (DEGS1 sample), focusing on cardiovascular diseases and risk factors due
to their relevance for overall morbidity and mortality, and their known association
with survivorship in other cancers. Using age- and sex-adjusted multivariate models
(Figure A), we found that AML survivors had a 2-fold higher risk of type 1/2 diabetes
(DM 1/2), and a 3.5-fold increased risk of congestive heart failure (CHF) compared
to the general population. Prevalence of hypertension, coronary artery disease (CAD)
and myocardial infarction were similar between the groups. To identify factors associated
with development of CHF among AML-LTS, we constructed multivariate models incorporating
patient- and treatment-related covariables. We found an increased risk of CHF for
AML-LTS who have had AML relapse (OR, 3.16; 95% CI: 1.46 – 6.83; P=0.004) and, in
trend, for patients with sAML or tAML (OR 2.19; 95%CI: 0.92 – 5.22, P=0.076). Disease-
or treatment-related factors did not significantly associate with any of the other
comorbidities we studied.
Furthermore, AML-LTS had significantly impaired kidney function, as assessed by the
glomerular filtration rate (eGFR) estimated using the CKD-EPI-formula. Figure B displays
the median eGFR according to age groups for AML-LTS and DEGS1. For each age group
AML-LTS had significantly lower eGFR in comparison to DEGS1. We did not find an association
between kidney function and time since diagnosis (p=.5) and no differences between
AML-LTS treated by chemotherapy or alloSCT (p=.5), survived a relapse (p=.9) or were
diagnosed with de novo-AML vs. sAML/tAML (P=.6).
Image:
Summary/Conclusion: Compared to the German general population, AML-LTS had increased
risks of CHF and diabetes, but not hypertension or CAD. We identified AML relapse
as a risk factor for developing of CHF, suggesting that cumulative chemotherapy exposure
might be causally involved. Notably, AML-LTS who had undergone alloSCT did not have
increased risks of CHF, CAD, hypertension or diabetes, compared to survivors treated
with chemotherapy only. In addition, AML-LTS had a higher prevalence of impaired renal
function. We did not identify treatment- or disease-related factors that might cause
the lower kidney function in AML-LTS.
P559: HUMORAL RESPONSE TO MRNA-BASED COVID-19 VACCINE IN PATIENTS WITH MYELOID MALIGNANCIES
A. Mori1,*, M. Onozawa2, S. Tsukamoto1, T. Ishio1, E. Yokoyama1, K. Izumiyama1, M.
Saito1, H. Muraki3,4, M. Morioka1, T. Teshima2, T. Kondo1
1Blood Disorders Center, Aiiku Hospital; 2Department of Hematology, Hokkaido University
Faculty of Medicine; 3Sapporo Clinical Laboratory Inc.; 4Division of Laboratory, Aiiku
Hospital, Sapporo, Japan
Background: The end of the pandemic of severe acute respiratory syndrome coronavirus
2 (SARS-CoV-2), the virus responsible for coronavirus disease-19 (COVID-19), is not
foreseen. Vaccination using two subtypes of mRNA-based vaccines, BNT162b2 or mRNA-1273,
is an effective public health measure to reduce the risk of infection and severe complications
from COVID-19. However, COVID-19 vaccine response data for patients with myeloid malignancy,
who are at severe risk in case of infection, has not emerged.
Aims: We investigated the antibody titers of COVID-19 in patients with myeloid malignancies
who received two doses of mRNA-based COVID-19 vaccine.
Methods: Previously treated, currently treated, and newly diagnosed 46 patients with
acute myeloid leukemia (AML) and 23 patients with myelodysplastic syndrome (MDS) were
included in this study. Anti-spike SARS-CoV-2 antibody titers were measured at 3 months
after the second vaccination and compared them to those in healthy controls.
Results: Seroconversion rates for AML and MDS were 94.7% and 100%, without significant
difference from healthy controls (100%). In AML patients, the median antibody titers
of patients in complete remission (CR) (816.5 [interquartile range (IQR): 250.0-2063.5]
U/ml vs 1023.0 [640.0-1535.0] U/ml, P=0.668), especially those who were under treatment-free
observation in CR (1630.0 [806.0-2454.0] U/ml vs 1023.0 [640.0-1535.0] U/ml, P=0.1220),
were comparable to those in healthy controls. On the other hand, even in CR, the antibody
titer in AML patients under maintenance therapy was significantly lower than that
in patients under treatment-free observation (154.0 [126.0-289.0] U/ml vs 1630.0 [806.0-2454.0]
U/ml, P=0.0003). Among the AML patients in CR, patients receiving maintenance treatment
had a significantly lower median absolute lymphocyte count (0.81 [0.71-1.46] x 109/l
vs 1.58 [1.29-1.93] x 109/l, P=0.0094) and a significantly lower median absolute neutrophil
count (1.45 [1.15-1.64] x 109/l vs 3.45 [2.68-4.28] x 109/l, P<0.0001) than that in
patients under treatment-free observation.
Significantly lower antibody titers were associated with current active treatment
(92.2 [37.5-216.3] U/ml vs 1630.0 [806.0-2454.0] U/ml, P<0.0001), AML with myelodysplasia-related
changes (50.8 U/ml [39.9-109.1] vs 816.5 [283.0-1935.3] U/ml, P=0.0022), advanced
age more than the median age of 68 years (195.0 [43.3-743.0] U/ml vs 1630.0 [806.0-3391.0]
U/ml, P=0.0002), and vaccine subtypes of BNT162b2 (285.0 [127.8-1045.3] U/ml vs 3037.0
[2198.50-4537.0] U/ml, P=0.0002) in AML patients and with current active treatment
(41.0 [10.7-227.5] U/ml vs 623.5 [173.8-1613.3] U/ml, P=0.0233), subtypes of excess
blasts (11.1 [4.8-34.1] U/ml vs 212.0 [81.7-600.0] U/ml, P=0.0293), and high and very
high risk of the revised international prognostic scoring system in MDS patients (9.0
[3.4-41.0] U/ml vs 169.0 [48.5-327.0] U/ml, P=0.0380).
Image:
Summary/Conclusion: This is one of the first studies on the effect of COVID-19 vaccines
focusing on patients with AML and MDS, and there are many new findings. The response
to COVID-19 vaccine appears to be related to disease and treatment status. Myeloid
malignancies may have less impact than lymphoid malignancies on the vaccine response.
AML patients under treatment-free observation in CR could be expected to have a vaccine
effect that is comparable to that in healthy individuals. In contrast, since the response
to vaccination might be insufficient in AML patients undergoing maintenance therapy,
maintenance therapy should be continued with strict measures for prevention of infection
even after vaccination.
P560: ANALYSIS OF GENE MUTATION SPECTRUM AND CORRELATION BETWEEN GENETIC ABNORMALITIES
IN 207 PATIANTS WITH T(8; 21) AML
Y. Nie1,*, Y. Li1, H. Zhang1, H. Li1, J. Zhang1, C. Wang1, Z. Chen1, S. Yang1, Y.
Lin1, K. Ru1
1Sino-us diagnostics, Tianjin, China
Background: The core binding factor (CBF) involves a subgroup of AML which is composed
of a and b subunits encoded by RUNX1(AML1) and CBFβ genes respectively, and CBF is
responsible for regulating multiple hematopoietic genes of myeloid differentiation.
AML with abnormal CBF accounts for 12%-15% of adult AML, including t(8;21)(q22;q22)
and inv (16)(p13q22), which produce RUNX1-RUNX1T1 (also known as AML1-ETO) and CBFβ-MYH11
fusion gene respectively.
Next generation sequencing(NGS) revealed that AML with t (8; 21) had other somatic
gene mutations at the same time. In addition to the known genetic abnormalities of
RAS and tyrosine kinase signaling pathways, the genes such as epigenetic regulation
showed recurrent genetic abnormalities. Some of these gene abnormalities, such as
KIT, JAK2 and FLT3-ITD mutations, have been reported in many studies and result in
poor prognosis. However, due to the relatively small sample or detected genes size,
there are some differences in the research conclusions. This study analyzed the molecular
correlation test results of 207 patients with t (8; 21) AML, and conducted a comprehensive
retrospective analysis by 56 gene targeted sequencing, so as to clarify the gene distribution
and related pathways involved in t (8; 21) AML patients in Chinese population, as
well as the correlation between different gene mutations and chromosome abnormalities.
Aims: To evaluate the gene mutational profile and its correlation with chromosomal
abnormalities in acute myeloid leukemia (AML) patients with t (8; 21).
Methods: The data of 207 patients with t(8;21) AML was retrieved from Sino-us diagnostics
lab from June 2015 to October 2021. We analyzed the gene mutational spectrum and characterized
the pathways of 56 AML-associated genes by NGS technology.
Results: Among these 207 patients with t(8;21) AML, the incidence of KIT(53%) gene
mutation was the highest, followed by NRAS (18%), ASXL2 (17%), FLT3 (12%), and EZH2
(10%). Most mutations were enriched in signal transduction, chromatin modification,
and transcriptional regulation pathways. According to their median VAF, we found that
DNA methylation genes, such as IDH1(50.8%), IDH2(49.0%), TET2 (43.8%) and DNMT3A (37.2%)
or Chromatin remodeling genes, like ASXL1 (42.1%), ASXL2 (37.3%) are present in the
majority of the cells. Meanwhile, alterations in RAS/RTK signaling genes like FLT3
(23.1%), KIT (32.5%) and the RAS GTPases NRAS (8.6%) and KRAS (13.3%) have the lower
VAF. In addition, most KIT mutations (121/154) were located in exon 17, mainly on
the SNV with D816 or N822. According to the correlation analysis, we found that IDH1/2
and complex karyotype (CK) or ZBTB7A mutation, ASXL2 and CCND2 mutation, t (x; 8;
21) abnormality and JAK2 or FLT3 mutation may co-mutate. On the contrary, KIT and
NRAS mutation were mutually exclusive.
Image:
Summary/Conclusion: From the patients with t(8;21) AML, most gene mutations occurred
in pathways involving signal transduction, chromatin modification, or transcriptional
regulation. DNA methylation or Chromatin remodeling genes may occur in the early stage
of leukemogenesis while RAS/RTK signaling genes may indicated later events. The coexistence
or mutual exclusion between genes and chromosomes may provide a basis for further
definitive diagnosis and therapeutic regimen.
P561: VIALE-T: A RANDOMIZED, OPEN-LABEL, PHASE 3 STUDY OF VENETOCLAX IN COMBINATION
WITH AZACITIDINE AFTER ALLOGENEIC STEM CELL TRANSPLANTATION IN PATIENTS WITH ACUTE
MYELOID LEUKEMIA
C. Craddock1,*, U. Platzbecker2, M. Heuser3, V. Pullarkat4, S. Chaudhury5, D. Wu6,
S. Addo7, B. Chyla7, Q. Jiang7, P. Lee7, J. E. Wolff7
1University of Birmingham, Birmingham, United Kingdom; 2University Hospital in Leipzig,
Leipzig; 3Hannover Medical School, Hannover, Germany; 4City of Hope, Duarte, CA; 5Ann
& Robert H. Lurie Children’s Hospital of Chicago, Northwestern Feinberg School of
Medicine, Chicago, United States of America; 6The First Affiliated Hospital of Soochow
University, Soochow, China; 7AbbVie Inc, North Chicago, United States of America
Background: Acute myeloid leukemia (AML) is an aggressive malignancy and is the most
common and second most common form of acute leukemia in adults and children, respectively.
The combination of the highly selective BCL-2 inhibitor venetoclax and the hypomethylating
agent azacitidine was shown to be safe and effective in clinical trials (DiNardo et
al. Blood. 2019;133:7-17; DiNardo et al. N Engl J Med. 2020;383:617-629) and is approved
by the United States Food and Drug Administration and European Medicines Agency for
the treatment of patients with AML who are not eligible to receive intensive chemotherapy.
Following allogeneic stem cell transplantation (alloSCT), most patients do not receive
antileukemic therapy; however, an unmet need remains as disease relapse and graft-versus-host
disease (GvHD) commonly occur posttransplant. In addition to the antileukemic effect
of venetoclax shown in clinical studies, preclinical studies suggest venetoclax may
mitigate the risk of GvHD. VIALE-T is a Phase 3, randomized, open-label trial in progress
(NCT04161885) evaluating the safety and efficacy of venetoclax in combination with
azacitidine versus best supportive care (BSC) as maintenance therapy following alloSCT
in patients with AML.
Aims: N/A
Methods: This Phase 3 study consists of 2 parts (Figure). Key inclusion criteria include
diagnosis of AML; plans to receive alloSCT or have received alloSCT within the past
30 days; bone marrow blasts <10% before pretransplant conditioning and <5% posttransplant;
have received myeloablative, or reduced intensity, or nonmyeloablative pretransplant
conditioning protocols. Grafts are allowed from various sources (bone marrow, peripheral
blood stem cells, cord blood cells). Patients must be ≥18 years old for Part 1 and
≥12 years old for Part 2. Additionally, patients must meet key laboratory values for
absolute neutrophil count (Part 1, ≥1500/µL; Part 2, ≥1000/µL), platelet count (Part
1, ≥80,000/µL; Part 2, ≥50,000/µL), bilirubin ≤3 times the upper limit of normal,
and creatinine clearance >30 mL/min. Patients who have received venetoclax and had
no history of disease progression while receiving venetoclax are eligible.
Part 1 evaluates dose levels of venetoclax combined with azacitidine to determine
the recommended Phase 3 dose (RP3D), which will be confirmed in approximately 12 additional
patients enrolled in the Safety Expansion Cohort. Part 2 will be a randomized, open-label
evaluation of the RP3D of venetoclax combined with azacitidine and BSC versus BSC
only in adults and children aged 12 years or older. All venetoclax-treated patients
will receive antibiotic prophylaxis during Cycle 1.
The primary endpoint for Part 1 is the frequency of dose-limiting toxicities. The
primary endpoint for Part 2 is relapse-free survival as assessed by an independent
review committee. Key secondary endpoints for Part 2 include overall survival, GvHD-free
relapse-free survival, and the rate of patients without higher grade GvHD at 90 days
after randomization. Enrollment into the Safety Expansion Cohort will be completed
in 2021. Part 2 will enroll approximately 400 patients across approximately 175 participating
study sites in 17 countries, with recruitment beginning in 2022.
Results: N/A
Image:
Summary/Conclusion: N/A
P562: LEMZOPARLIMAB (LEMZO) WITH VENETOCLAX (VEN) AND/OR AZACITIDINE (AZA) IN PATIENTS
(PTS) WITH ACUTE MYELOID LEUKEMIA (AML) OR MYELODYSPLASTIC SYNDROMES (MDS): A PHASE
1B DOSE ESCALATION STUDY
J.-Z. Hou1,*, N. G. Daver2, D. A. Stevens3, T. Yamauchi4, Y. Moshe5, C. Y. Fong6,
A. Marzocchetti7, R. Adamec8, M. Patel8, S. Lambert9, K. Y. Wu8, C. Röllig10
1UPMC Hillman Cancer Center, Pittsburgh, PA; 2Department of Leukemia, The University
of Texas MD Anderson Cancer Center, Houston, TX; 3Norton Cancer Institute, Louisville,
KY, United States of America; 4University of Fukui Hospital, Fukui, Japan; 5Tel Aviv
Sourasky Medical Center, Tel Aviv, Israel; 6Department of Clinical Haematology, Austin
Health, Heidelberg, Victoria, Australia; 7AbbVie S.r.l., Rome, Italy; 8AbbVie Inc.,
North Chicago, IL; 9AbbVie Inc., South San Francisco, CA, United States of America;
10Universitätsklinikum Carl Gustav Carus an der TU Dresden, Dresden, Germany
Background: Despite treatment advances, pts with AML or higher-risk MDS who are ineligible
for standard intensive treatments still have poor survival, highlighting the need
for novel treatments.
Overexpression of CD47 is common in leukemic stem cells and AML blasts and correlates
with poor clinical outcomes. Lemzo is an anti-CD47 antibody with red blood cell–sparing
properties. Treatment with ven plus aza has shown favorable safety and efficacy in
older/unfit pts with AML and higher-risk MDS. Blocking CD47 is hypothesized to hypersensitize
AML cells to the antitumor activity of ven and aza.
This study will evaluate the safety and dose-limiting toxicities (DLTs) of lemzo with
ven + aza for pts with treatment-naïve AML, as well as lemzo with aza ± ven for treatment-naïve
higher-risk MDS.
Aims: N/A
Methods: This phase 1b, open-label, dose-escalation study (NCT04912063) is enrolling
adults with: (1) treatment-naïve AML with adverse cytogenetic/molecular risk not suitable
for induction therapy, with an Eastern Cooperative Oncology Group Performance Status
(ECOG-PS) 0–2 (aged ≥75 years) or 0–3 (aged ≥18–74 years) excluding acute promyelocytic
leukemia; or (2) treatment-naïve higher-risk MDS (Revised International Prognostic
Scoring Score >3) with <20% bone marrow blasts, ECOG-PS 0–2, and no immediately planned
stem cell transplant.
For each 28-day cycle, aza is administered subcutaneously or intravenously (IV) daily
for 7 days within the first 9 days (7-0-0 or 5-2-2 schedule); ven is administered
orally daily on days 1–28 (AML, following dose ramp-up) or days 1–14 (ven-containing
MDS cohorts). Lemzo is administered IV at a schedule that is to be determined in this
study.
Dose escalation has Bayesian optimal interval design and may be expanded to investigate
alternate dosing for lemzo. Dose expansion will initiate at recommended phase 2 dose.
Treatment discontinuation criteria are unacceptable toxicity, progressive disease,
lack of partial/complete remission or clinical benefit within 6 cycles, or at physician’s
discretion. Pts who discontinue study treatment without progression will continue
with posttreatment follow-up; pts who progress will enter survival follow-up.
The primary endpoints are DLTs of lemzo. Secondary endpoints for both cohorts include
best overall responses of complete remission, duration of response, event-free survival,
and overall survival. Exploratory biomarker endpoints are included. Safety assessments
include adverse event (AE, graded per National Cancer Institute Common Terminology
Criteria for AEs v5.0) monitoring, physical examinations, vital signs, electrocardiograms,
and laboratory tests.
Response rates will be analyzed with estimates and 95% confidence intervals based
on exact binomial distribution. Time-to-event endpoints will be analyzed using Kaplan–Meier
methodology.
Results: N/A
Summary/Conclusion: N/A
P563: A RANDOMIZED, DOUBLE-BLIND, 2-ARM, MULTICENTER, PH3 STUDY OF VENETOCLAX & ORAL
AZACITIDINE VS. ORAL AZACITIDINE AS MAINTENANCE THERAPY FOR PTS WITH AML IN FIRST
REMISSION AFTER INTENSIVE CHEMOTHERAPHY
V. Ivanov1,*, S.-P. Yeh2, J. Mayer3, L. Saini4, A. Unal5, M. Boyiadzis6, D. M. Hoffman7,
K. Kang7, S. N. Addo7, W. L. Mendes7, A. T. Fathi8
1Almazov National Medical Research Centre, Saint Petersburg, Russia; 2China Medical
University Hospital, Taichung City, Taiwan; 3Fakultni Nemocnice Brno, Lískovec, Czechia;
4London Health Sciences Center, Ontario, Canada; 5Erciyes University Medical School,
Kayseri, Turkey; 6Genentech Inc., South San Francisco, CA; 7AbbVie Inc, North Chicago,
IL; 8Massachusetts General Hospital, Boston, MA, United States of America
Background: Acute myeloid leukemia (AML) is an aggressive, heterogenous hematologic
malignancy with poor prognosis. For eligible patients, standard treatment includes
intensive chemotherapy during induction, followed by consolidative chemotherapy or
stem cell transplantation. Despite current therapies, the prevention of relapse is
still a major therapeutic challenge and an unmet need for patients in remission.
Venetoclax (Ven) is a selective, potent, oral BCL-2 inhibitor that induces apoptosis
in AML cells. Ven in combination with Azacitidine (Aza) leads to prolonged overall
survival (OS) and rapid, durable remissions in treatment-naïve AML patients ineligible
for intensive chemotherapy. The QUAZAR AML-001 (NCT01757535) trial showed that the
median OS in patients receiving maintenance oral Aza was superior to patients receiving
placebo following upfront induction and consolidation; this benefit was also observed
for patients who achieved either complete remission (CR) or CR with incomplete blood
count recovery (CRi).
VIALE-M is a randomized, double-blind, two-part study (NCT04102020) to determine the
recommended Phase 3 dose (RPTD) of Ven in combination with oral Aza (CC-486) that
can be safely administered as maintenance therapy, and to evaluate the safety and
efficacy of Ven in combination with oral Aza + BSC compared to placebo and oral Aza
+ BSC in patients with AML who have achieved CR or CRi after intensive induction and
consolidation.
Aims: N/A
Methods: The study is enrolling patients aged ≥18 years with newly diagnosed AML per
WHO 2016 classification, with confirmed CR or CRi following intensive induction and
consolidation therapies, from > 200 sites worldwide. Patients must have achieved first
CR or CRi (after induction) ≤120 days of first dose of study drug or ≤75 days past
last dose of last consolidation cycle, have intermediate or poor risk cytogenetics
per NCCN 2016 categorization, ECOG ≤2, and no history of acute promyelocytic leukemia
or active central nervous system involvement with AML. For the dose finding part of
the study, patients will receive Ven QD for up to 24 cycles and oral Aza QD on D1-14
of each 28-day cycle for up to 24 cycles to determine the RPTD. For safety expansion,
patients will receive Ven QD for up to 24 cycles and oral Aza QD on D1-14 of each
cycle for up to 24 cycles at the RPTD. For the randomization part, patients will be
randomized 1:1 to receive placebo or Ven QD at the RTPD and oral Aza QD on D1-14 of
each cycle for 24 cycles. In all parts, treatment may discontinue due to relapse/unacceptable
toxicity, or continue beyond 24 cycles under investigator’s discretion.
For the randomization portion, the primary endpoint is relapse-free survival; secondary
outcomes include OS, minimal residual disease conversion, and improvement in quality
of life.
Results: N/A
Summary/Conclusion: N/A
P564: FINAL RESULTS OF THE QOLESS AZA-AMLE RANDOMIZED TRIAL TO EVALUATE THE EFFICACY
OF 5-AZA FOR POST-REMISSION THERAPY OF ACUTE MYELOID LEUKEMIA IN ELDERLY PATIENTS
E. N. Oliva1,*, P. Salutari2, F. Di Raimondo3, G. Reda4, D. Capelli5, G. Iannì6, G.
Tripepi7, C. Alati1, C. Mammì8, M. G. D’Errigo8, P. Niscola9, C. Selleri10, P. Musto11,
E. Vigna12, A. Volpe13, N. Cascavilla14, M. C. Cannatà8, D. Mannina15, A. Candoni16
1U.O.C. di Ematologia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, Reggio
di Calabria; 2U.O. di Ematologia, Ospedale Civile di Pescara, Pescara; 3U.O.C. di
Ematologia, Policlinico Catania, Catania; 4U.O.C. di Ematologia, Fondazione IRCCS
Ca’ Granda Ospedale Maggiore Policlinico, Milano; 5Clinica di Ematologia, Ospedali
Riuniti di Ancona, Ancona; 6Dielnet SRL; 7Ifc-CNR; 8U.O.S.D. Genetica Medica, Grande
Ospedale Metropolitano Bianchi Melacrino Morelli, Reggio di Calabria; 9U.O. di Ematologia,
Ospedale Sant’Eugenio, Roma; 10Dipartimento di Oncoematologia, AOU San Giovanni di
Dio e Ruggi d’Aragona, Salerno; 11Department of Emergency and Organ Transplantation,
“Aldo Moro” University School of Medicine, and Unit of Hematology and Stem Cell Transplantation,
AOUC Policlinico, Bari; 12U.O. di Ematologia, Ospedale L’Annunziata, Cosenza; 13U.O.
di Ematologia, Azienda Ospedaliera San Giuseppe Moscato, Avellino; 14U.O. Ematologia,
Ospedale Casa Sollievo della Sofferenza IRCCS, San Giovanni Rotondo; 15U.O.C. di Ematologia,
Azienda Ospedaliera Papardo, Messina; 16Clinica Ematologica, Centro Trapianti e Terapie
Cellulari, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
Background: Elderly patients in complete remission (CR) after first-line therapy for
acute myeloid leukemia (AML) relapse within months unless additional therapy is given.
In elderly patients with AML in CR after intensive chemotherapy, the HOVON 97 study
reported on the efficacy and safety of azacitidine (5-Aza) post-remission therapy
and the randomized QUAZAR trial showed that CC-486 as maintenance therapy was associated
with significantly longer overall and relapse-free survival than placebo. We report
final results of the QoLESS AZA-AMLE Phase III, randomized, open label, Italian multicentre,
trial to evaluate the efficacy of 5-Aza for post-remission therapy of elderly AML
patients compared to best supportive care (BSC).
Aims: The primary endpoint is disease-free survival (DFS, from CR to relapse/death
at 2 and 5 years. Secondary endpoints are post-remission hospitalizations, overall
survival (OS) at 2 and 5 years and changes in quality of life (QoL).
Methods: Patients aged ≥ 61 years with untreated AML, “de novo” or evolving from MDS,
fit for intensive chemotherapy but ineligible for stem cell transplantation received
induction chemotherapy consisting of 2 courses of 3 + 7 (Daunorubicin 40 mg/m2 daily
days 1-3 and cytarabine 100 mg/m2 intravenous infusion days 1-7. Cases in CR received
cytarabine 800 mg/m2 3-hour infusion twice daily for 3 days. Patients in CR were randomized
to receive BSC or 5-Aza 50 mg/m2 for 7 days every 28 days and dose increase after
1st cycle to 75 mg/m2 for further 5 cycles, followed by cycles every 56 days for 4.5
years. QoL was assessed with QOL-E version 3 and EORTC QLQ-C30 version 3.0.
Results: 149 patients were enrolled to finally randomize 54 patients alive and in
CR (Table 1). After randomization, no patients died before relapse, 7 cases completed
the study while 43 subjects relapsed: at 2 years, 22 cases in BSC (median DFS: 6.0
months, 95% CI: 0.2–11.7) versus 18 in the 5-Aza arm (median DFS: 10.8 months, 95%
CI: 1.9-19.6; p=0.20); at 5 years, 1 subject on BSC withdrew consent and 23 subjects
relapsed (median DFS: 6.0 months, 95% CI: 0.2–11.7) while 2 subjects on 5-Aza withdrew
consent, 1 discontinued for bladder cancer and 20 relapsed (median DFS: 10.8 months,
95% CI: 1.9-19.6; p=0.23). Age modified the effect of the allocation arm on DFS. Data
adjustment for cytogenetic risk further amplified the efficacy of 5-Aza in patients
aged >68 years, both at 2 (HR: 0.24, 95% CI: 0.08-0.69, P=0.008) and 5 years (HR:
0.28, 95% CI: 0.10-0.76, P=0.012). The effect of 5-Aza versus BSC tended to be statistically
significant on DFS at 2 (P=0.053) and 5 years (P=0.068) in Cox models including cytogenetic
risk and MRD as covariates. Kaplan-Meier analyses of the effect of 5-Aza on DFS (at
2 and 5 years) showed that patients aged >68 years and positive MRD tended to have
a benefit in terms of DFS at both 2 and 5 years (P=0.056). In the 5-Aza arm, 5 subjects
out of 27 (19%) were hospitalised whereas no patient was hospitalised in the BSC arm
(P=0.023). QoL was similar in both arms after randomization. Twenty grade 3-4 adverse
events, mainly neutropenia, occurred in 5-Aza versus 1 in BSC arm (p=0.002).
Image:
Summary/Conclusion: 5-Aza is well tolerated in elderly patients with AML who have
achieved CR following standard chemotherapy. MRD remains an important predictor of
DFS in the long-term, independent of 5-Aza maintenance. No statistically significant
differences were observed between the two arms in terms of DFS, but after data adjustment
it is suggested that patients aged >68 years and positive MRD may benefit from 5-Aza
maintenance.
P565: IMPROVED OUTCOME OF PATIENS WITH ACUTE MYELOID LEUKEMIA HARBORING FLT3 MUTATION
IN THE ERA OF TARGETED THERAPY
G. Oñate1,2,*, M. Pratcorona1, A. Garrido1, A. Artigas1, A. Bataller3, M. Tormo4,
M. Arnan5, S. Vives6, R. Coll7, O. Salamero8, F. Vall-Llovera9, A. Sampol10, A. Garcia11,
M. Cervera12, S. Garcia Avila13, J. Bargay14, X. Ortin15, J. Esteve3, J. Sierra1
1Hematology, Hospital de la Santa Creu i Sant Pau; 2Universitat Autònoma de Barcelona;
3Hematology, Hospital Clínic, Barcelona; 4Hematology, Hospital Clínico Universitario,
Biomedical Research Institute INCLIVA, Valencia; 5Hematology, Institut Català d’Oncologia,
Hospital Duran i Reynals, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL),
Universitat de Barcelona, Hospitalet de Llobregat; 6Hematology, ICO-Hospital Germans
Trias i Pujol, Badalona; 7Hematology, ICO-Hospital Josep Trueta, Girona; 8Hematology,
Hospital Universitari Vall d’Hebron, VHIO; 9Hematology, Hospital Universitari Mútua
Terrassa, Barcelona; 10Hematology, Hospital Son Espases, Palma de Mallorca; 11Hospital
Universitari Arnau de Vilanova, Lleida; 12ICO-Hospital Joan XXIII, Tarragona; 13Hematology,
Hospital del Mar, Barcelona; 14Hospital Son Llàtzer, Palma de Mallorca; 15Hematology,
Hospital Verge de la Cinta, Tortosa, Spain
Background: In recent years, the treatment of AML is rapidly evolving due to the advances
in targeted therapy, risk-adapted protocols and MRD-guided decisions. The prognosis
of patients harbouring FLT3-ITD differs according to its allelic ratio and the presence
of NPM1 co-mutation (NPM1mut). Nonetheless, FLT3-ITD prognostic impact and allogeneic
hematopoietic cell transplantation (alloHCT) indication in this setting might be redefined
by the widespread use of FLT3 inhibitors. Since 2012, chemotherapy eligible patients
are treated following a risk-adapted and MRD-guided protocol in the CETLAM group (AML-12).
Since 2016, targeted therapy with midostaurin was progressively incorporated in the
protocol for FLT3-mutated (FLT3mut) patients.
Aims: To analyse the outcome of FLT3mut AML treated with a risk-adapted protocol in
two different time periods, mainly differentiated by the advent of midostaurin.
Methods: All adult patients with de novo AML and available FLT3 status treated with
the intensive protocol AML-12 (clinicaltrials.gov #NCT04687098) in the Spanish CETLAM
centres from 2012 to 2020 were retrospectively analysed. Molecular testing of NPM1,
and FLT3-ITD and its allelic ratio were studied as previously described. Patients
were divided into two study cohorts: the early (2012-2015) and the late cohort (2016-2020).
All patients received the same chemotherapy protocol except for the addition of midostaurin
since 2016 in most FLT3mut patients. Complete remission (CR), overall survival (OS),
event-free survival (EFS) and risk of relapse (RR) followed ELN criteria. OS and EFS
were studied with the Kaplan-Meier method and log-rank test, cumulative incidence
and Grey test were used to estimate relapse risk (CIR) and non-relapse mortality (NRM).
Results: A total of 906 patients were selected, 390 from the early and 516 from the
late cohort. Median follow-up was 38 months; FLT3mut was present in 227 cases (25%)
and constitute the focus of this study. There were not differences on main variables
between cohorts (age, cytogenetic risk, ELN category, frequency of NPM1 mutation or
FLT3-ITD allelic ratio). TKD mutations were only available since 2015. Midostaurin
was administered to 62% of FLT3mut patients from the late cohort: 63 with ITDs (20
low and 43 high ratio) and 20 TKDs. CR rates were similar between cohorts. AlloHCT
in first CR (CR1) was performed in 60% (early) and 67% (late) patients (p=ns). A higher
RR was observed in the early group in comparison to the late cohort (2-year RR 42±11%
vs 28±10%; p=0.014), without differences in NRM (Figure 1B), translating into an improved
EFS (median EFS 9.4 vs. 26 months, p=0.014) and OS in the late cohort (median OS:
15 months vs. non-reached, p=ns, Figure 1A). Among NPM1mut AML patients, FLT3-ITD
and ELN 2017 categories retained its prognostic value in the early cohort, with a
worse OS, EFS, and higher RR among FLT3-ITD vs wt patients (HR for OS 1.9 95% CI 1.15-3.2,
p=.011; figure 1C) and FLT3mut ELN-intermediate vs favourable patients (OS p<0.001;
figure 1D). On the contrary, in the late cohort, FLT3-ITD and FLT3 high allelic ratio
lost their adverse prognostic impact, with comparable RR, EFS, and OS regardless of
FLT3-ITD status (Figure 1E and F).
Image:
Summary/Conclusion: This real-life study demonstrated an improved outcome among FLT3mut
AML patients treated in the most recent period, probably reflecting the beneficial
effect of midostaurin on relapse prevention in this context. Future analyses should
revise the role of alloHCT in CR1 for non-favourable FLT3mut AML patients treated
with FLT3 inhibitors who achieve MRD eradication.
P566: IMPACT OF FRONTLINE INDUCTION APPROACH ON POST-STEM CELL TRANSPLANT (SCT) OUTCOMES
IN OLDER ADULTS WITH NEWLY DIAGNOSED ACUTE MYELOID LEUKEMIA (AML)
F. Ong1,*, F. Ravandi1, U. Popat2, T. M. Kadia1, N. Daver1, C. DiNardo1, M. Konopleva1,
G. Borthakur1, E. J. Shpall2, B. Oran2, G. Al-Atrash2, R. Mehta2, E. J. Jabbour1,
M. Yilmaz1, G. C. Issa1, G. Garcia-Manero1, A. Maiti1, H. A. Abbas1, R. E. Champlin1,
N. J. Short1
1Leukemia; 2Stem Cell Transplantation and Cellular Therapy, The University of Texas
MD Anderson Cancer Center, HOUSTON, United States of America
Background: A hypomethylating agent plus venetoclax (VEN) is standard of care for
older, unfit patients (pts) with newly diagnosed AML. However, the optimal induction
regimen for older pts with AML who are adequately fit for SCT is not well-established.
Aims: We investigated the post-SCT outcomes of older adult pts with newly diagnosed
AML based on the intensity of the induction regimen received.
Methods: This is a retrospective analysis of pts ≥60 years (yr) of age with newly
diagnosed AML who received allogeneic SCT after achieving first remission (CR, CRi
or MLFS) between 9/2012 and 7/2021 at our cancer center. Out of 127 pts, 44 pts received
intensive chemotherapy (IC) without VEN as induction therapy, 36 pts received low-intensity
therapy (LIT) without VEN, and 47 pts received LIT with VEN. Overall survival (OS),
relapse-free survival (RFS), cumulative incidence of relapse (CIR) and non-relapse
mortality (NRM) after SCT were compared among the groups.
Results: The baseline characteristics of the groups are shown in Table 1. Pts in the
IC cohort was significantly younger (median age of 63 yrs) than pts who received LIT
with or without VEN (median age of 68 yrs, p<0.01). Pts in the IC group were more
likely to have ECOG PS of 0 (34%) as compared to those who received LIT (14%, p=0.07).
Most pts had adverse ELN cytomolecular risk on presentation (43% in IC, 50% in LIT
without VEN, and 55% in LIT with VEN). The median numbers of cycles prior to SCT were
3 in all groups. Pts in LIT without VEN group were more likely to receive matched
unrelated donor (75%) than those in the LIT with VEN (49%) and IC groups (50%) and
were less likely to receive a haploidentical SCT (3% vs. 17% and 14%, respectively).
Pts in LIT with VEN group were more likely to receive reduced-intensity conditioning
(92%) prior to SCT, compared with 53% and 58% in the IC and LIT without VEN groups,
respectively (p<0.01). HLA matching was well-balanced in 3 groups. The majority of
pts were in CR/CRi prior to SCT (100% in the IC group, 95% in the LIT without VEN
group, and 92% in the LIT with VEN group), while the rest of pts achieved MLFS. MRD
negativity prior to SCT was achieved in more pts treated with IC (77%) or LIT with
VEN (66%), compared with LIT without VEN (49%). For the entire cohort, the median
follow-up was 37 months (mos) post-SCT. Post-SCT outcomes are shown in Figure 1. The
2-yr OS was highest in the LIT with VEN group (73%) when compared with the IC or LIT
without VEN groups (58% and 44%, respectively); OS was statistically superior with
LIT with VEN compared with LIT without Ven (p=0.02) and there was a trend towards
superior OS compared with IC (p=0.17). 2-yr RFS was also superior in the LIT with
VEN group (62%) as compared with the IC or LIT without VEN groups (54% and 42%, respectively).
2-yr CIR was highest in LIT without VEN group (36%), and was similar in the LIT with
VEN (19%) and IC groups (18%). 2-yr NRM was highest in IC group (27%) as compared
with the LIT with VEN (11%) and LIT without VEN groups (22%) (p=0.02 for IC vs. LIT
with VEN).
Image:
Summary/Conclusion: LIT with VEN resulted in comparable—and possibly superior—outcomes
compared with IC in SCT eligible patients ≥60 yrs with newly diagnosed AML. Despite
the older age and worse PS of the LIT with VEN group, their post-SCT NRM was lower
than those who received IC, which translated to numerically superior OS. These results
support LIT with VEN as an effective and tolerable induction strategy in older SCT-eligible
pts with newly diagnosed AML.
P567: CENTRAL NERVOUS SYSTEM INVOLVEMENT IN ADULT PATIENTS WITH ACUTE MYELOID LEUKEMIA
– INCIDENCE AND OUTCOME
M. Virijevic1,2,*, I. Djunic1,2, M. Mitrovic1,2, N. Pantic1, Z. Pravdic1, N. Sabljic1,
A. Novkovic3, M. Cvetovic2, J. Rajic1, A. Vidovic1,2, M. Todorovic-Balint1,2, N. Suvajdzic-Vukovic1,2
1Clinic of Hematology, University Clinical Center of Serbia; 2Faculty of Medicine,
University of Belgrade; 3Department of Hematology, Clinical Hospital Center “Zemun”,
Belgrade, Serbia
Background: Examination of central nervous system (CNS) involvement is not routine
diagnostic in adult patients with acute myeloid leukemia (AML). Therefore, its impact
on the disease course is not well defined. There are studies showing that CNS involvement
in AML is associated with a worse outcome, whereas others did not confirm such impact
on long-term survival.
Aims: To determine the incidence of CNS involvement, its impact on the disease free
and overall survival, and to define risk factors for CNS involvement.
Methods: This single center, retrospective study involved 645 adult patients with
nonpromyelocytic AML, diagnosed in period of 2013. and 2021. In patients with the
presence of CNS symptoms, with increased leukocyte count (≥30x109/L), FLT3 mutation,
monocyte phenotype, CD56 positivity, a lumbar puncture was performed. All cerebrospinal
fluid (CSF) samples were examined by flow cytometry (FCM). Treatment of CNS+ include
intrathecal CT: cytarabine 100 mg. Assessment of remission of CNS+ was performed after
8 i.t. CT. The following parameters were estimated as risk factors for CNS+ at diagnosis
of AML: age, WBC (<30x109/L vs. ≥30x109/L), ECOG PS, HCT CI score, elevated lactate
dehydrogenase (LDH), leukemia-related parameters (cytogenetics (including ELN risk
stratification), flow cytometry). The methods of descriptive and analytical statistics
were used. Univariate and multivariate Cox Proportional Hazards models were used to
identify risk factors.
Results: CNS involvement examination was performed in 272 patietns. A total of 55/272
(20.2%) patients had proven CNS involvement, while 217/272 (79.8%) were without neuroleukemia.
Complete remission was achieved by 168/272 (61.7%) of patients, but there was no statistically
significant difference between CNS positive and CNS negative patients (p=0.619). Patients
with CNS involvement at diagnosis had a significantly higher frequency of hyperleukocytosis
(≥30x109/L), age ≤50 years, French–American–British M5 subtype, expression of CD56
and CD15 antigen. Multivariate analysis identified CD56+ as the most important risk
factor for CNS+ at diagnosis in AML patients: p=0.003, HR 2.271; 95% confidental interval
(CI)= 1.320-3.908. There were no statistically significant differences in 5-year overall
survival and disease free survival between CNS positive and CNS negative patients.
Summary/Conclusion: Our study showed a high incidence of CNS involvement in AML patients,
compared to studies in which the incidence was significantly less (3.3%). Our study
confirmed the accuracy of the factor we used to assess CNS involvement. There was
no difference in the outcome between patients with CNS involvement and those without.
P568: REAL-LIFE EXPERIENCE OF TREATMENT REFRACTORY/RELAPSED ACUTE MYELOID LEUKEMIA
PATIENTS WITH VENETOCLAX COMBINATION THERAPY
H. Pomares Marin1,*, J. Villarreal Hernández2, M. Condom Esteve2, S. Vives Polo3,
M. Cervera Calvo4, R. Coll Jorda5, C. Maluquer Artigal1, G. Ibarra Fernández3, A.
Torrent Catarineu3, M. Galiano Barajas1, A. M. Sureda Balari1, M. Arnan Sangerman1
1Servei d’Hematologia, Institut Català d’Oncologia, Hospital Duran i Reynals, Institut
d’Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L’Hospitalet
de Llobregat; 2Servei d’Hematologia, Institut Català d’Oncologia, Hospital Duran i
Reynals, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de
Barcelona, L’Hospitalet de Llobregat; 3Servei d’Hematologia, Institut Català d’Oncologia,
Hospital Germans Trias i Pujol, Institut de Recerca contra la Leucèmia Josep Carreras,
Universitat Autònoma de Barcelona, Badalona; 4Servei d’Hematologia, Institut Català
d’Oncologia, Hospital Universitari Joan XXIII, Tarragona; 5Servei d’Hematologia, Institut
Català d’Oncologia, Institut d’Investigació Biomèdica de Girona (IDIBGI), Universitat
de Girona, Girona, Spain
Background: Venetoclax, a BCL-2 specific inhibitor, used in combination with azacitidine
in patients with newly-diagnosed acute myeloid leukemia (AML) who were ineligible
for intensive chemotherapy treatment, has shown high response rate and overall survival
compared with azacitidine monotherapy (Di Nardo et al. N Engl J Med 2020; 383:617-629).
Moreover, Venetoclax in combination with hypomethylating agents or with low-dose cytarabine
is being explored in other settings being frequently used in relapsed/refractory (R/R)
AML.
Aims: We performed a retrospective study of patients with R/R AML receiving venetoclax
combinations in the Catalan Institute of Oncology (ICO) in order to determine the
efficacy and safety of the combination.
Methods: We analyze 60 patients diagnosed with R/R AML at 4 hospitals belonging to
ICO in Spain, treated with venetoclax (400mg/24h; initial daily dose of 100mg with
a 3-day ramp-up to target dose of 400mg) in combination with hypomethylating agents
(Azacitidine 75mg/m2 7/28 days or Decitabine 20mg/m2 5/28 days) or low-dose cytarabine
(20mg/m2 10/28 days) from May 2019 until December 2021. Event was defined as death,
refractoriness to treatment or progressive disease.
Results: Characteristics of our cohort are described in Table 1. Notably, 31 (52%)
patients had high-risk AML according to ELN 2017 classification. Fourteen (23%) patients
received venetoclax in combination with decitabine, 34 (57%) patients with azacitidine,
and 12 (20%) patients with low-dose cytarabine. The median number of cycles received
was 3 (range 1-28), with a median of one cycle to achieve the best response (range:
1-4). Early mortality in the first 30 days was 15% (9 patients), 5 due to progression
disease, 2 due to infection and 2 due to clinical worsening. Overall response rate
after first cycle (Complete response (CR) + complete response without haematological
recovery (CRi) + partial response (PR)) was 58%. Three patients were treated in a
molecular-MRD positive status, and all of them achieved molecular response. Six of
19 patients (32%) were transitioned to allogeneic stem cell transplantation (alloSCT).
The median event-free survival and overall survival was 5.03 months and 7.2 months,
respectively. Response to treatment after 3-4 cycles, discriminate two groups of patients
with an OS of 12.66 months in those patients who achieved CR or PR vs 2.4 months in
non-responders (p0.000). Patients relapsed after alloSCT (7 patients) presented a
poor outcome (median OS was 1.6 months).
Image:
Summary/Conclusion: Our study showed that real-world experience of treating patients
with R/R AML with venetoclax in combination with hypomethylating agents or low-dose
cytarabine is feasible as salvage treatment in patients relapsed to hypomethylating
agent and as bridge therapy to alloSCT with a rapid response rate. Moreover, rapid
responses shown with the combination, allow us to identificate those patients who
may benefit from this approach.
P569: GILTERITINIB AND QUIZARTINIB IN RELAPSED/REFRACTORY (R/R) ACUTE MYELOBLASTIC
LEUKEMIA (AML) WITH FLT3 MUTATIONS: A REAL-LIFE EFFECTIVENESS AND SAFETY STUDY
D. Quintela1,*, M. Morgades1, A. Serrano2, M. Cervera3, A. Balerdi4, M. Díaz-Beyá5,
M. Arnan6, A. Garrido7, R. Coll8, M. Tormo9, J. López-Marin10, B. Merchan11, S. Garcia11,
M. Casado12, A. Sampol13, J. Esteve5, D. Martínez-Cuadrón2, J. Sierra7, M. Á. Sanz2,
J. M. Ribera1, P. Montesinos2, S. Vives1
1Clinical Hematology, ICO-Hospital Universitari Germans Trias i Pujol, Institut de
Recerca Josep Carreras, Badalona; 2Clinical Hematology, Hospital Universitari i Politècnic
La Fe, Valencia; 3Clinical Hematology, ICO-Hospital Joan XXIII, Tarragona; 4Clinical
Hematology, Hospital Universitario Cruces, Barakaldo; 5Clinical Hematology, Hospital
Clínic de Barcelona, Barcelona; 6Clinical Hematology, ICO-Hospital Duran i Reynals,
L’Hospitalet del Llobregat; 7Clinical Hematology, Hospital de la Santa Creu i Sant
Pau, Barcelona; 8Clinical Hematology, ICO-Hospital Josep Trueta, Girona; 9Clinical
Hematology, Hospital Clínico Universitario de Valencia, Valencia; 10Clinical Hematology,
Hospital General Universitario de Alicante, Alicante; 11Clinical Hematology, Hospital
del Mar, Barcelona; 12Clinical Hematology, Hospital Universitario de Badajoz, Badajoz;
13Clinical Hematology, Hospital Universitari Son Espases, Palma, Spain
Background: Patients with R/R AML have poor prognosis. FLT3 mutations worsened the
prognosis but offer a targeted therapy. FLT3 inhibitors have demonstrated efficacy
as monotherapy in FLT3-positive R/R AML.
Aims: The objective of the present study was to analyze the efficacy and safety of
Gilteritinib and Quizartinib in R/R FLT3-mutated AML in real-life context.
Methods: Between December 2016 and April 2021, 22 patients were treated with Gilteritinib
and 5 patients with Quizartinib in 13 centers of the Spanish PETHEMA and CETLAM groups.
Results: The median age was 62 years (range 25; 81) and 56% were women. Most patients
presented ITD FLT3 (80%), 56% were refractory and 44% were relapsed (CR1 duration
≥6 months in 60%). The 1st line therapy was intensive chemotherapy (78%, 38% with
midostaurin) or HMA (22%). A total of 13/27 patients (48%) had previously received
a FLT3 inhibitor (midostaurin=6, sorafenib=2, quizartinib=2, midostaurin + crenolanib
vs placebo=2 and midostaurin + quizartinib=1). Seventy percent of the patients received
>1 line of treatment prior to Gilteritinib/Quizartinib (the most frequent was FLAG-IDA).
Compared with diagnosis, patients in R/R had poorer functional status (ECOG<2 88%
vs. 73%) but lower leukocyte counts, LDH level and FLT3 ITD ratio (21.5 x109/L vs
4.7x109/L, 563 U/L vs 330 U/L and 0.60 vs 0.42, respectively). Three patients displayed
clonal evolution in R/R with acquisition of cytogenetic alterations in patients with
altered karyotype at diagnosis and appearance of alterations in patients with normal
karyotype. The ORR was 63% (21% CR, 21% CRi and 21% PR) and the median OS was 5.8
(95%CI, 3.8; 7.9) months. Time to achieve better response was 1.8 months (range 0.9;4.1).
Eight patients received the FLT3 inhibitor as a bridge to SCT, and six of them were
finally transplanted. Gilteritinib dose was 120mg/day, increasing to 200mg/d in 41%
of patients (due to lack of response after 28 days). Toxicity was observed in 69%
of patients (g3/4 in 30%). Febrile neutropenia was the most frequent toxicity (35%)
followed by hepatotoxicity (28%) and QTc prolongation (16%). One patient died of toxicity
attributed to Gilteritinib (febrile neutropenia).
Summary/Conclusion: Treatment with Gilteritinib and Quizartinib as monotherapy is
an effective and tolerable option for patients with R/R FLT3-mutated AML in real-life,
with similar response rates and toxicity to those reported in Phase 3 trials, despite
the fact that the study population from our series was more heterogeneous.
P570: REAL-WORLD EFFICACY OUTCOMES OF VENETOCLAX PLUS AZACITIDINE VS INTENSIVE CHEMOTHERAPY
FOR INDUCTION THERAPY IN ADULT PATIENTS WITH ACUTE MYELOID LEUKEMIA
A. M. Zeidan1,*, D. A. Pollyea2, U. Borate3, A. Vasconcelos4, R. Potluri5, D. Rotter5,
Z. Kiendrebeogo5, L. Gaugler4, G. Bonifacio4, T. Prebet4, C. Chen4
1Yale University School of Medicine, New Haven, CT; 2University of Colorado School
of Medicine, Aurora, CO; 3Oregon Health & Science University, Portland, OR; 4Bristol
Myers Squibb, Princeton, NJ; 5SmartAnalyst Inc., New York, NY, United States of America
Background: Intensive chemotherapy (IC) is commonly used to achieve remission in patients
with newly diagnosed acute myeloid leukemia (AML), although the relapse rate remains
high. The combination of venetoclax, a BCL2 inhibitor, plus intravenous or subcutaneous
azacitidine (ie, VEN-AZA), is FDA- and EMA-approved for the treatment of patients
with newly diagnosed AML aged ≥ 75 years or with comorbidities that preclude the use
of IC. Because of the challenging induction and maintenance decisions for some patients,
it is important to understand the real-world patterns of use and outcomes of VEN-AZA
vs IC.
Aims: To compare US real-world outcomes for the use of VEN-AZA vs IC, particularly
in older populations.
Methods: This retrospective analysis was performed on a cohort of patients from the
US-based Flatiron Health electronic health record-derived de-identified database,
who had newly diagnosed AML and received induction therapy with VEN-AZA or IC between
21 Nov 2018 and 01 Jun 2021. Using an intention-to-treat analysis (including patients
with less than 2 months of follow-up), patient groups were propensity score-matched
in a 1:1 ratio using nearest neighbor matching with calipers set to 0.01 to reduce
bias. Matched-patient assessments included morphological complete remission, overall
survival (OS; defined as from first day of therapy to death), and relapse-free survival
(RFS; from first day of therapy to relapse or death). Univariate analyses were used
to investigate possible treatment-associated OS and RFS benefit in patient subgroups,
including those with high-risk mutations.
Results: Of 7484 patients in the Flatiron database with newly diagnosed AML, 1188
met the selection criteria and 436 were propensity score-matched resulting in 218
per treatment group. In the matched VEN-AZA vs IC cohorts, patients had a mean age
of 69.9 vs 69.0 y and Charlson Comorbidity Index of 0.5 vs 0.7, respectively; 39%
vs 38% were female. The rates of patients with complete remission (CR) and those with
stem cell transplant (SCT) were significantly lower with VEN-AZA vs IC at 46% vs 62%
(P=0.001) and 16% vs 31% (P<0.001), respectively. Fewer VEN-AZA– vs IC-treated patients
received an early posttreatment bone marrow biopsy, with respective rates of 31% vs
69% (1 mo) and 57% vs 86% (2 mo). Median OS for VEN-AZA vs IC in the matched population
was 13.0 vs 17.2 mo (P=0.263), respectively; it was 11.0 vs 15.6 mo (P=0.410) with
censoring for SCT. Median RFS was numerically shorter for VEN-AZA than for IC (9.5
vs 11.0 mo; P=0.689); when including censoring for SCT, RFS was longer for VEN-AZA
(7.5 vs 5.7 mo; P=0.027). Among subgroups, including those with high-risk mutations,
no significant factor was found to be associated with OS in the matched patients (Figure).
The frequency of known patients with high-risk mutations in RUNX1, ASXL1, and TP53
was similar for both treatment groups. Similar subgroup results were observed for
RFS.
Image:
Summary/Conclusion: Rates of CR and SCT were significantly higher with IC. OS was
nominally longer with IC vs VEN-AZA, with and without censoring for SCT; differences
were not statistically significant. None of the subgroups tested, including patients
with high-risk mutations, showed a significant OS association. Longer follow-up time
may contribute to better characterized outcomes achieved with IC vs VEN-AZA in routine
clinical practice. These data support more investigation on strategies to improve
survival outcomes, including the impact of maintenance therapy.
P571: ORAL AZACITIDINE VS MIDOSTAURIN AS MAINTENANCE TREATMENT FOR FLT3 MUTANT ACUTE
MYELOID LEUKEMIA IN COMPLETE REMISSION: AN INDIRECT TREATMENT COMPARISON
E. N. Oliva1,*, C. Chen2, S. N. Rahim2, B. Seuro3, A. Vasconcelos2, L. Gaugler2, B.
Skikne2, T. Prebet2, H. Cameron3, C. Beach2, G. Thompson2
1Grande Ospedale Metropolitano Bianchi Melacrino Morelli, Reggio Calabria, Italy;
2Bristol Myers Squibb, Princeton, NJ; 3Eversana, San Diego, CA, United States of America
Background: Patients (pts) with FLT3-mutant (mut) acute myeloid leukemia (AML) have
a high risk of relapse, poor prognosis, and limited treatment (tx) options. A trend
toward improved survival with oral azacitidine (Oral-AZA) vs placebo (PBO) as maintenance
treatment (MT) was observed in pts in first complete remission (CR) after intensive
chemotherapy (IC) induction ± consolidation in an exploratory analysis of the phase
3 QUAZAR AML-001 study (QUAZAR; NCT01757535). Midostaurin (MIDO) plus chemotherapy
was FDA-approved as induction and consolidation tx for pts with FLT3-mut AML based
on results of the phase 3 RATIFY study (NCT00651261); absence of randomization prior
to MT makes inferences about the effectiveness of MIDO as MT difficult, although MIDO
was EMA-approved for MT. The relative efficacy of MT with Oral-AZA vs MIDO has not
been compared directly in clinical trials.
Aims: To compare the survival outcomes of MT with Oral-AZA vs MIDO in pts with FLT3-mut
AML who achieved first CR after IC (± consolidation tx) by performing an indirect
tx comparison (ITC) of data from QUAZAR and RATIFY.
Methods: An ITC of overall survival (OS) and relapse-free survival (RFS) comparing
Oral-AZA vs MIDO was performed, in which data from QUAZAR pts with FLT3-mut AML who
had achieved a CR with full blood count recovery (matching RATIFY eligibility criteria)
were compared with efficacy outcomes from the RATIFY MT phase (Larson RA, et al. Leukemia
2021). Primary analyses utilized the anchored Bucher method, which assumes the relative
tx effect is comparable across studies with similar effect modifiers. Time-varying
methods (parametric and spline models) were also explored to account for violation
of the proportional hazards (PH) assumption.
Results: Of 472 pts in the intent-to-treat population of QUAZAR, 56 pts with FLT3-mut
AML achieving a CR (Oral-AZA, 24; PBO, 32) were included in the primary analysis population,
as were all 205 RATIFY MT-phase pts (MIDO, 120; PBO, 85). In the QUAZAR and RATIFY
populations, respectively, mean age was 66.4 and 42.7 y, 55% and 50% were female,
36% and 29% had point mutations in the FLT3 tyrosine kinase domain, 0% and 57% had
favorable cytogenetic risk, and 0% and 5% underwent stem cell transplantation during
first CR. Bucher ITCs of both OS and RFS appeared to favor Oral-AZA over MIDO, but
were not statistically significant (Table). To assess age imbalance (QUAZAR pts were
≥ 55 y; RATIFY pts, 18–59 y), an exploratory analysis with QUAZAR pts (n = 9; age-matched
to RATIFY pts) showed OS and RFS results with Oral-AZA vs PBO were similar to those
of all QUAZAR pts. Evidence suggests the PH assumption had been violated in some of
these analyses, so best fitting parametric models were explored. Landmark OS and RFS
rates (generalized gamma model) were found to be higher with Oral-AZA than MIDO (Table).
Similar results were obtained using a spline model.
Image:
Summary/Conclusion: Despite more favorable prognostic factors among pts in RATIFY
(eg, younger age), the Bucher ITC analysis showed a trend toward longer OS and RFS
with Oral-AZA vs MIDO as MT for pts with FLT3-mut AML. Analyses that accounted for
the PH assumption violation also indicated a survival benefit for Oral-AZA vs MIDO.
Limitations included trial differences and small sample size. More research is needed
to support this observation.
P572: VENETOCLAX AND HYPOMETHYLATING AGENT COMBINATIONS FOR THE TREATMENT OF ADVANCED
MYELOPROLIFERATIVE NEOPLASMS AND ACUTE MYELOID LEUKEMIA WITH EXTRAMEDULLARY DISEASE
K. Sanber1,*, K. Ye2, H.-L. Tsai1, M. Newman3, A. Ambinder1, A. DeZern1, T. Jain1
1Department of Oncology; 2School of Medicine; 3Department of Pharmacy, Johns Hopkins
University, Baltimore, United States of America
Background: Progression of myeloproliferative neoplasms (MPN) and myelodysplastic/myeloproliferative
neoplasms (MDS/MPN), into accelerated phase (AP) or blast phase (BP) disease is associated
with poor prognosis and decreased responsiveness to intensive chemotherapy regimens
that are typically used in young and fit patients with acute myeloid leukemia (AML).
Although the combination of a hypomethylating agent (HMA) and venetoclax (HMA/venetoclax)
is being increasingly utilized to treat patients with advanced MPN or MDS/MPN as well
as AML with extramedullary disease (EMD), outcomes in these subgroups of patients
is not well characterized as they were largely excluded from the pivotal VIALE-A trial.
Aims: We sought to evaluate the outcomes associated with HMA/venetoclax in disease
groups that were not well-represented in the pivotal phase III VIALE-A trial.
Methods: We performed an IRB-approved, retrospective chart review of patients who
received HMA and venetoclax (for at least 14 days) between 1/1/2016 and 5/1/2021 at
Johns Hopkins University. Composite complete response included patients who attained
a complete molecular response (CMR), complete cytogenetic response (CCR) or acute
leukemia response-complete (ALR-C) for patients with advanced MPN or MDS/MPN based
on consensus guidelines (Mascarenhas et al., 2012), as well as complete response (CR)
and CR with incomplete hematologic recovery (CRi) for patients with AML and EMD based
on the ELN 2017 criteria (Dohner et al., 2017).
Results: The composite CR rate and median overall survival (OS) were: 42.9% and 9.7
months for the overall population (n=35), 42.9% and 10.2 months for the BP cohort
(n=21), 60% and 9.0 months for the AP cohort (n=5), 33.3% and 8.1 months for the AML
with EMD (n=9). A summary of patient outcomes and their mutational landscape is represented
in Figure 1. Five out of 15 patients who achieved a composite CR went on to receive
an allogeneic bone marrow transplant. Patients with advanced MDS/MPN had numerically
higher composite CR rate (70.0% versus 31.3%; P= 0.1054) and median OS (11.9 months
versus 5.1 months; P= 0.1585) compared to patients with advanced MPN. Patients with
a mutation in the SRSF2 gene had a higher composite CR rate (80% versus 20%; p= 0.0082)
and median OS (10.9 months versus 8.0 months; P= 0.2614) compared to those with a
wild-type SRSF2 gene. On the other hand, there was a trend towards lower composite
CR in AML/blast phase patients who had EMD compared to those without EMD (33% vs 67%;
P=0.1414).
Image:
Summary/Conclusion: The composite CR rate (42.9%) and median OS (9.7 months) for the
overall patient population in our study are lower than those reported in the VIALE-A
trial (42.9% versus 66.4% and 9.7 months versus 14.7 months, respectively). This is
not unexpected since advanced MPN and extramedullary AML are known to have a worse
prognosis than de novo AML. While limited by a small sample size, our study suggests
that HMA/venetoclax may be associated with more favorable outcomes in patients with
advanced MDS/MPN compared to those with advanced MPN. Mutations in the SRSF2 gene
may also be associated with more favorable outcomes. These findings will need to be
confirmed in larger studies and their biological basis will need to be further investigated.
P573: PHARMACOKINETIC EXPOSURE EQUIVALENCE AND PRELIMINARY EFFICACY AND SAFETY FROM
A RANDOMIZED CROSS OVER PHASE 3 STUDY OF AN ORAL HYPOMETHYLATING AGENT DEC-C COMPARED
TO IV DECITABINE IN AML PATIENTS
K. Geissler1, Z. Koristek2, T. Bernal del Castillo3, J. Novák4, G. Rodriguez Macias5,
S. K. Metzelder6, A. Illes7, A. Nagy8, J. Mayer9, M. Arnan10, M.-M. Keating11, J.
Krauter12, M. Lunghi13, N. Stefano Fracchiolla14, U. Platzbecker15, V. Santini16,
Y. Sano17,*, A. Oganesian17, H. Keer17, M. Lübbert18
1Clinic Hietzing, Vienna, Austria; 2University Hospital Ostrava, Ostrava, Czechia;
3Hospital Universitario Central de Asturias, Oviedo, Spain; 4Department of Haematology,
3rd Faculty of Medicine, Charles University and Faculty Hospital Kralovske Vinohrady,
Prague, Czechia; 5Hospital General Universitario Gregorio Marañón, Madrid, Spain;
6Philipps-Universität Marburg, Marburg, Germany; 7Division of Haematologoy, Department
of Internal Medicine, Faculty of Medicine, University of Debrecen, Debreccen; 81st
Department of Internal Medicine, University of Pécs, Pécs, Hungary; 9Fakultní Nemocnice,
Brno, Czechia; 10Hematology Department, Servei d’Hematologia, Institut Català d’Oncologia,
Hospital Duran i Reynals, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL),
Universitat de Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain; 11Queen Elizabeth
II (QEII) Health Sciences Centre, Halifax, Nova Scotia, Canada; 12Städtisches Klinikum
Braunschweig, Braunschweig, Germany; 13Azienda Ospedaliero-Universitaria Maggiore
della Carità Novara, Novara; 14Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico
di Milano, Milano, Italy; 15University Hospital Leipzig, Leipzig, Germany; 16MDS Unit,
AOU Careggi, DMSC, University of Florence, Firenze, Italy; 17Astex Pharmaceuticals,
Inc., Pleasanton, United States of America; 18Universitaetsklinikum Freiburg, Freiburg,
Germany
Background: Parenterally administered hypomethylating agents (HMAs), decitabine (DEC)
and azacitidine (AZA), are approved in Europe for adult patients with acute myeloid
leukemia (AML) who are not candidates for standard induction chemotherapy as single
agent or in combination with venetoclax. ASTX727 (DEC-C) is a fixed dose combination
(FDC) tablet of 35 mg DEC and 100 mg cedazuridine, a novel cytidine deaminase inhibitor
(CDAi). In clinical trials with myelodysplastic syndromes (MDS)/chronic myelomonocytic
leukemia (CMML) patients, DEC-C provides DEC exposures that are equivalent to IV DEC
at the approved dose of 20 mg/m2 daily×5 and is approved as INQOVI® in the US, Canada,
and Australia.
Aims: To demonstrate DEC exposure bioequivalence of oral DEC-C and IV-DEC and generate
clinical data using DEC-C in AML patients.
Methods: The ASCERTAIN study was a randomized cross over design. Patients were randomized
1:1 to either Sequence A: DEC-C (35 mg DEC/100 mg cedazuridine) in Cycle 1 followed
by IV-DEC at 20 mg/m2 in Cycle 2, or Sequence B: receiving IV-DEC in Cycle 1 followed
by DEC-C on Cycle 2 to compare PK [primary endpoint Area Under the Curve (AUC) equivalence
over 5 days of dosing]. All patients received DEC-C from Cycle 3 onwards until treatment
discontinuation to assess safety and clinical efficacy. Patients were eligible as
per the EMA-approved decitabine label (newly diagnosed AML who are not candidates
for standard induction chemotherapy). Clinical responses were assessed according to
modified International Working Group (IWG) 2003 response criteria.
Results: 89 patients were randomized, of whom 87 were treated. The median age of patients
was 78.0 years (range, 61 to 92) with 31 (35.6%) males and 56 (64.4%) females. Cytogenetic
risk classification was poor-risk in 33 (37.9%) and intermediate-risk in 45 (51.7%)
patients. For the primary endpoint, preliminary PK data was available from 69 patients
who successfully completed PK assessments for both IV DEC and DEC-C cycles, and the
DEC AUC0-24 (h*ng/mL) 5-Day geometric mean estimate was 904 for DEC-C and 907 for
IV-DEC resulting in an oral/IV geometric LSM AUC ratio of 99.64% (90% CI of 91.23-108.8%).
Safety findings were consistent with those anticipated for IV-DEC (related Grade ≥
3 AEs in more than 10% were thrombocytopenia, anemia, febrile neutropenia, neutropenia,
and pneumonia). As of the data cutoff date (10 SEP 2021), median follow up was 7.95
months (IQR 6.11-11.86). Of the 77 patients who had ≥6 months of follow up or discontinued
treatment, the best response was complete response (CR) in 17 (22.1%, 95% CI: 13.4,
33.0%). In addition, 4 patients (5.2%) had CR with incomplete blood cell count recovery
(CRi), with 1 patient (1.3%) who had CR with incomplete platelet recovery (CRp), resulting
in composite response rate [CR + CRp] of 23.4% [18/77 patients, 95% CI: 14.5, 34.4%].
These results obtained with DEC-C are consistent with those observed for IV DEC. Based
on preliminary and limited study follow-up with ~46% censored observations, the median
survival was approximately 7.9 months (95% CI: 5.9, 13.0).
Summary/Conclusion: This randomized phase 3 study in AML patients not candidates for
standard induction chemotherapy demonstrates that the oral FDC of DEC-C (35mg/100 mg)
resulted in an equivalent DEC AUC exposure to IV-DEC at 20 mg/m2 over 5 days. In addition,
safety findings and preliminary clinical activity is also consistent with published
data from IV-DEC, suggesting that DEC-C has the potential to be an oral alternative
to the standard IV decitabine Daily×5 regimen.
P574: CPX-351 COMBINED WITH HEMATOPOIETIC CELL TRANSPLANTATION WITH REGULATORY AND
CONVENTIONAL T CELL IMMUNOTHERAPY FOR HIGH-RISK ACUTE MYELOID LEUKEMIA
S. Sciabolacci1,*, L. Ruggeri1, V. Cardinali1, L. Brunetti1, S. Tricarico1, S. Saldi2,
F. Marzuttini1, M. Griselli1, G. Perta1, V. Viglione1, G. Cimino1, A. Osmani1, M.
Caridi1, R. Sembenico1, A. Terenzi1, T. Zei1, R. Iacucci Ostini1, M. F. Martelli1,
B. Falini1, A. Velardi1, C. Aristei2, C. Mecucci1, A. Carotti1, M. P. Martelli1, A.
Pierini1
1Division of Hematology and Clinical Immunology; 2Department of Surgical and Biomedical
Science, Perugia General Hospital, Perugia, Italy
Background: Patients with newly diagnosed acute myeloid leukemia with myelodysplasia-related
changes (MRC-AML) or therapy-related AML (t-AML) have an extremely poor prognosis.
CPX-351, a dual-drug liposomal encapsulation of cytarabine and daunorubicin, improved
significantly rates of remission and overall survival (Lancet JE et al, JCO 2018).
Allogeneic hematopoietic stem cell transplantation (HSCT) remains the only curative
strategy for such patients, but incidence of post-transplant relapse is still unacceptably
high. We developed a novel HSCT strategy that combines an age-adapted myeloablative
irradiation-based conditioning regimen with regulatory and conventional T-cell adoptive
immunotherapy (Treg/Tcon HSCT). HLA-haploidentical Treg/Tcon HSCT ensured low relapse
rate (4%) and resulted in an unprecedented chronic graft versus host disease/relapse
free survival (CRFS, 75%) in AML patients (Pierini et al., Blood Adv. 2021).
Aims: We report preliminary outcomes of the combination therapy with CPX-351 and Treg/Tcon
HSCT for MRC-AML and t-AML at our center.
Methods: We included in the study all patients with MRC-AML or t-AML who were referred
at our center from September 2019 to January 2022 and could be treated with CPX-351.
Treg/Tcon HSCT consisted of a myeloablative conditioning regimen with total marrow/lymphoid
irradiation, thiotepa, fludarabine and cyclophosphamide followed by an infusion of
2x106/Kg donor Tregs on day -4, 1x106/Kg Tcons on day -1 and a megadose (>6x106/Kg)
of purified CD34+ hematopoietic progenitor cells on day 0. No pharmacological GvHD
prophylaxis was given post-transplant (clinicaltrials.gov: #NCT03977103).
Results: Seventeen patients with a median age of 57.5 years (46-72) were treated with
CPX-351: 13 with newly diagnosed AML (11 MRC-AML and 2 t-AML) and 4 patients who relapsed
after a previous HSCT and who acquired novel cytogenetic abnormalities (2 complex
karyotypes, CK). 9/13 patients with newly diagnosed AML had karyotype abnormalities:
4 monosomy 7, 1 CK, 1 t(3;3), 1 isolated del(20q) and 2 (both t-AML) t(11;20) which
resulted in NUP98-TOP1 fusion. Nine patients achieved complete remission (CR), 2 partial
response (PR) and 2 had persistent disease. No patient experienced grade>3 extrahematologic
toxicity.
Ten/13 patients underwent a first HSCT (8 in CR, 2 in PR). Eight underwent Treg/Tcon
HSCT (3 from HLA-matched, 5 from HLA-haploidentical donor), while 2 unmanipulated
haploidentical HSCT followed by post-transplant cyclophosphamide (PT-Cy). All patients
achieved full-donor engraftment. No patients experienced non-relapse mortality (NRM).
Three/10 patients had a grade ≥ III acute GvHD; all of them are alive and off immunosuppressive
therapy. At a median follow up of 16.2 months (range 9-29 months), 9/10 patients are
alive and in CR with no chronic GvHD. No relapse occurred in the 8 patients who underwent
Treg/Tcon HSCT, while one patient who underwent HSCT followed by PT-CY relapsed.
The 4 patients who were treated after one previous HSCT received one single induction
cycle with CPX-351, achieved CR and experienced no grade>3 extra-hematologic toxicity.
All of them could undergo a second HSCT. NRM occurred in 1 patient (septic shock),
while 3 patients are alive to date.
Summary/Conclusion: CPX-351 allowed most of MRC-AML and t-AML patients to reach HSCT
in CR despite high risk genetics and even when used in patients who relapsed after
a 1st HSCT. Combination of CPX-351 with Treg-Tcon HSCT is safe and exert powerful
antileukemic activity. Thus, it appears to be a promising strategy to ensure high
rate of CRFS in very high-risk AML.
P575: IMPACT OF MOLECULAR RESPONSE AND CHEMOTHERAPY REGIMEN ON OUTCOMES IN CORE BINDING
FACTOR AML
J. Senapati1,*, F. Haddad1, F. Ravandi1, T. Kadia1, C. DiNardo1, N. Daver1, N. Pemmaraju1,
Y. Alvarado1, M. A. Brandt1, H. Kantarjian1, G. Borthakur1
1Leukemia, MD Anderson Cancer Center, Houston, United States of America
Background: Fludarabine based induction/consolidation regimens result in excellent
outcomes in core binding factor acute myeloid leukemia (CBF-AML) Attainment of early
polymerase chain reaction (PCR) based optimal minimal residual disease (MRD) responses
positively impact long term outcomes and provide early decision timepoints for alternative
therapies including stem cell transplantation (SCT).
Aims: We studied factors that affected early and late optimal PCR responses (OPR)
and survival in patients (pts) of CBF AML who were treated with fludarabine based
regiemns (fludarabine, cytarabine and GCSF) with idarubicin (FLAG-IDA) or gemtuzumab
(FLAG-GO).
Methods: This is a prospective study of pts with N/D CBF-AML treated at our center
on FLAG based regimens. Pts received up to 7 cycles of therapy with appropriate dose
reductions based on age or toxicity. PCR for the disease defining transcripts (CBFB::MYH11
or RUNX1::RUNX1T1) were monitored after C1, C3 and end of therapy (EOT). Based on
our earlier report (Boddu et. al., Leukemia 2018) OPR was defined as PCR transcript
<0.1% after C1 and <0.01% for later time points.
Results: Between 4/2007 to 12/2019, 174 pts, with a median age of 51 years (range,
19-78) were included. A total of 109 pts (63%) received FLAG-IDA, while the rest got
FLAG-GO therapy. This was mostly due to withdrawal of approval of GO in the US for
a period of time. Median number of cycles received was 6 (range, 1-7) Baseline parameters
are described in Table 1.
PCR data is available for 94% pts after C1, 90% after C3 and 92% at EOT. EOT was considered
the timepoint at which pt stopped further FLAG based therapy beyond C1. The percent
of patients achieving OPR at each time point by regimen are: 68% vs. 41% (p=0.002)
after C1, 75% vs. 39% (P< 0.0001) after C3 and 92% vs. 55% (p<0.0001) at EOT for FLAG-GO
and FLAG-IDA respectively. On univariate analysis, FLAG-GO regimen favored attainment
of post C1 OPR over FLAG-IDA (OR, odds ratio=3.1, 95%CI 1.6-6.2) while age, baseline
BM blast, cytogenetic subtype of CBF, ACA and kinase mutations did not. The benefit
of FLAG-GO in attaining OPR after C1 was maintained even on a multivariate (MV) model.
For EOT OPR, FLAG-GO (OR= 8.5, 95% CI 3.4-26) fared better over FLAG-IDA. Absence
of KIT mutation (OR=2.4) and attainment of post C1 OPR (OR= 11.3) were the other relevant
factors. On MV analysis, only chemo arm and post C1 OPR remained significant.
At a median follow up of 92 months (mos), the RFS was 124 mos and OS not reached (NR)
for the whole cohort (RFS 68 mos and OS 102 mos for FLAG-IDA treated pts vs. both
NR for FLAG-GO treated pts, p=0.004 and 0.04 respectively). Across the 2 groups there
was no difference in RFS or OS between pts who attained early or late OPR. However,
on stratifying serial PCR responses with therapy arms, even in pts with early OPR
(after C1), RFS was better (NR) in FLAG-GO arm vs 101 mos for FLAG-IDA arm (p= 0.03),
though OS was similar, likely reflecting efficacy of salvage therapies in this subset
of AML. In a Cox proportional hazards model that included age, therapy arm, kinase
mutations, cumulative therapy cycles, post C1 and EOT PCR responses; only younger
age and EOT OPR favorably affected RFS; however FLAG-GO treated pts had lower hazards
for RFS at any level of PCR response compared to FLAG-IDA treated pts (Fig. 1).
Image:
Summary/Conclusion: In CBF AML, FLAG-GO regimen leads to higher rate of earlier and
end of therapy OPR than FLAG-Ida and substantially better RFS. At any combination
of PCR response, FLAG-GO treated patients had lower hazards for relapse than FLAG-IDA
treated pts.
P576: A PHASE I/II STUDY OF MILADEMETAN (DS3032B) IN COMBINATION WITH LOW DOSE CYTARABINE
WITH OR WITHOUT VENETOCLAX IN ACUTE MYELOID LEUKEMIA
J. Senapati1,*, J. Ishizawa1, H. A. Abbas1, S. Loghavi1, G. Borthakur1, G. C. Issa1,
S. I. Dara1, R. Pourebrahim1, N. Daver1, E. J. Jabbour1, S. M. Kornblau1, N. Pemmaraju1,
G. Garcia-Manero1, F. Ravandi1, M. Muftuoglu1, J. Khoury1, C. DiNardo1, M. Andreeff1
1Leukemia, MD Anderson Cancer Center, Houston, United States of America
Background: In TP53 wild type (WT) acute myeloid leukemia (AML), inhibition of MDM2
increases p53 protein expression and mediates antileukemic effects. MDM2i monotherapy
in AML has shown only modest responses; combining MDM2i with anti-leukemic therapies
with additive or synergistic pre-clinical activity and non-overlapping toxicities
might potentiate therapeutic benefit.
Aims: To evaluate safety and efficacy of milademetan (DS-3032b), an MDM2i, with low
dose cytarabine (LDAC) ± venetoclax (VEN) in patients (pts) with AML.
Methods: This open label, single arm Phase I/II trial (NCT03634228) included pts with
AML and WT TP53. The phase I utilized a 3 + 3 design to identify safety and tolerability
of the combination and determine the recommended combination dose to proceed to the
expansion phase (phase II). Pts included were adult (≥18 years), ECOG PS ≤ 3, with
adequate renal and hepatic function. Key exclusion criteria included acute promyelocytic
leukemia, prior therapy with MDM2i, or stem cell transplantation (SCT) in the preceding
60 days.
Doses of milademetan and combination therapy are provided in Table 1. Toxicity was
graded according to the NCI-CTCAE v.5 and responses according to the ELN 2017 AML
response criteria.
Results: A total of 21 pts were screened; 16 pts were enrolled and treated in phase
I. Median age was 70 years (range, 23-80) and 11 (69%) were women. Median number of
prior therapies was 3 (range, 1-7), with 6 pts (37%) who had undergone prior SCT.
Two pts (12.5%) were newly diagnosed with treated secondary AML; having received 3
and 2 lines of therapy for prior MDS. Milademetan combination therapy was reasonably
well tolerated at all dose levels, the most common adverse events being gastrointestinal.
One pt had an attributable grade 3-4 toxicity (diarrhea), which was considered a dose
limiting toxicity. Pts received a median of 1 cycle (range, 1-4). Two pts (12.5%)
achieved an overall response (CRp, 12.5%, 1 each at dose level 1 and 2). At a median
follow up of 10.5 mos (range, 0.6-17), 5 pts are alive (31%), all on subsequent lines
of therapy. Ten of 11 deaths occurred in non-responding pts and were attributed to
disease progression or infection. One responding pt died of infection in cycle 3.
Given the minimal response rates at all dose levels, the phase II expansion portion
of the trial was not conducted.
p53 immunohistochemistry was performed on baseline bone marrow (BM) samples from 11
pts, all of whom had WT expression pattern. BM samples of 1 amongst 7 pts evaluated
post C1 demonstrated acquisition of mutant p53. We performed CyTOF analysis of sequentially
collected samples to interrogate signalling pathways, the p53-MDM2 axis, and the abundance
of pro/anti-apoptotic molecules. We observed that CD34+ leukemia blasts expressed
high levels of Bcl-2 and were eliminated after therapy in a patient with CRp while
Bcl-2 low non-malignant monocytic cells preferentially enriched. Comparative single-cell
proteomic analysis in responders and non-responders are underway to identify proteomic
correlates of favourable response vs. therapy resistance.
Image:
Summary/Conclusion: The combination of milademetan with LDAC ± venetoclax was reasonably
well tolerated but showed only minimal clinical responses at all 3 dose levels in
a heavily pre-treated AML population. Further studies are needed to decipher the most
appropriate combination regimen and pt population to benefit from milademetan.
P577: 20% BLAST CRITERIA FOR DIAGNOSING AML/MDS: IS IT TIME TO MOVE BEYOND?
V. Sheth1,*, R. Walter2,3, C. Shaw3, E. Estey2,3, M.-B. Percival2,3
1Deprtment of hematology and Bone marrow transplant, UAB, Birmingham; 2Division of
hematology, University Washington; 3Clinical research division, Fred Hutch, SEATTLE,
United States of America
Background: Classification of acute myeloid leukemia (AML) and myelodysplastic syndromes
(MDS) is driven by an arbitrary blast percentage cut off (20%, using the 2016 WHO
criteria), which subsequently affects treatment options (including enrollment in clinical
trials) without taking genomic and biological characteristics into consideration.
It has recently been posited that AML and MDS with excess blasts (EB) exist on a continuum
based on biological features rather than blast percentages, and their outcomes post
treatment are very similar.
Aims: We therefore hypothesized that the IPSS-R (MDS prognostication using hematologic
parameters and cytogenetic features) should predict outcomes for WHO-defined AML,
and similarly, ELN2017 (AML prognostication using molecular and cytogenetic features)
should predict outcomes for WHO-defined MDS-EB (5-19% blasts).
Methods: We retrospectively analyzed 1034 adult patients from the UW/FHCRC registry
database, who received treatment for AML or MDS-EB between the years 2005-2018. WHO-defined
AML occurred in 777 patients (67%) and WHO-defined MDS-EB in 257 (23%). Median follow
up was 5 years. Outcomes were relapse-free survival (RFS) and overall survival (OS).
Results: Median age of patients in AML group was 63 years (range 18-91), and in MDS
group was 60 years (range 22-85). The majority of the patients in AML cohort had intermediate
(37%) or high-risk disease (37%) using ELN2017 classification; many were reclassified
to very high risk by IPSS-R (54%). Most of the patients in MDS cohort were very high
risk (54%) using IPSS-R; many were reclassified to ELN high risk (46%). For AML patients,
classification using the IPSS-R was highly predictive of OS for AML in univariate
(p=0.001) and multivariate analysis (reference low risk/intermediate risk) (p=0.001,
HR-2.1, 95% confidence interval (CI) 1.1-2.9 for very high risk and p=0.2, HR-1.4,
95%CI 0.76- 1.9 for high risk in AML), as well as RFS in univariate (p=0.001) and
multivariate analysis (reference low risk/intermediate risk) (p=0.001, HR-2.08, 95%CI
1.2-2.8 for very high risk and 0.2, HR-1.4, 95%, CI 0.2-1.8 for high risk in AML).
Similarly, ELN 2017 was predictive of OS in MDS cohort in univariate (p=0.025), and
multivariate analysis (reference ELN adverse (p=0.005, HR-0.1, 95%CI 0.02-0.5, ELN
favorable risk), as well as RFS in univariate (p=0.043) and multivariate analysis
(reference ELN adverse) (p=0.019, HR-0.25 for ELN favorable risk, 95%CI 0.04- 0.8).
In the subgroup of AML patients who achieved CR/CRi, IPSS-R was predictive of RFS
in univariate analysis (p=<0.001), and multivariate analysis (ref low/int) (p=0.001,
HR=2.08 for very high risk). Similarly, in the subgroup of MDS patients who achieved
CR/CRi, ELN 2017 was predictive of RFS (p=<0.001), but this effect was not sustained
in multivariable analysis.
Summary/Conclusion: Data from our cohort suggest that IPSS-R can be predictive of
survival outcomes in WHO-defined AML patients and ELN 2017 can similarly be predictive
in WHO-defined MDS patients. Therefore, we suggest that diagnostic criteria for AML
and MDS should include more biological and genomic features rather than a blast percentage
cut-off, using tools such as the recently presented IPSS-molecular for MDS. Clinical
trials of new therapies should include all patients having 35% blasts (MDS-EB and
AML). This study, however, carries a potential drawback of retrospective classification
and lack of complete molecular data through comprehensive targeted next generation
sequencing panel in patients treated at earlier time periods
P578: GILTERITINIB FOR RELAPSED/REFRACTORY FLT3 MUTATED ACUTE MYELOID LEUKEMIA A REAL-WORLD,
MULTI-CENTER, MATCHED ANALYSIS
S. Shimony1,2,3, J. Canaani3,4, E. Kugler2,3,*, B. Nachmias5, R. Ram6,7, A. Frisch8,
C. Ganzel9, V. Vainstein5, Y. Moshe3,10, S. Aumann5, M. Yeshurun2,3, Y. Ofran9, P.
Raanani2,3, O. Wolach2,3
1Leukemia, Dana Farber Cancer Institute, Boston, United States of America; 2hematology,
Rabin medical center, Petah Tikva; 3Sackler Medical School, Tel Aviv University, Tel
Aviv; 4Leukemia, Chaim Sheba Medical Center, Ramat Gan; 5hematology, Hadassah Medical
Center and Hebrew university Faculty, Jerusalem; 6BMT Unit; 7Sackler Medical School,
Tel Aviv Sourasky Medical Center, Tel Aviv; 8Department of Hematology and Bone Marrow
Transplantation, Rambam Health Care Campus, and Rappaport Faculty of Medicine - Technion,
Haifa; 9hematology, Shaare Zedek Medical Center, and Faculty of Medicine, Hebrew University
of Jerusalem, Jerusalem; 10hematology, Tel Aviv Sourasky Medical Center, Tel Aviv,
Israel
Background: Patients with FLT3-mutated relapsed or refractory (R/R) acute myeloid
leukemia (AML) have a dismal prognosis. Gilteritinib is a FLT3 tyrosine kinase inhibitor
(TKI) recently approved for patients with R/R AML.
Aims: We aimed to characterize real-world data regarding gilteritinib treatment in
FLT3-mutated R/R AML and to compare outcomes with matched FLT3-mutated R/R AML patients
treated with chemotherapy-based salvage regimens.
Methods: Survival analysis was performed by Kaplan Meyer with log rank test to compare
between subgroups. Cox proportional hazards regression models were fitted to predict
effect of covariates on overall survival (OS) and event free survival (EFS). All statistical
analyses used 2-sided p value with a threshold of 0.05.
Results: Twenty-five patients from six academic centers were treated with gilteritinib
for FLT3-mutated R/R AML. The median age was 58 (IQR1-3 47-73) years, 80% (n=20) were
treated with a prior intensive induction regimen and 40% of them received prior TKI
therapy. After a median time of seven months post gilteritinib initiation (range 1-34),
12 patients (48%) achieved complete response (CR) with gilteritinib (figure 1A). The
estimated median overall survival (OS) of the entire cohort was eight (CI 95% 0-16.2)
months and was significantly higher in patients who achieved CR compared to those
who did not (16.3 months, CI 95% 0-36.2 vs. 2.6 months, CI 95% 1.47-3.7; p value=0.046,
figure 1B). In a multivariate cox regression analysis, achievement of CR was the only
predictor for longer OS (HR 0.33 95% CI 0.11-0.97, p=0.044). Prior TKI exposure did
not affect OS but was associated with better event-free survival (HR 0.15 95% CI 0.03-0.71,
p=0.016). An age and ELN-risk matched comparison between patients treated with gilteritinib
and intensive salvage revealed similar response rates (50% in both groups); median
OS was 9.6 months (CI 95% 2.3-16,8) vs. 7 months (CI 95% 5.1-8.9) in gilteritinib
and matched controls, respectively (p = 0.869, figure 1C).
Image:
Summary/Conclusion: In the real-world setting gilteritinib is effective, including
in heavily pre-treated, TKI exposed patients.
P579: A PHASE 4 STUDY OF FRACTIONATED GEMTUZUMAB OZOGAMICIN ON QT INTERVAL AND SAFETY
IN PATIENTS WITH RELAPSED/REFRACTORY CD33-POSITIVE ACUTE MYELOID LEUKEMIA
P. Montesinos1,*, V. Kota2, J. Brandwein3, P. Bousset4, R. J. Benner5, E. Vandendries6,
M. F. McMullin7
1Department of Hematology, Hospital Universitario i Politècnico la Fe, Valencia, Spain;
2Department of Medicine: Hematology and Oncology, Medical College of Georgia, Augusta
University, Augusta, United States of America; 3Department of Medicine, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, Canada; 4Pfizer Inc., Paris,
France; 5Pfizer Inc., Groton; 6Pfizer Inc., Cambridge, United States of America; 7School
of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast,
United Kingdom
Background: Gemtuzumab ozogamicin (GO) is a CD33-directed antibody-drug conjugate
indicated for treatment of relapsed/refractory (R/R) acute myeloid leukemia (AML).
Fractionated dosing of GO has demonstrated an enhanced safety profile without hampering
efficacy.
Aims: To assess the effect of fractionated GO as monotherapy on corrected QT (QTc)
interval, safety, pharmacokinetics (PK), and immunogenicity of GO in patients (pts)
with R/R AML.
Methods: Pts aged ≥12 y with R/R AML were enrolled in a single-arm, open-label, phase
4 study (NCT03727750) and received a fractionated dose of GO 3 mg/m2 on days 1, 4,
and 7 of each cycle, up to 2 cycles. Pts receiving ≥1 dose of GO were evaluable for
QTc, safety, and PK. The primary endpoint was mean change from baseline in QTc. Secondary
endpoints included PK parameters, adverse events (AEs), including veno-occlusive disease/sinusoidal
obstruction syndrome (VOD/SOS), incidence of anti-drug antibodies (ADAs)/neutralizing
antibodies (NAb), overall survival (OS), and response rate of complete remission (CR)/CR
with incomplete hematologic recovery (CRi). This analysis includes data from pts aged
≥18 y.
Results: Of 51 adult pts enrolled (median age 67 y), 50 (98%) pts received ≥1 dose
of GO during cycle 1; 9 (18%) pts were treated with GO during cycle 2. The upper limit
of the 2-sided 90% CI for least squares mean differences in QTc according to Fridericia’s
formula (QTcF) was <10 msec for all timepoints during cycle 1 (Table). Most pts had
a QTcF ≤450 msec (90%) and a maximum increase from baseline QTcF ≤30 msec (94%). No
pts had a post-baseline QTcF >480 msec or a change from baseline >60 msec. Treatment-emergent
AEs (TEAEs) occurred in 98% of pts; 54% were grade 3/4. The most common (>10%) grade
3/4 TEAEs were febrile neutropenia (36%) and thrombocytopenia (18%). One pt experienced
grade 3 atrial fibrillation and supraventricular tachycardia, unrelated to GO. No
pts experienced grade 4 or higher cardiac conduction TEAEs. VOD/SOS was not reported;
however, one pt experienced treatment-related grade 5 capillary leak syndrome associated
with pleural effusion, ascites, hyperbilirubinemia, and endothelial syndrome. The
overall incidence of ADAs and NAbs were 12% and 2%, respectively. ADAs were persistent
in 5/6 pts (83%). Of the 6 pts with treatment-induced ADAs, 2 experienced pyrexia
(grade 1 and grade 3) within 24 h of the infusion. Among ADA-negative pts (n=44),
infusion-related reactions were observed in 7 (16%) pts. These were grade 1/2, except
for 1 instance of grade 3 urticaria. Median (95% CI) OS was 2.8 (1.7–4.2) mo. Disease
progression was the most common cause of death in 35/45 (78%) pts. A CR/CRi was observed
in 5/51 (10%) pts.
Image:
Summary/Conclusion: Fractionated dosing of GO (3 mg/m2 per cycle) is not predicted
to pose a clinically significant safety risk for QT prolongation in pts with R/R AML,
with the upper limit of the 2-sided 90% CI for the least squares mean change from
baseline falling below the 20 msec threshold of clinical concern for oncology drugs.
TEAEs reported in this study are consistent with the known safety profile of GO, and
the presence of ADAs does not appear to be associated with any potential safety concerns.
Response rates were lower than previously reported in the MyloFrance 1 study, which
included less heavily pretreated patients (i.e., patients only in first relapse; Taksin
et al, Leukemia, 2007).
P580: THE USE OF LEUKAEMIA Q-FUSION GENE SCREENING ASSAY (Q30) IN THE DIAGNOSTIC EVALUATION
OF ACUTE MYELOID LEUKAEMIA (AML)
S. Tayabali1,*, R. Baker1,2, E. Nacheva1,2, J. O’Nions1,2, R. Gupta1,2, A. J. Wilson1,2,
K. Xu1,2
1University College London Hospital; 2UCLH Specialist Integrated Haematology Malignancy
Diagnostic Service, Health Services Laboratories, London, United Kingdom
Background: AML is a clinically heterogenous disease characterised by chromosomal
and genetic abnormalities which guide risk stratification and management. Hence timely
cytogenetic and molecular profiling is essential to initiate appropriate treatment.
Approximately 30% of AML cases have fusion genes. We use a combination of fluorescence
in situ hybridization (FISH) to rapidly screen for common chromosomal abnormalities
and fusion genes, and chromosomal microarray (CMA) to assess copy number variations
across the whole genome. However, cytogenetic analysis is resource intensive and may
take several days which can delay patient management.
The QuanDx© Leukaemia Q-Fusion Genes Screening Kit (Q30) uses multiplex reverse transcription
real-time PCR (RT-qPCR) to allow simultaneous detection of 30 fusion genes with 140+
breakpoints within hours. The use of Q30 may allow earlier risk stratification and
treatment before FISH and CMA results are available.
Aims:
To evaluate the Q30 Kit to detect rearrangements on bone marrow and peripheral blood
specimens from newly diagnosed AML patients; and compare concordance of Q30 and FISH
in identifying leukaemia fusion genes.
Methods:
We retrospectively reviewed FISH, CMA and Q30 data for new AML cases diagnosed in
the specialist integrated haematological malignancy diagnostic service (SIHMDS) at
University College London Hospital over a 2 year period: 01 Jan 19 to 31 Dec 20.
Results:
114 cases were included. The median age was 59 [range 19 to 91]. Male 59 (52%), Female
55 (48 %).
ELN Cytogenetic Risk Group (n;%)
Favourable 19 (17%)
Intermediate 69 (61%)
Adverse 26 (23%)
Type of AML (n;%)
De novo 105 (92%)
Secondary 7 (6%)
Therapy Related 2 (2%)
30 (26%) of 114 patients had a detectable fusion gene by Q30 (Figure 1). 25/30 were
confirmed by FISH either on our standard panel or subsequently using targeted probes.
The 5/30 fusion genes detected by Q30, but not by FISH were:
- t(3;5)(q25;q34) NPM1-MLF1 (n=1)
- t(9;22)(q34;q11) BCR-ABL1 with a high cycle threshold (Ct) value (n=3)
- t(16;21)(p11;q22) TLS-ERG (n=1)
The discordant results were due to high Ct values (detecting low level gene fusions
below the limit of FISH sensitivity), or a lack of commercially available fusion probes.
Image:
Summary/Conclusion: The ability to rapidly risk-stratify newly diagnosed AML patients
allows earlier initiation of targeted therapies. We found that Q30 is highly sensitive
and showed 100% concordance in identifying fusions associated with good cytogenetic
risk AML in our cohort. Q30 can also identify high risk fusion genes including DEK-CAN,
MLL-AF6 and other high risk MLL translocations included in the panel which are rare
and for which FISH fusion probes are not routinely used. These may have been otherwise
undetected, but can subsequently be confirmed by FISH, and importantly allow the generation
of molecular measurable residual disease (MRD) assays to assess treatment response.
We conclude that Q30 is a rapid, simple and sensitive adjunct to conventional FISH
in the diagnosis of AML and, in combination with CMA, may preclude the requirement
for conventional and time consuming G-banding analysis.
P581: PHASE II STUDY OF LOWER-INTENSITY FRONTLINE THERAPY FOR NEWLY DIAGNOSED PATIENTS
WITH AML WHO ARE UNFIT OR OTHERWISE NOT ELIGIBLE FOR FRONTLINE CLINICAL TRIALS
S. Venugopal1,*, E. Jabbour1, N. Pemmaraju1, G. Montalban-Bravo1, K. S. Chien1, N.
Daver1, N. Jain1, J. Burger1, Y. Alvarado1, A. Maiti1, C. D. DiNardo1, G. Borthakur1,
R. Malla1, G. Garcia-Manero1, F. Ravandi1, H. Kantarjian1, T. Kadia1
1Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, United States
of America
Background: Pts with newly diagnosed (ND) AML frequently present with abnormal organ
function, poor performance status (PS), concurrent active malignancies, and active
infections. These factors often preclude these pts from enrollment on frontline clinical
trials, since standard eligibility routinely exclude them.
Aims: Therefore, we designed a lower-intensity regimen of cladribine plus low-dose
cytarabine (LDAC) alternating with decitabine (DAC) with less stringent inclusion
criteria in ND pts unfit or otherwise ineligible for existing clinical trials.
Methods: Pts >18 years with untreated AML and ineligible for other frontline AML clinical
trials were enrolled (NCT01515527). Eligibility criteria included either creatinine
≥2mg/dL; or total bilirubin ≥2 mg/dL; or ECOG PS of 3 or 4; or ineligible for participation
in a higher priority protocol. Pts with active concurrent malignancies and ongoing
infection related to their AML could be enrolled. Induction was cladribine 5 mg/m2
IV on D1-5, Cytarabine 20 mg SQ twice daily on D 1-10, followed by consolidation with
cladribine 5 mg/m2 IV on D1-3, Cytarabine 20 mg SQ twice daily on D 1-10 alternating
with decitabine 20 mg/m2 IV, daily on D 1-5. The primary objective was 60-d survival
rate.
Results: A total of 25 pts have been enrolled. The median age was 73 years (range,
52-82) with 76% of the cohort older than 70 years. Six pts (24%) had concurrent active
malignancy, 4 (16%) had baseline creatinine >2mg/dL, and 9 (36%) had PS ≥3. 76% of
them were adverse risk per ELN 2017. Baseline characteristics are shown in figure.
Among 25 evaluable pts, 17 (68%) achieved a composite complete response (CRc) including
10 (40%) CR and 7 (28%) CR with incomplete count recovery (CRi). Among responders,
6 pts (35%) achieved MRD neg by flow. Of 7 pts with no response, all were ELN adverse
risk. Median cycles to response was 1 (range: 1 - 4). 30- and 60 d mortality was 8%
and 16%, respectively, including 2 pts (8%) who died due to pseudomonal sepsis on
D8, and the other due to pneumonia on D11.
At a median follow up of 9.4 months (range, 0.4- 19.9 m),median OS (6-mo OS% - 61
%), and EFS was 8.3 mo each (6-mo RFS% - 56 %) with a 60-d OS and EFS rate of 83%
each, with a median RFS of 5.8 mo (2-mo RFS%-66%; 6-mo RFS% - 49 %). In this challenging
patient population, this lower-intensity regimen was well tolerated, with an acceptable
toxicity profile.
Image:
Summary/Conclusion: In an unfit patient population with a high comorbidity burden,
that were ineligible for other clinical trials, induction therapy with Cladribine
plus LDAC was feasible and effective in newly diagnosed pts with AML. The regimen
produced high rates of response, encouraging EFS, OS, and low early mortality in a
cohort predicted to have a high risk of early death. Treating this pt population on
a clinical trial is feasible and can allow pts to achieve remission and move on to
effective post-remission therapy.
P582: FIRST RESULTS OF A PHASE II STUDY (STIMULUS-AML1) INVESTIGATING SABATOLIMAB
+ AZACITIDINE + VENETOCLAX IN PATIENTS WITH NEWLY DIAGNOSED ACUTE MYELOID LEUKEMIA
A. M. Zeidan1,*, J. Westermann2, T. Kovacsovics3, S. Assouline4, A. C. Schuh5, H.-J.
Kim6, G. Rodriguez Macias7, D. Sanford8, M. R. Luskin9, E. M. Stein10, K. Malek11,
J. Lyu12, M. Stegert11, J. Esteve13
1Yale University and Yale Cancer Center, New Haven, United States of America; 2Charité-University
Medical Center Berlin, Campus Virchow Clinic, Berlin, Germany; 3University of Utah
and Huntsman Cancer Institute, Salt Lake City, United States of America; 4Jewish General
Hospital, McGill University, Montreal; 5Princess Margaret Cancer Centre, University
of Toronto, Toronto, Canada; 6Catholic Hematology Hospital, Seoul St. Mary’s Hospital,
College of Medicine, The Catholic University of Korea, Seoul, South Korea; 7Hospital
General Universitario Gregorio Marañón, Madrid, Spain; 8BC Cancer, University of British
Columbia, Vancouver, Canada; 9Dana-Farber Cancer Institute, Harvard Medical School,
Boston; 10Memorial Sloan Kettering Cancer Center, New York, United States of America;
11Novartis Pharma AG, Basel, Switzerland; 12Novartis Institutes for BioMedical Research,
Shanghai, China; 13Hospital Clínic, Barcelona, Spain
Background: The BCL-2 inhibitor venetoclax (VEN) in combination with a hypomethylating
agent (HMA) has improved outcomes for patients with newly diagnosed (ND) acute myeloid
leukemia (AML) who are unfit for intensive chemotherapy (IC; DiNardo CD, N Engl J
Med, 2020); however, responses are often transient. Sabatolimab is a novel immuno-myeloid
therapy targeting TIM-3, an immune regulator expressed on immune cells and myeloid
leukemic progenitors but not on normal hematopoietic stem cells. Sabatolimab + HMA
has shown promising durable responses in a Phase Ib study in patients with ND-AML
and myelodysplastic syndrome (Wei AH, ASH 2021; NCT03066648). Treatment with sabatolimab
+ VEN + azacitidine (AZA) may also induce different pathways of cancer cell elimination.
Aims: In this first presentation of the study, we report findings from the dose-escalation
part of STIMULUS-AML1 (NCT04150029), a Phase II, single-arm study of sabatolimab +
AZA + VEN in adult patients with AML.
Methods: Adult patients with ND-AML ineligible for IC were enrolled. In dose-escalation
part 1 (safety run-in), patients received either 400 mg (Cohort 1) or 800 mg (Cohort
2) sabatolimab every 4 weeks (Q4W) plus 400 mg VEN once a day every day, plus 75 mg/m2
AZA on Days (D) 1-7, or D1-5+D8-9, or D1-6+D8 of a 28-day cycle (Zeidan A, ESH-AML
2022). For the safety run-in part, the primary endpoint was assessment of the incidence
of dose-limiting toxicities (DLTs) between Cycle 1 D8 and the end of Cycle 2.
Results: As of the Sep 6, 2021 data cutoff, 18 patients have been treated in part
1 of the study with sabatolimab + AZA + VEN (Cohort 1, n=5; Cohort 2, n=13). Baseline
demographics are shown in Table 1. Treatment was ongoing for 9 (50%) patients (Cohort
1, n=1; Cohort 2, n=8). Reasons for study treatment discontinuation included adverse
events (AEs; n=3), progressive disease (n=3, including a case of chloroma), planned
stem cell transplant (n=2), and physician decision (n=1). Only 1 DLT (elevated troponin
T/asymptomatic myocarditis) was reported in Cohort 2 and no DLTs were reported in
Cohort 1. Table 2 summarizes AEs observed in >20% of patients in the study. Consistent
with AEs often observed during AZA + VEN therapy (DiNardo CD, N Engl J Med, 2020),
the 5 most common AEs regardless of relationship to study treatment were constipation
(all grades [gr]: 39%; gr ≥3: 0%) and hematologic events, including anemia (33%; 28%),
decreased platelets (33%; 33%), neutropenia (39%; 39%), and febrile neutropenia (gr:
50%). Treatment-related AEs gr ≥3 in >25% of all patients included neutropenia (39%),
decreased platelet count (33%), and decreased neutrophil count (28%). Treatment-related
AEs led to study treatment discontinuation in 3 patients (platelet count decreased
[n=2], troponin level increased/asymptomatic myocarditis [n=1]). AEs led to sabatolimab
dose interruption in 6 patients, all in Cohort 2. No sabatolimab dose reduction was
observed. Fourteen patients had dose interruption of VEN due to AEs. Five patients
had dose reduction of VEN (none due to AEs). Serious AEs were reported in 14 (78%)
patients; the most common was febrile neutropenia (44%). No other serious AEs were
reported in >1 patient. Efficacy data will be presented at the EHA meeting.
Image:
Summary/Conclusion: Safety and tolerability of sabatolimab + AZA + VEN were comparable
at 2 dose levels (400 and 800 mg) of sabatolimab and were overall comparable to the
reported safety profile of VEN + AZA doublet therapy. These findings supported initiation
of the expansion cohort of STIMULUS-AML1 at a sabatolimab 800 mg dose.
P583: PATIENTS AT HIGH RISK OF RELAPSE POST-TRANSPLANT: A PHASE 1 STUDY DESIGN WITH
A NOVEL TREATMENT STRATEGY USING THE ESTIMAND FRAMEWORK
R. Zeiser1,*, C. Schmid2, G. Al-Atrash3, Y. Xu4, H.-J. Weber5, L. Eldjerou6, S. Weber7,
L. Widmer7, C. Craddock8
1Department of Medicine I, Medical Center, University Freiburg, Freiburg; 2Department
of Hematology and Oncology, Augsburg University Hospital and Medical Faculty, Augsburg,
Germany; 3Department of Stem Cell Transplantation and Cellular Therapy, The University
of Texas MD Anderson Cancer Center, Houston; 4Oncology Analytics, Novartis Pharmaceutical
Corporation, East Hanover, United States of America; 5Oncology Analytics, Novartis
Pharma AG, Basel, Switzerland; 6Clinical Development, Novartis Pharmaceuticals Corporation,
East Hanover, United States of America; 7Advanced Exploratory Analytics, Novartis
Pharma AG, Basel, Switzerland; 8Centre for Clinical Haematology, University Hospitals
Birmingham NHS Foundation Trust, Birmingham, United Kingdom
Background: The risk of disease relapse in patients (pts) allografted for acute myeloid
leukemia (AML) is high and remains the main cause of transplant failure. The great
majority of pts destined to relapse post-allogeneic stem cell transplant (SCT) will
do so within the first 12 months and survival after early relapse is dismal. Therefore,
strategies to prevent relapse are urgently required. A potent graft-versus-leukemia
(GvL) effect is exerted after SCT, and strategies that may augment a GVL effect have
the potential to reduce relapse risk post-SCT. Siremadlin (HDM201) is a novel investigational
MDM2 inhibitor with single-agent (SA) anti-AML activity. MDM2 inhibitors, including
siremadlin, possess potent immunomodulatory effects (IME) in murine solid tumor and
AML models. The post-allogeneic SCT setting is ideal for investigating the IME of
MDM2 inhibition on enhancing GvL effect. Here we present a phase Ib/II proof-of-concept
study designed to assess safety and preliminary efficacy of a treatment strategy with
siremadlin as monotherapy and, subsequently, in combination with donor lymphocyte
infusions (DLIs) for AML in post-SCT setting using the estimand framework (ICH E9
(R1) addendum).
Aims: The objective of the planned treatment strategy is to enhance GvL post-SCT by
siremadlin as monotherapy and in combination with DLIs. Due to the risk of Graft-vs-Host
Disease (GvHD) with DLI early after SCT, siremadlin will be given as priming monotherapy
to prevent early relapse prior to starting siremadlin/DLI combination therapy. SA
siremadlin maintenance after the combination therapy is intended to prolong and enhance
the GvL reaction to eradicate residual leukemic blasts.
Methods: The estimand framework mandates a careful definition of the experimental
intervention according to the clinical question of interest. In this trial, the experimental
intervention follows a treatment strategy: Patients (pts) with AML in complete remission
(CR) post-SCT but at high risk for relapse based on pre-SCT risk factors will start
with siremadlin priming monotherapy. Pts who tolerate the siremadlin dose with no
evidence of GvHD are eligible to start siremadlin/DLI combination therapy. After combination
therapy, pts may continue with SA siremadlin maintenance. Depending on the treatment
journey, pts may receive only a part of the intervention’s sequence (e.g., pts in
siremadlin priming monotherapy who are not eligible for DLI combination will continue
the priming part). The clinical questions addressed in this trial are to determine
the recommended dose (RD) of siremadlin across all parts of the treatment strategy
and to assess preliminary efficacy regardless of the parts of the treatment strategy
pts received. The doses of interest were preselected based on SA studies and in pts
who received siremadlin after SCT.
Results: The proposed approach is tailored to investigate safety and efficacy of the
treatment strategy. The RD of siremadlin will be determined over 2 parts: A Bayesian
logistic regression model will guide dose finding of siremadlin during priming (Cycle
1) and a Bayesian time-to-DLT model will assess the siremadlin dose for combination.
Efficacy will be assessed by proportion of pts remaining in CR for at least 6 months.
Image:
Summary/Conclusion: In clinical practice, treatment decisions follow a strategy. The
estimand framework guided the design of a clinical study in AML post SCT to conceptualize
the assessment of a treatment strategy. Accordingly, safety and efficacy of the treatment
strategy as a whole will be assessed taking into account different patient journeys.
P584: OVERALL SURVIVAL WITH INTENSIVE CHEMOTHERAPY (IC) VS NON-IC IN PATIENTS (PTS)
WITH NEWLY DIAGNOSED (ND) AML FROM THE CONNECT® MYELOID DISEASE REGISTRY INELIGIBLE
FOR RANDOMIZED CLINICAL TRIALS (RCT)
H. Erba1,*, D. Pollyea2, M. Sekeres3, G. Garcia-Manero4,5, K. Seiter5, I. DeGutis6,
P. Kiselev6, A. McBride6, E. Yu6, G. Roboz7,8
1Duke University, Durham; 2University of Colorado, Aurora; 3Sylvester Comprehensive
Cancer Center, University of Miami Health System, Miami; 4The University of Texas
MD Anderson Cancer Center, Houston; 5New York Medical College, Valhalla; 6Bristol
Myers Squibb, Princeton; 7Weill Cornell Medicine; 8New York-Presbyterian Hospital,
New York, United States of America
Background: Pts with AML in RCTs often do not reflect the population seen in clinical
practice due to strict eligibility criteria.
Aims: To evaluate patient outcomes of IC vs venetoclax (VEN)-containing regimens based
on eligibility criteria from a recent RCT (the VIALE-A trial) in a broad cohort of
real-world pts with AML from the Connect® Myeloid Disease Registry (NCT01688011).
Methods: Pts were stratified into 3 groups based on the non-IC phase 3 VIALE-A trial
eligibility criteria: 1) “eligible” pts who would have met all VIALE-A inclusion criteria;
2) “unfit” pts who would have been ineligible for VIALE-A due to ≥ 1 of the following:
abnormal liver/kidney function, high ECOG, recent prior malignancy, comorbidities
score ≥ 2 by ACE-27, hepatic grade ≥ 1, AIDS grade ≥ 1; 3) “fit” pts who would have
been ineligible for a VEN-based regimen in VIALE-A because they would have qualified
for IC (defined as: ≤ 74 y of age, low ECOG, no apparent cardiovascular/renal comorbidities,
and did not meet any criteria in #2). Baseline characteristics were summarized by
eligibility group. Overall survival by group was estimated using the Kaplan–Meier
method. Induction regimens were categorized as IC (any regimen containing 7 + 3, MEC,
CLAG, FLAG) or VEN-based. Hazard ratios (HR) for induction regimens among each group
were estimated using Cox models adjusted for age, ELN risk, ECOG, frailty score, and
comorbidity index.
Results: Of 734 enrolled pts with AML (Dec 2021), most were male (61%) and white (84%);
median age 71 y (range, 55–97). Only 26% of pts (n = 192) were eligible for a non-IC
RCT, 45% (n = 327) were ineligible due to unfitness, and 29% (n = 215) were ineligible
due to overall fitness. The main reason for non-IC RCT ineligibility was high overall
comorbidity grade (n = 265 [36%]). At baseline, fit pts intended to undergo transplant
more often compared with unfit pts. Median duration of overall survival for eligible,
unfit, and fit pts was 14, 10, and 22 months, respectively (HR [95% CI], eligible
vs unfit, 0.02 [−0.19 to 0.22], P = 0.8735; eligible vs fit, 0.57 [0.33–0.81], P <
0.0001; unfit vs fit, 0.55 [0.33–0.77], P < 0.0001). Among unfit pts, those receiving
IC had significantly longer overall survival compared with pts receiving a VEN-based
regimen (median overall survival, 14 vs 6 months, respectively; HR, 0.51 [95% CI,
0.27–0.98]; P = 0.042; Figure). Unfit patients who received IC went on to transplant
more frequently than those who received VEN-based therapies (16% [n = 17] vs 1% [n
= 1], respectively). Eligible pts who received IC (n = 31) tended to have shorter
median overall survival (13 months) vs pts who received VEN-based therapies (n = 27;
23 months; HR, 1.45, 95% CI, 0.66–3.17; not sig.). Among fit pts, median overall survival
was 19 months for those receiving IC (n = 69) but could not be estimated for pts receiving
VEN-based therapies due to small sample size (n = 10).
Image:
Summary/Conclusion: The majority of pts with ND AML in the Connect® Myeloid Disease
Registry would have been ineligible for a non-IC RCT due to being too fit or unfit.
Among pts ineligible for an RCT due to unfitness, there was an association with increased
overall survival in pts receiving IC vs those receiving VEN-based therapies, and pts
in the IC group were more likely to receive a transplant. This analysis suggests that
RCTs may be excluding pts who appear unfit but can potentially tolerate IC and experience
improved survival outcomes.
P585: VENETOCLAX IN COMBINATION WITH INTENSIVE TREATMENT PROTOCOLS FOR PATIENTS WITH
HIGH-RISK ACUTE MYELOID LEUKEMIA – A MULTICENTER REAL-WORLD ANALYSIS
O. Wolach1,*, A. Frisch2, L. Shargian1, M. Yeshurun1, A. Apel3, V. Vainstein4, Y.
Moshe5, S. Shimony1, O. Amit5, Y. Bar On5, Y. Ofran6, P. Raanani1, B. Nachmias4, R.
Ram5
1Hematology, Rabin Medical Center, Petah Tikva; 2Hematology, Rambam Health Care Campus,
Haifa; 3Hematology, Shamir Medical Center, Zriffin; 4Hematology, Hadassah Medical
Center, Jerusalem; 5Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv; 6Hematology,
Shaare Zedek Medical Center, Jerusalem, Israel
Background: The addition of venetoclax, a selective BCL2-inhibitor, to low intensity
therapies was shown to improve the survival of patients with acute myeloid leukemia
(AML) that are ineligible for intensive induction chemotherapy. Based on the encouraging
anti-leukaemic properties of venetoclax and safety profile, several studies are currently
exploring venetoclax in combination with high-intensity chemotherapy in high-risk
AML scenarios such as ELN adverse-risk AML and in relapsed and refractoy (R/R) AML.
The addition of venetoclax to established intensive chemotherapy protocols in these
trials are generally reported to be associated with enhanced efficacy, frequently
at the cost of significant hematological and infectious toxicities that requires dose
modifications of both chemotherapy and venetoclax dosing
Aims: To assess the safety and efficacy of venetoclax in combination with intensive
treatment protocols in a ‘real-world’ setting.
Methods: A multicenter retrospective cohort study in six academic centers in Israel.
The decision to treat a patient with venetoclax in addition to an intensive treatment
protocol as well as the choice of protocol and dosing were at discretion of the treating
physician.
Results: Twenty-nine patients were included in analysis (median age 53.4 years). Most
patients were treated for R/R AML (n=27, 93%) with a median of one (0-5) previous
lines of therapy and 48% of patients (n=14) having prior allogeneic hematopoietic-cell
transplantation (HCT).
Most patients were treated with venetoclax in combination with FLAG-IDA protocol (n=25,
86%). Median follow-up was 6.3 (0.7-23.7) months. Platelet and neutrophil recovery
were observed at a median of 31 (95%CI 17.6-38.3) and 24 (95%CI 20-28) days, respectively.
Infectious complications were common (blood stream infections, 41% and invasive fungal
infections, 28%) and 30-day mortality was 14%.
Composite complete remission (CRc) was 72% for the entire cohort and 91% in patients
treated for post-HCT relapse. Incidences of relapse-free and overall survival at 12
months were 90% (95% CI 76%-98%) and 55% (95% CI 21%-63%), respectively.
Image:
Summary/Conclusion: The addition of venetoclax to intensive therapy is effective in
high-risk AML, especially in post-HCT relapse setting. Prophylaxis and surveillance
for infections are crucial.
P586: IADADEMSTAT COMBINATION WITH AZACITIDINE SHOWS ENCOURAGING SAFETY AND EFFICACY
DATA IN ELDERLY AND UNFIT AML PATIENTS
O. Salamero1,*, T. Somervaille2, A. Molero1, E. Acuña-Cruz3, J. A. Perez-Simon4, R.
Coll5, M. Arnan Sangerman6, B. Merchan7, A. Perez8, I. Cano3, R. Rodriguez-Veiga3,
M. Arevalo9, S. Gutierrez9, C. Buesa9, F. Bosch8, P. Montesinos3
1Dep. of Hematology, Vall d’Hebron University Hospital. Vall d’Hebron Institute of
Oncology (VHIO), Barcelona, Spain; 2The Christie Hospital NHS Foundation Trust, Cancer
Research Manchester Institute, Manchester, United Kingdom; 3Hospital Universitario
y Politécnico La Fe, Valencia; 4Hospital Universitario Virgen del Rocio; Instituto
de Biomedicina de Sevilla (IBIS)/CSIC/U. de Sevilla, Sevilla; 5ICO Hospital Dr Josep
Trueta, Girona; 6Institut Català d’Oncología (ICO), Hospital de Bellvitge, Hospitalet
de Llobregat; 7Hospital del Mar; 8Vall d’Hebron University Hospital. Vall d’Hebron
Institute of Oncology (VHIO), Barcelona; 9Oryzon Genomics SA, Cornella de Llobregat,
Spain
Background: Acute Myeloid Leukemia (AML) is an hematological malignancy with highest
incidence and lower survival rates in elderly. Previously reported ORR with hypomethylating
agents such as azacitidine alone is less than 30%. Recently, combinations of hypomethylating
agents with venetoclax have shown improved response ratios. However, refractoriness
or relapse is still a challenge for most patients (pts), particularly in the elderly
and the high-risk subpopulations. It has been shown that LSD1 is involved in malignant
transformation in AML. Iadademstat (iada) is a potent and selective LSD1 inhibitor
that has been administered to more than 100 oncology pts in Ph1 and Ph2 trials, showing
manageable toxicity and signs of preliminary activity.
Aims: ALICE (EudraCT 2018-000482-36) is a Phase IIa study to assess the safety, tolerability
and the recommended Phase II dose (RP2D) of iada (PO 5d ON, 2d OFF every week in 28d
cycles) in combination with azacitidine (sc., d1-5 and 8-9 every cycle) for the treatment
of adult patients diagnosed with AML, as per WHO 2016 classification, who have not
received prior treatment and are ineligible for intensive chemotherapy. We present
an update of the ongoing study six months after completion of recruitment.
Methods: Two doses of iada are studied in the trial: 60 and 90µg/m2/d. Besides the
safety and tolerability primary endpoints, secondary endpoints investigate anti-leukemic
activity including ORR according to ELN recommendations, time to response (TTR) and
duration of responses (DoR). Additional assessments include residual detectable disease
status, overall survival and PK/PD determinations.
Results: At EHA 2021, ALICE data from 27 pts were reported. In October 2021, recruitment
of the targeted 36 pts was completed. Patient baseline characteristics are shown in
Table 1. Main safety events by the end of 2021 included 348 adverse reactions (AR).
The most frequently reported AR was platelet reduction, observed in 44% of pts, however,
Grade ≥3 thrombocytopenia was already present at baseline in 61% of pts (Table 1).
Only three Grade 3-4 nonhematological treatment-related ARs were observed in two pts
(asthenia, dysgeusia and weight decrease). Among the 71 serious AR reported, only
2 were considered as potentially related to iada, corresponding to a differentiation
syndrome and a fatal intracranial hemorrhage, previously presented at EHA 2021.
By the time this abstract is written, among the pts intended to treat (34), 27 have
had at least 1 bone marrow evaluation after cycle 1 and are therefore evaluable for
efficacy. 78% of them (21 of 27) achieved an objective response; of which, 62% were
CR/CRi. Moreover, 80% of the CRi pts also achieved partial hematology recovery (CRh).
Among CR/CRi pts, 5 out of the 8 already assessed by MRD presented no residual detectable
disease by flow cytometry. DoR is encouraging with 77% of the CR/CRi lasting more
than 6 months, with a mean DoR increasing as the study progresses, with the longest
CR to date above 1,100 days. Iada at 90 µg/m2/d is the confirmed RP2D for the combination,
showing deep responses with manageable toxicity.
Image:
Summary/Conclusion: ALICE data confirms that the combination of iada with azacitidine
gives robust, fast and durable responses in unfit, previously untreated, AML patients.
Considering iada’s efficacy, pharmacologic properties, its manageable toxicity and
low anticipated drug-drug interactions, iada combinations may offer additional therapeutic
options for AML patients in first line or in the R/R setting.
P587: COVALENT-101: A PHASE 1 STUDY OF BMF-219, A NOVEL ORAL IRREVERSIBLE MENIN INHIBITOR,
IN PATIENTS WITH RELAPSED/REFRACTORY ACUTE LEUKEMIA, DIFFUSE LARGE B-CELL LYMPHOMA,
AND MULTIPLE MYELOMA
F. Ravandi-Kashani1,*, A. Kishtagari2, H. Carraway3, G. Schiller4, E. Curran5, B.
Yadav6, A. Cacovean6, S. Morris6, T. Butler6, J. Lancet7
1Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson
Cancer Center, Houston; 2Vanderbilt-Ingram Cancer Center, Nashville; 3Cleveland Clinic
Foundation, Cleveland; 4University of California Los Angeles, Los Angeles; 5University
of Cincinnati Medical Center, Cincinnati; 6Biomea Fusion, Inc, Redwood City; 7Department
of Malignant Hematology, Moffitt Cancer Center, Tampa, United States of America
Background: Menin, a protein involved in transcriptional regulation, impacting cell
cycle control, apoptosis, and DNA damage repair, plays a direct role in oncogenic
signaling in multiple cancers. Inhibition of menin is therefore a novel approach to
cancer treatment. Preclinical data of BMF-219, a highly selective, orally bioavailable,
small-molecule irreversible inhibitor of menin, show sustained potent abrogation of
menin-dependent oncogenic signaling in vitro and in vivo. BMF-219 exhibited a strong
anti-proliferative effect on various menin-dependent acute myeloid leukemia (AML)
cell lines, diffuse large B-cell lymphoma (DLBCL) cell lines representing Double/Triple
Hit Lymphoma (DHL/THL) & Double Expressor Lymphoma (DEL), and multiple myeloma (MM)
cell lines with diverse mutational backgrounds. BMF-219 also showed high potency ex
vivo in patient samples from MLL-rearranged and NPM1-mutant AML, THL and MYC-amplified
DLBCL, and bone marrow mononuclear cells from treatment-naive and relapsed/refractory
(R/R) MM.
Aims: COVALENT-101 (BF-MNN-101: NCT05153330) is a prospective, open-label, multi-cohort,
non-randomized, multicenter Phase I study evaluating the safety, tolerability, and
clinical activity of escalating doses of once daily oral BMF-219 in patients with
R/R acute leukemia (AL), DLBCL and MM who have received standard therapy.
The primary objective of the study is to determine independently for each cohort/indication,
the optimal biological dose (OBD)/ recommended Phase 2 dose (RP2D) of BMF-219 oral
monotherapy. Key secondary objectives include further evaluation of safety and tolerability,
characterization of the pharmacodynamics and pharmacokinetics of BMF-219, and assessment
of its antitumor activity based on best overall response rate (ORR), duration of response
(DOR), progression-free survival (PFS), and time to progression (TTP) per disease-specific
response criteria as assessed by the investigator. Food-effect studies will be performed
in DLBCL and MM patients at certain dose levels.
Methods: Utilizing an accelerated titration design, doses of BMF-219 will be escalated
in single-subject cohorts independently for each indication until 1 subject experiences
either a ≥ Grade 2 related adverse event or dose limiting toxicity (DLT). At that
point, the cohort will switch to a classical “3 + 3” design. Treatment will continue
in 28-day cycles until progression or intolerability. Expansion cohorts for each indication
will enroll patients to obtain further safety and efficacy data.
Patients with R/R AL, R/R DLBLC who received ≥ 2 but ≤ 5 therapies, and R/R MM who
received ≥ 3 therapies and have either failed or are ineligible for any standard therapies
are eligible. Patients must have ECOG PS ≤ 2, and adequate organ function. Key exclusion
criteria include known CNS disease involvement, prior menin inhibitor therapy, and
clinically significant cardiovascular disease.
Results: The study is ongoing.
Summary/Conclusion: The enrollment commenced in January 2022.
P588: IMPROVED OUTCOME OF RELAPSED/REFRACTORY ACUTE MYELOID LEUKEMIA BY ADDITION OF
LIPOSOMAL ADRIAMYCIN TO CLAG CHEMOTHERAPY
H. Yao1,*, C. Zhang1, J. Li2, X. Yin3, J. Rao1, X. Tan1, T. Chen1, L. Gao1, P. Kong1,
X. Zhang1
1Medical Center of Hematology, Xin Qiao Hospital, Chong Qing; 2Hematology, Changsha
Central Hospital Affiliated to Nanhua University, Hu Nan; 3Department of Hematology,the
923rd Hospital of the Joint Logistics Support Force of the People′Liberation Army
of China, Guang Xi, China
Background: Prognosis of refractory and relapsed acute myeloid leukemia (r/r AML)
is challenge and no standard of treatment exists. Cladribine based regimen in treatment
of r/r AML proved to be effective,while the total rate of response is limited.
Aims: To explore whether the addition of liposomal adriamycin can improve the response.
Methods: We registered a clinical trial, which is CLAG plus liposomal adriamycin (CLAG+PLD)
regimen for re-induction of r/r AML (ChiCTR1800017569), to observe the response, overall
survival (OS) and disease free suivival (DFS) of the patients. In addition to that,
CLAG alone as salvage therapy (no PLD group) for RR-AML in our medical center were
chosen as historical comparison
Results: To observe the response, overall survival (OS) and disease free suivival
(DFS) of the patients. In addition to that, CLAG alone as salvage therapy (no PLD
group) for RR-AML in our medical center were chosen as historical comparison.Of the
55 patients who were evaluable for remission after CLAG +PLD treatment(PLD group),
27(49.1%)achieved CR and negative of MRD, 12(21.8%)achieved CR and positive of MRD,
5(9.1%)achieved partial response, and 11(20%)no response. In Contrast, no PLD group
has inferior response rate,with the corresponding data is 24(46.6%), 6(10.9%),10(18.2%),and
13(23.6%)respectively (p= 0.008).As for the 2-year OS and DFS, the median OS and DFS
for PLD group has not reached and 10 months for noPLD group (p= 0.023 and p= 0.045
respectively). The safety observation did not find increasing toxicity in PLD group.In
subgroup analysis of PLD group, the response between FLT3-ITD mutated group and wild
group showed significant difference (p=0.006). Moreover, hematopoietic stem cell transplantation
(HSCT) group exert superiority in both OS and DFS compared to no HSCT group with 2-year
of follow-up(p<0.0001 and p<0.00001 respectively).
Image:
Summary/Conclusion: In conclusion, CLAG + PLD is a prospective salvage regimen for
r/r AML which brings improved response rates, 2-year OS and DFS and similar toxicities
compared to CLAG.
P589: AGE AND AGE-ADAPTED RISK FACTORS HIGHLY INFLUENCE SURVIVAL PROBABILITY IN AML
PATIENTS – A RETROSPECTIVE ANALYSIS
K. Nachtkamp1, C. Zahner1,*, A. Fricke1, T. Ulrych1, F. I. Schulz1, A. Kündgen1, P.
S. Jäger1, G. Kobbe1, U. Germing1
1Klinik für Hämatologie, Onkologie und Klinische Immunologie, Uniklinik Düsseldorf,
Düsseldorf, Germany
Background: Advanced age is strongly associated with inferior overall survival in
patients with acute myeloid leukemia (AML). Despite advances in modified treatment
regimen and genetic analyses of underlying mutations the prognosis for patients above
the age of 65 remains poor.
Aims: The aim was to determine patient-related risk factors and disease-associated
variables.
Methods: 915 AML patients (498 < 65 years, 417 ≥ 65 years) treated at the University
Hospital of Düsseldorf were analyzed. Patients were diagnosed between 2008 and 2020
comprising 484 males and 431 females. Median age was 63 years. Patients’ survival
was calculated using Kaplan-Meier-plots and multivariate regression analyses were
performed to identify prognostic factors for survival.
Results: Age itself proved to be an independent risk factor for survival: 6.9 months
within the group of patients ≥ 65 years, 166 months in patients < 65 years (p < 0.001).
In patients ≥ 65 years aberrant cytogenetic findings and an inferior ECOG status were
more frequent at the time of diagnosis. Comparison of survival within the same cytogenetic
risk group yielded significant differences between the two age-stratified cohorts:
Bearing cytogenetic aberrations associated with a favorable prognosis in the group
with patients aged over 65 was associated with a median survival of 18 months whereas
patients younger than 65 did not reach estimated median survival time (p < 0.001).
When comparing patients with the same ECOG-status, advanced age was the most decisive
prognostic factor for survival. This was highly significant in patients with ECOG
status 0. In the group with higher ECOG status an influence of age was notable but
of lower significance. In patients with pathologic lung function tests age turned
out to be significant for survival: 26 months (≥ 65 years) vs. not reached (< 65 years)
(p = 0.008). In patients with pathologic cardiac ultrasound findings results were
similar with 26 months (≥ 65 years) vs. not reached (< 65 years) (p = 0.001). Whether
AML was de novo or secondary affected survival probability only in the group aged
below 65 (166 months de novo AML, 69 months secondary AML).
Summary/Conclusion: Age has significant influence on patient- and leukemia-related
factors, thus modulating the impact of other parameters on the prognosis. We were
able to confirm the following independent negative predictive parameters on survival
in AML patients: increasing age ≥ 65 years, high-risk genetic aberrations, comorbidities
(lung, heart) and higher ECOG-status.
P590: PRELIMINARY SAFETY AND EFFICACY OF BGB-11417, A POTENT AND SELECTIVE B-CELL
LYMPHOMA 2 (BCL2) INHIBITOR, IN PATIENTS (PTS) WITH ACUTE MYELOID LEUKEMIA (AML)
J. Shortt1,*, S. Y. Tan2, P. Cannell3, T. F. Ng4, C. Y. Fong5, S. Ramanathan6, R.
Rajagopal7, S. Leitch8, R. Gasiorowski9, C. Grove10, D. Lenton11, P. Tan12, C. DiNardo13,
M. T. Ling14, S. Cheng14, Y. Liu14, M. Co14, W. Y. Chan14, D. Simpson14, A. H. Wei12,15
1School of Clinical Sciences, Monash University and Monash Health, Clayton; 2St Vincent’s
Hospital Melbourne, Fitzroy, Victoria; 3Fiona Stanley Hospital, Murdoch, Western Australia;
4Gold Coast University Hospital, Southport, Queensland; 5Austin Health, Heidelberg,
Victoria; 6The Saint George Hospital-Kogarah, Kogarah, New South Wales, Australia;
7Middlemore Hospital; 8North Shore Hospital, Auckland, New Zealand; 9Concord Repatriation
General Hospital, Concord West, New South Wales; 10Linear Clinical Research & Sir
Charles Gairdner Hospital, Nedlands, Western Australia; 11Orange Health Service (Central
West Cancer Care Centre), Orange, New South Wales; 12One Clinical Research, Nedlands,
Western Australia, Australia; 13University of Texas MD Anderson Cancer Center, Houston,
TX; 14BeiGene (Shanghai) Co., Ltd., Shanghai, China and BeiGene USA, Inc., San Mateo,
CA, United States of America; 15Department of Clinical Haematology, Peter MacCallum
Cancer Centre and Royal Melbourne Hospital, Melbourne, Victoria, Australia
Background: BCL2, a key regulator of apoptosis promoting cancer cell survival, is
aberrantly expressed in many hematologic malignancies. The BCL2 inhibitor, venetoclax,
is standard of care for the treatment of newly diagnosed AML in adults unfit for induction
chemotherapy. Although venetoclax-based treatments have improved outcomes for pts
with AML, there are concerns surrounding disease resistance after continued use and
adverse events (AEs) such as gastrointestinal toxicities and neutropenia. Clinical
findings have shown that 20-30% of pts using venetoclax are refractory to treatment,
and a majority of pts who initially achieve remission ultimately relapse (N Engl J
Med. 2020;383(7):617-629). The highly selective investigational BCL2 inhibitor, BGB-11417,
demonstrated more potent antitumor activity than venetoclax in preclinical studies
(Cancer Res. 2020;80[suppl, abstr]:3077).
Aims: To present preliminary safety and efficacy results of BGB-11417 in combination
with azacitidine in pts with AML enrolled in BGB-11417-103 (NCT04771130).
Methods: BGB-11417-103 is an ongoing phase 1b/2, global, multicenter, dose-escalation
and expansion study evaluating the combination of BGB-11417 and azacitidine in pts
with AML (either treatment-naïve [TN] unfit for intensive induction chemotherapy or
relapsed/refractory [R/R]), myelodysplastic syndrome, or myelodysplastic syndrome/myeloproliferative
neoplasm. Pts who received prior azacitidine or BCL2 inhibitors were excluded. For
pts with AML, the 10-day regimen consisted of BGB-11417 at 40 mg (Cohort 1), 80 mg
(Cohort 2), or 160 mg (Cohort 3) in combination with azacitidine (75 mg/m2 x 7 days).
In Cycle 1, a 4-day ramp-up of BGB-11417 was utilized starting at 1/8 of the target
dose. The window to assess dose-limiting toxicity (DLT) was up to Day 28 for nonhematologic
toxicities and Day 42 or initiation of Cycle 2 for hematologic toxicities. Treatment-emergent
AEs were graded per Common Terminology Criteria for AEs v5.0. Responses were assessed
using the 2017 European LeukemiaNet criteria (Blood. 2017;129(4):424-447). All pts
gave informed consent.
Results: As of 10 January 2022, 27 pts with AML were treated with the combination
therapy (6 in Cohort 1; 15 in Cohort 2; 6 in Cohort 3; Table). Median age was 80 yrs
(TN; 18 pts) and 70 yrs (R/R AML; 9 pts); 44% of pts had adverse karyotype. Median
prior line of therapy for R/R AML was 1 (range 1-2). At a median study follow-up of
2.1 months and median duration of treatment of 1.84 months (range 0.3-7.6), 2 of 23
evaluable pts experienced DLTs (Grade 4 neutropenia and Grade 4 thrombocytopenia at
the 80 mg x 10 day dose level), which did not meet the safety stopping protocol criteria.
Laboratory tumor lysis syndrome (TLS) was observed in 1 pt with a known history of
chronic kidney disease treated with 160 mg x 10 day; pt was asymptomatic and TLS resolved
within 4 days. Most common nonhematologic AEs were constipation (37%) and azacitidine
injection-site reaction (33%). Most common Grade ≥3 hematologic AEs were neutropenia
(44%), thrombocytopenia (41%), and anemia (37%). No pts required dose reductions of
BGB-11417. Majority (n=17; 63%) of pts are continuing study treatment. Ten pts discontinued
due to AEs (n=3), proceeding to transplant (n=3), consent withdrawal (n=2), or disease
progression (n=2). Preliminary CR/CRh rates for TN and R/R were 56% and 44%, respectively.
Seven of the 9 CRs were achieved by end of Cycle 1.
Image:
Summary/Conclusion: Preliminary results show that a 10-day regimen of BGB-11417 plus
azacitidine resulted in a majority of CR observed by the end of Cycle 1 and was well
tolerated in pts with AML.
P591: PRELIMINARY RESULTS OF A PHASE I STUDY OF CLIFUTINIB, A HIGHLY SELECTIVE, POTENT
ORAL FLT-3 INHIBITOR, IN PATINETS WITH FLT3-MUTATED RELAPSED OR REFRACTORY ACUTE MYELOID
LEUKEMIA
W. Yu1, X. Wei2, Z. Ge3, Y. Li4, Z. Jiang5, L. Yang6, L. Lin7, Y. Yao8, X. Deng9,
X. Du10, Y. Li11, T. Chen12, X. Feng13, J. Zhou14, M. Hou15, R. Fu16, J. Lan17, X.
Hu18, S. Huang19, J. Wang20, X. Du21, H. Yang22, H. Yang23, H. Wang1, L. Zheng24,
Z. Wang24, B. Liu24, N. Kang24, Y. Zhuang24, Y. Zhang24, J. Jin1,*
1The First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou;
2Henan Cancer Hospital, Zhengzhou; 3Zhongda Hospital, Southeast University, Nanjing;
4Zhujiang Hospital of Southern Medical University, Guangzhou; 5The First Affiliated
Hospital of Zhengzhou University, Zhengzhou; 6The Second Hospital of Shanxi Medical
University, Taiyuan; 7Hainan General Hospital, Haikou; 8Baoji Central Hospital, Baoji;
9Weihai Municipal Hospital, Weihai; 10Guangdong General Hospital; 11Sun Yat-sen Memorial
Hospital of Sun Yat-sen University, Guangzhou; 12Huashan Hospital, Shanghai; 13The
Affiliated Hospital of Qingdao University, Qingdao; 14Wuhan Tongji Hospital, Wuhan;
15Qilu Hospital of Shandong University, Jinan; 16Tianjin Medical University General
Hospital, Tianjin; 17Zhejiang Provincial People’s Hospital, Hangzhou; 18Beida Medical
Treatment Luzhong Hospital, Zibo; 19Xi’an Gaoxin Hospital, Xian; 20Aerospace Center
Hospital, Beijing; 21The Second People’s Hospital of Shenzhen, Shenzhen; 22Shunde
Hospital of Southern Medical University, Foshan; 23The 1st Affiliated Hospital of
He’nan University of Science and Technology, Luoyang; 24Sunshine Lake Pharma Co.,
Ltd, Dongguan, China
Background: An internal tandem duplication (ITD) in the juxtamembrane domain of FLT3
on chromosome 13q12, which accounts for 30% of patients with Acute Myeloid Leukemia
(AML), is associated with poor prognosis, including a lower complete remission (CR)
rate, a reduced duration of remission (DOR), a higher relapse rate, and a decreased
overall survival. Clifutinib, a highly selective oral FLT-3 inhibitor, demonstrated
potent activity against FLT3-ITD mutated human cell lines, both in vitro and in vivo.
Aims: This present study (NCT04827069) is a first-in-human study of Clifutinib, including
dose-escalation and dose-expansion phases, with a purpose to evaluate the safety,
tolerability, pharmacokinetics (PK) and antitumor activity in Chinese patients with
relapsed/ refractory (R/R) FLT3-mutated AML. Here we report the preliminary results
of the study.
Methods: AML patients aged≥18 years with refractory to ≥ 2 cycles of standard induction
chemotherapy, or relapsed after achieved remission from prior treatments were enrolled.
Previous use of FLT3 inhibitors was allowed. The primary endpoints included safety
and tolerability, including dose limiting toxicity (DLT), adverse event (AE). Secondary
endpoints were PK and anti-leukemic effect [CR/ CRh (Complete Remission with Partial
Hematologic Recovery) rate, Composite Complete Remission (CRc) rate, DOR, and overall
survival (OS)]. Clifutinib was administered orally on an empty stomach at 10~70 mg
daily, 28 consecutive days as a treatment cycle. Resumption of Clifutinib was not
allowed after hematopoietic stem cell transplantation (HSCT). Modified 3 + 3 and accelerated
titration design was utilized.
Results: As of December, 2021, fifty-seven patients were enrolled, including one at
10 mg, one at 20 mg, 6 at 55 mg, 34 at 40 mg, 15 at 70mg dose groups. The median age
was 52 years, and one third of the subjects received prior FLT-3 inhibitors treatments.
Two subjects discontinued from the study due to AEs. Eighteen subjects were evaluable
for DLT determination. Only one subject in the 55 mg group experienced DLT (Grade
3 QT prolongation), and MTD had not been reached. Fifty-three subjects were evaluable
for safety analysis. The most common treatment-related AEs of any grade included platelet
count decreased (69.8%), decreased white cell count (69.8%), neutrophil count decreased
(60.4%), anemia (43.3%), lymphocyte count decreased (32.1%). Plasma concentrations
of Clifutinib increased with increasing dose, with tmax between 2 and 6 hours. Accumulation
was observed following repeat dosing, with estimated t1/2 values of 68.3 to 114.2
hours based on accumulation index. Approximately dose-linear PK parameters were observed
over the dose range for both single and repeat dosing. Of 53 FLT3-ITD positive, evaluable
subjects, CR/CRh rate was 17.0% (9/53), and CRc rate was 45.3% (24/53). Among subjects
administered with Clifutinib 40 mg daily, the CR/CRh rate was 18.2% (6/33) with 3
patients achieved CR, median DOR of CR/CRh was 5.7 months, the CRc rate was 48.5%
(16/33), and median OS was 7.4 months. Among patients experienced only one prior regimen,
the CR/CRh rate of 40 mg dose group was 25% (4/16), the CRc rate was 50% (8/16), and
the median OS was 13.0 months.
Summary/Conclusion: Preliminary results of this phase 1 study demonstrated that Clifutinib
has an acceptable safety profile and promising antitumor activity, especially at the
dose of 40 mg daily. A confirmatory phase 3 study is planned to further evaluate the
efficacy and safety of Clifutinib at 40 mg daily in FLT3-mutated R/R AML.
P592: GILTERITINIB IN COMBINATION WITH VENETOCLAX, LOW DOSE CYTARABINE AND ACTINOMYCIN
D FOR FLT3 MUTATED RELAPSED OR REFRACTORY ACUTE MYELOID LEUKEMIA
A. Žučenka1,*, R. Pileckytė1, K. Maneikis1, V. Vaitekėnaitė1, L. Kevličius1, L. Griškevičius1
1Hematology, Oncology, Transfusion Medicine Center, Vilnius University Hospital Santaros
Klinikos, Vilnius, Lithuania
Background: A second generation FLT3 inhibitor Gilteritinib has become a standard
of care for FLT3 mutated relapsed or refractory acute myeloid leukemia (FLT3m R/R
AML). However, remission duration and overall survival remain unsatisfactory. Preliminary
results of doublet Gilteritinib + Venetoclax and triplet Gilteritinib + Venetoclax
+ Hypomethylator regimens are encouraging. Herein, we report the quadruplet regimen
consisting of Gilteritinib, Venetoclax, Low Dose Cytarabine and Actinomycin D (ACTIVE
+ G) for the treatment of FLT3m R/R AML in the clinical practice setting.
Aims: To evaluate the efficacy and safety of the ACTIVE + G regimen.
Methods: This was an observational, retrospective study. The patients were at least
18 years of age and had FLT3m R/R AML. All patients provided informed consent for
treatment and data collection. The ACTIVE + G regimen consisted of Venetoclax 600mg/d
p/o from day 1 up to day 28, Cytarabine 20mg/m2 s/c on days 1-10, Actinomycin D 12.5
µg/kg i/v on days 1-3 (on days 1-2 for patients ≥65 years) and Gilteritinib 120mg/d
p/o starting from either day 4 or day 10 and continued up to day 28. Indications for
stopping Venetoclax and Gilteritinib before day 28 were life-threatening infections
or faster hematological recovery in responding patients. A second ACTIVE + G cycle
was administered in non-responders without evidence of progressive disease after Cycle
1 or in responders with positive measurable residual disease (MRD). Responders after
ACTIVE + G could proceed to either allogeneic stem cell transplantation (alloSCT)
or maintenance therapy with Venetoclax, Low Dose Cytarabine and Gilteritinib. We evaluated
baseline characteristics, composite CR (CRc = CR + CRi + CRp), overall response (ORR
= CRc + MLFS), MRD negativity rates (<0.1% by multiparameter flow cytometry), overall
survival (OS), relapse-free survival (RFS), grade 3-5 non-hematological toxicities
and day 30 and day 60 mortality rates.
Results: Fifteen patients had been treated with ACTIVE + G, of whom 8 (53%) were female.
The median age was 66 years (34-87), median ECOG was 2 (0-3). FLT3-ITD mutation was
confirmed in 80% (12/15) of cases and 20% (3/15) had FLT3-TKD. The most common co-mutations
were NPM1 (53%, 8/15), DNMT3A (27%, 4/15), IDH1 (20%, 3/15) and IDH2 (20%, 3/15).
Three patients (20%) had adverse cytogenetics. The median number of previous treatment
lines was 2 (1-5). Twelve patients (80%) had received prior intensive chemotherapy,
3 patients (20%) had prior Venetoclax exposure and 9 patients (60%) had been previously
treated with FLT3 inhibitors (Midostaurin – 7, Sorafenib – 1, Gilteritinib – 1). Three
patients (20%) had relapsed after alloSCT. The majority of patients (80%, 12/15) had
received 1 cycle of ACTIVE + G, 3 patients (20%) had been treated with 2 cycles. The
CRc and the ORR were 67% (10/15) and 93% (14/15), respectively. MRD negativity was
confirmed in 50% (5/10) of CRc cases. The median OS and RFS were 8.6 and 12.9 months,
respectively. The most common non-hematological grade 3-5 adverse events were febrile
neutropenia (73%, 11/15), sepsis/bacteremia (53%, 8/15), pneumonia (33%, 5/15) and
secondary hemophagocytosis (13%, 2/15). Day 30 and day 60 mortality rates were 13%
(2/15) and 20% (3/15), respectively.
Image:
Summary/Conclusion: A quadruplet regimen ACTIVE + G demonstrated high efficacy in
this small group of R/R FLT3m AML patients irrespective of their prior exposure to
FLT3 inhibitors or Venetoclax. The main toxicities were infectious complications attributable
to prolonged myelosuppression. Prospective clinical trials are needed to verify our
results.
P593: MULTIDIMENSIONAL ANALYSIS OF THE B CELL RECEPTOR OFFERS INSIGHT INTO THE ONTOGENETIC
RELATIONSHIP OF MONOCLONAL B-CELL LYMPHOCYTOSIS WITH CHRONIC LYMPHOCYTIC LEUKEMIA
A. Agathangelidis1,*, C. Galigalidou2, A. Iatrou2, L. Zaragoza-Infante2, L. Scarfò3,
M. C. Maniou2, P. Ranghetti3, N. Pechlivanis2, V. Junet4, A. Skaftason5, M. Tsagiopoulou2,
F. Psomopoulos2, R. Rosenquist5, P. Ghia6, A. Chatzidimitriou2, K. Stamatopoulos2
1Department of Biology, Section of Genetics and Biotechnology, National and Kapodistrian
University of Athens, Athens; 2Institute of Applied Biosciences, Centre for Research
and Technology Hellas, Thessaloniki, Greece; 3Division of Experimental Oncology, Università
Vita-Salute San Raffaele/Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)
Ospedale San Raffaele, Milan, Italy; 4Institute of Biotechnology and Biomedicine,
Universitat Autònoma de Barcelona, Barcelona, Spain; 5Department of Molecular Medicine
and Surgery, Karolinska Institutet, Stockholm, Sweden; 6Division of Experimental Oncology,
Università Vita-Salute San Raffaele/Istituto di Ricovero e Cura a Carattere Scientifico
(IRCCS) Ospedale San Raffaele, Milan, Greece
Background: The clonotypic B-cell receptor immunoglobulin (BcR IG) is key to the pathogenesis
of CLL. Against that, however, scant information exists regarding the role of BcR
IG in early stages of CLL ontogenesis, i.e. monoclonal B cell lymphocytosis (MBL).
Aims: Here, we aimed to address this issue through the integrative analysis of: (i)
the BcR IG gene repertoire, (ii) the transcriptome, and, (iii) the BcR IG reactivity
profile, of both the high and the low-count MBL subtypes (HC-MBL and LC-MBL, respectively),
complemented by comparisons to CLL.
Methods: The study group comprised 17 individuals with LC-MBL and 14 with HC-MBL.
“CLL-like” cells were flow-sorted. BcR IG NGS data was annotated with IMGT/HighV-QUEST
and TRIPR. Clonotypes were defined as IG gene rearrangements with identical IG variable
gene and CDR3 amino acid sequence. Stereotypy analysis in MBL cases focused on abundant
clonotypes, which were compared against data from >39,000 CLL cases. Intraclonal diversification
was assessed through graph networks of all distinct nucleotide variants of each clonotype.
Transcriptome profiling was performed on polyA-selected RNA from 16 LC-MBL and 8 HC-MBL
cases; comparisons were undertaken against RNAseq data from 74 CLL patients. Count
normalization, differential gene expression and splice event analysis were performed
with DESeq2, Limma and SGSseq, respectively; gene enrichment analysis was performed
with Enrichr (p<0.05). BcR IG reactivity profiling was performed for 6 LC-MBL cases,
10 HC-MBL cases and 9 CLL cases against 123 autoantigens using a protein microarray;
validation tests were performed with ELISA.
Results: Clonality assessment revealed a monoclonal profile for most HC-MBL cases
(86%), whereas LC-MBL cases were mainly oligoclonal (53%). Stereotypy analysis revealed
a stronger immunogenetic connection of HC-MBL to CLL, since 19/47 abundant clonotypes
(40%) from HC-MBL were assigned to CLL stereotyped subsets as opposed to only 19/167
(11%) from LC-MBL. Intraclonal diversification analysis was undertaken for: (i) 26
monoclonal cases (from HC-MBL or LC-MBL), (ii) 10 oligoclonal LC-MBL cases and 73
monoclonal CLL cases. Significant differences (p<0.05) were evident between oligoclonal
MBL vs monoclonal MBL vs CLL; the former showed wide branching of variants due to
distinct somatic hypermutations (SHM). In contrast, monoclonal MBL and CLL displayed
progressively higher levels of variant connectivity due to the presence of shared
SHMs, likely reflecting stronger antigenic pressure. Comparative transcriptomic analysis
of either type of MBL vs CLL disclosed significant (p<0.05) down-regulation of genes
in the Notch, AMPK and BcR signaling pathways. Moreover, LC-MBL showed overexpression
of genes implicated in antigen processing and presentation and OXPHOS compared to
either HC-MBL or CLL. In addition, both MBL types were characterized by virtual absence
of alternative splicing, contrasting CLL where splicing events were identified in
14 genes of the BcR signaling pathway. Finally, BcR IG reactivity analysis revealed
different patterns among individual cases, however all examined cases from MBL and
CLL consistently bound a set of autoantigens, including myelin, centromere protein
B, β2-microglobulin, C-reactive protein and the nucleosome.
Summary/Conclusion: In conclusion, we highlight important differences between MBL
and CLL stemming from the attributes of the BcR IG and the related signaling pathway.
Overall, the reported findings allude to different maturation processes that likely
contribute to shaping a distinct BcR IG signaling capacity in MBL, particularly LC-MBL,
vs CLL.
P594: INCREASED FREQUENCIES OF IGH LOCUS SUICIDE RECOMBINATION POINTS ON CHRONIC LYMPHOCYTIC
LEUKEMIA WITH LOW RATE OF AID RELATED SOMATIC MUTATIONS, MYC OVEREXPRESSION AND SHORT
TELOMERES
I. Al Jamal1,2,*, M. Parquet1, H. Boutouil1, K. Guiyedi1, D. Rizzo1,3, M. Dupont1,3,
M. Boulin1,3, S. Aoufouchi4, S. Al Hamaoui2, N. Makdissy2, J. Feuillard1,3, N. Gachard1,3,
S. Peron1
1Centre National de la Recherche Scientifique (CNRS), UMR 7276/INSERM U1262,Limoges
University, Limoges, France; 2Faculty of Sciences, GSBT Genomic Surveillance and Biotherapy
Team, Lebanese University, Tripoli, Lebanon; 3Limoges University Hospital Center,
Biological Hematology Laboratory, Limoges; 4UMR 9019 Genome Integrity and Cancers,
Villejuif, France
Background: In normal B-cells, Activation Induced-cytidine deaminase (AID) is the
key enzyme for class switch recombination (CSR) and IGHV somatic hypermutation (SHM).
AID is also implicated in another IgH rearrangement, the Locus Suicide Recombination
(LSR). LSR occurs in activated B-cells and recombines the IgH locus between the switch
µ (Sµ) region and one 3’a2 regulatory region (3’a2RR) of the IgH locus. LSR results
in the complete deletion of the cluster of IgH constant genes. When LSR hits the active
IgH locus, it induces the loss of BCR expression and the death of the concerned B
cell. Chronic lymphocytic leukemia (CLL) is an indolent non-Hodgkin B-cell lymphoma.
Tumor CLL B-cells weakly express a B cell receptor (BCR) on the surface which is composed,
in the vast majority of cases, of immunoglobulins (Ig) of the mu (µ) and delta (δ)
isotypes and Ig class-switched CLL are rare, raising the question of abnormalities
in the Ig gene recombination machinery in this B-cell cancer.
Aims: Searching for abnormalities of IgH locus recombination in CLL, we investigated
CSR and LSR in CLL.
Methods: In this study, we used different molecular biology technics as high throughput
Sequencing (HTS) to analyze CSR and LSR junctions, IgHV, and PIM-1 mutation. Quantitative
reel time PCR (quantification of AID, cMYC and IgH locus transcripts). Samples are
from CLL patients (N=47) with more than 98% blood tumor cell infiltration and controls
consisted in healthy volunteers (HV) (N=9). Bioinformatics uses CSReport tool. Statistics
are from graph pad or R.
Results: CSR levels were lower in CLL than in HV samples contrariwise to LSR that
was found at comparable levels in both HV and CLL groups (Fig. A, B). Moreover, some
patients exhibited increased LSR counts. Because distribution of LSR counts was bimodal
with a valley at 27, that value being also the mean of LSR counts in HVs, we separated
CLL patients in two groups so called LSR-High and LSR-Low with that threshold of 27
LSR count per sample. We analyzed the diversity of the LSR junctions using Shannon
Index. LSR junction diversity was significantly higher in LSR-High than in LSR-Low
CLLs and HVs. This result is in opposition with the IgHV mutation rate since LSR-High
CLLs exhibited a stronger homology to IgHV reference sequences (unmutated CLLs). We
also found that LSR-High CLLs exhibited a marked lowest rate of AID off-target PIM1
mutations (Fig. C). AID expression was low and at comparable levels in both LSR-Low
and LSR-High CLLs. Because diversity of LSR junctions in LSR-High CLLs is evocative
of an on-going process, we analyzed the expression of productive and non-productive
transcripts of the constant part of IgH locus, which we found increased in LSR-High
when compared to LSR-Low CLLs. We also observed shorter telomere lengths and c-Myc
overexpression in the LSR-High group. Both shorter telomeres and c-Myc overexpression
are very likely to reflect an increased number of DNA replication cycles in the history
of the CLL tumor cell (Fig. D, E). Consistently, Kaplan Meyer curves of Treatment
Free Survival (TFS) show that TFS of LSR-High CLLs was significantly shorter than
the one of LSR-Low counterpart, an indication of a more rapidly evolving CLLs (Fig.
F).
Image:
Summary/Conclusion: Altogether, these results indicate the accessibility of IgH locus
and the proliferation in CLL patients with high rate and increased diversity of LSR
junctions could be increased in Myc dependent manner resulting in shorter survival
and pointing on an AID independent mechanism of IgH recombination.
P595: DISCOVERY OF NOVEL CIRCULAR RNAS (CIRCRNAS) OF THE APOPTOSIS-RELATED BAX AND
BCL2L12 GENES WITH A MULTIFACETED ROLE IN CHRONIC LYMPHOCYTIC LEUKEMIA, USING CUTTING-EDGE
NANOPORE SEQUENCING TECHNOLOGY
P. Artemaki1,*, N. Machairas1, P. Karousi1, V. Pappa2, A. Scorilas1, S. Papageorgiou2,
M. Batish3, C. Kontos1
1Department of Biochemistry and Molecular Biology, National and Kapodistrian University
of Athens; 2Second Department of Internal Medicine and Research Unit, University General
Hospital “Attikon”, Athens, Greece; 3Department of Medical and Molecular Sciences,
University of Delaware, Newark, DE, United States of America
Background: Circular RNAs (circRNAs), a novel RNA type generated by back-splicing,
are key indirect regulators of gene expression, with deregulated expression and established
involvement in several human malignancies. Many circRNAs have been shown to bind microRNAs
(miRNAs), thus making miRNAs unavailable to regulate protein-coding gene expression.
Moreover, some of them have been described to interact with chromatin and affecting
its structure. Despite their pivotal roles in the development and progression of cancer,
only three studies have investigated their expression and role(s) in chronic lymphocytic
leukemia (CLL), so far. The products of BCL2 and its homologues, including BAX and
BCL2-like 12 (BCL2L12), are implicated in CLL, as apoptosis regulators. Moreover,
expression levels of both BAX and BCL2L12 mRNAs are deregulated in CLL and are associated
with patients’ response to therapy and overall survival. However, to the best of our
knowledge, nothing is known about circRNAs produced by these two genes and their regulatory
potential.
Aims: We sought to further elucidate the contribution of both BAX and BCL2L12 in CLL
by unraveling the identity of their circRNAs in a B-cell leukemic cell line (EHEB)
and by studying in silico their potential interactions with miRNAs having an already
established role in CLL.
Methods: EHEB cells were propagated according to ATCC instructions. After total RNA
extraction, 2μg of total RNA were reverse-transcribed using random hexamers. Next,
nested PCR with divergent primers was performed, starting from each exon of BAX and
BCL2L12 genes; thus, only cDNAs from BAX and BCL2L12 circRNAs were amplified, respectively.
After having mixed and purified all nested-PCR products, nanopore sequencing libraries
were built according to an optimized protocol. Following nanopore sequencing and basecalling
in a MinION Mk1C 3rd-generation sequencer, sequencing reads were aligned and corrected
using Minimap2 and TranscriptClean, respectively. circRNA sequences were identified
using an in-house–built, PERL-based algorithm. A single-molecule resolution fluorescent
in situ hybridization (FISH) method called circFISH was used to visualize the circRNA
distribution in the cells. Lastly, miRDB was used to predict interactions of the most
abundant BAX and BCL2L12 circRNAs with miRNAs.
Results: We discovered 18 and 21 novel circRNAs produced by BAX and BCL2L12 genes,
respectively, in this leukemic cell line. The exon structure of these novel circRNAs
showed remarkable diversity, also comprising genomic regions that have previously
been considered as intronic, as they are not included in messenger RNAs (mRNAs). Interestingly,
one of the two most abundant BAX circRNAs is composed of a single exon, also spanning
two introns. Similar intron retentions have been observed in a few BCL2L12 circRNAs.
The gamut of miRNAs predicted to be bound by novel circRNAs includes miR-484, miR-181a-2-3p
and miR-214-3p. Particularly, the inhibition of miR-214-3p is associated with elevated
apoptosis in leukemic B-cells.
Summary/Conclusion: Alternative splicing and back-splicing mechanisms of the primary
BAX and BCL2L12 transcripts produce multiple distinct circRNAs in this B-cell CLL
cell line. Interestingly, one of the two most abundant circular transcripts of BAX
is a circRNA spanning three exons and two intervening introns of this gene. Lastly,
several miRNAs are predicted to be sequestered by circRNAs of either of these apoptosis-related
genes, including miR-181 family members that have been associated with chemosensitivity.
Our data suggest a multifaceted role of BAX and BCL2L12 circRNAs in B-cell CLL.
P596: CLINICAL IMPACT OF TP53 DISRUPTION IN CHRONIC LYMPHOCYTIC LEUKEMIA PATIENTS
TREATED WITH A BCR INHIBITOR. A CAMPUS CLL EXPERIENCE
R. Bomben1,*, F. M. Rossi1, F. Vit1, T. Bittolo1, A. Zucchetto1, R. Papotti2, E. Tissino1,
F. Pozzo1, M. Degan1, E. Zaina1, I. Cattarossi1, P. Nanni1, R. Marasca3, G. Reda4,
L. Laurenti5, J. Olivieri6, A. Chiarenza7, L. Ballotta8, A. Cuneo9, M. Gentile10,
F. Morabito11, A. Tafuri12, F. Zaja13, R. Foà14, F. Di Raimondo15, G. Del Poeta16,
V. Gattei1
1Clinical and Experimental Onco-Heamtology Unit, Centro di Riferimento Oncologico
IRCCS, Aviano; 2International PhD School in Clinical and Experimental Medicine, University
of Modena and Reggio Emilia; 3Hematology Unit, Department of Oncology and Hematology,
Azienda-Ospedaliero Universitaria (AOU) of Modena, Policlinico, Modena; 4Division
of Ematologia, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico di Milano,
Milano; 5Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione
Universitaria Policlinico A Gemelli di Roma, Roma; 6Clinica Ematologica, Centro Trapianti
e Terapie Cellulari “Carlo Melzi” DISM, Azienda Ospedaliera Universitaria S. Maria
Misericordia, Udine; 7Division of Hematology, Policlinico, Department of Surgery and
Medical Specialties, University of Catania, Catania; 8Dipartimento Clinico di Scienze
Mediche, Chirurgiche e della Salute, University of Trieste, Trieste; 9Hematology Section,
Department of Medical Sciences, University of Ferrara, Ferrara, Ferrara; 10Hematology,
Unit AO of Cosenza; 11Biothecnology Research Unit, AO of Cosenza, Cosenza; 12Department
of Clinical and Molecular Medicine and Hematology, Sant’Andrea - University Hospital
- Sapienza, University of Rome, Rome; 13Department of Medical, Surgical and Health
Sciences, University of Trieste, Trieste; 14Hematology, Department of Translational
and Precision Medicine, ‘Sapienza’ University, Roma; 15Division of Hematology, Policlinico,
Department of Surgery and Medical Specialties, University of Catania, Catania; 16Division
of Haematology, University of Tor Vergata, Roma, Italy
Background: In chronic lymphocytic leukemia (CLL), the presence of TP53 aberration
(mutations and/or deletion) predicts an increased risk of relapse and death after
chemo-immunotherapy (CIT). Although TP53 deletion and mutations mostly co-occur and
are considered equal prognosticators, the clinical impact of isolated or concomitant
mutations and deletions remains unclear in the context of targeted agents.
Aims: To investigate the clinical relevance of isolated or concomitant TP53 aberrations
in a large cohort of CLL patients treated with BCR inhibitors.
Methods: In the framework of an institutional Italian multicenter working group on
CLL (Campus CLL), a retrospective analysis of 229 CLL patients (51 treatment naïve,
and 178 relapsed/refractory) treated with ibrutinib was carried out. All patients
referred to a single institution for del17p analyses by FISH (167-kb 17p13 orange
probe, MetaSystems) and TP53 mutations by NGS, both analyses carried out on CD19-purified
(>85% pure) CLL samples within 6 months prior the start of ibrutinib treatment. The
median follow-up from ibrutinib treatment was 36.3 months (95% CI 29.5-41.5 months).
Overall survival (OS) and progression free survival (PFS) were computed from the start
of ibrutinib treatment.
Results: In the CLL cohort, 76 patients with del17p showed a trend for inferior OS
respect to wt cases (P=0.0662, Fig.1A), while a significant correlation was found
with PFS (P=0.0162). With regard to TP53 mutations, 296 TP53 mutations were found
in 126 patients (range of mutations/patients 1-11). As with CIT, TP53-mutated patients,
irrespective of VAF (Fig.1B), experienced a significantly worse OS and PFS than wt
cases also by univariate analyses (P=0.0160, and P=0.0378, respectively). The combination
of del17p with TP53 mutation data identified 95 cases with no TP53 aberrations, 8
cases del17p only, 58 cases TP53-mutated only, and 68 cases bearing both del17p deletion
and TP53 mutation (Fig.1CD). Only patients with concomitant TP53 mutations and del17p
experienced significantly shorter OS and PFS compared to TP53wt cases (P=0.0122, and
P=0.0076, respectively; Fig.1CD). Conversely, patients presenting a single aberration
showed no significant differences compared to wt patients (Fig.1CD). The simultaneous
presence of TP53 mutations and del17p remained an independent predictor factor both
for OS and PFS by multivariate analysis, together with the previous therapy lines
(0-1 vs. >1), and anemia. The evolution of TP53-mutated clones was assessed by longitudinal
NGS analysis of sequential PB samples collected from 38 patients (16 relapsed, and
22 on treatment) corresponding to 127 TP53 mutations. Among relapsed cases, 7 showed
a prominent expansion of the TP53-mutated clone, 8 remained stable, and the remaining
case displayed an evident decrease (Fig.1E). In non-relapsed patients, 3 cases presented
an increase of TP53 mutations, 13 remained stable, and 6 showed a reduction (Fig.1F),
with no significant difference compared to relapsed cases (P=0.0623, c2 test). BTK
and PLCG2 mutations were found in 9/16 (56%) relapsed cases and in 3/22 (14%) patients
still on ibrutinib (P=0.0492, c2 test). Of note, only 3/7 relapsed cases that presented
a positive selection for TP53 mutations showed the presence of BTK mutations at the
time of relapse.
Image:
Summary/Conclusion: This retrospective study indicates that only the concomitant presence
of TP53 mutations and deletion is an independent negative prognostic factor for OS
and PFS in patients with CLL on ibrutinib treatment; the simultaneous investigation
of del17p and TP53 mutations may lead to a more precise risk assessment.
P597: HIGH DOSE IRON IMPAIRS MALIGNANT B-CELL VIABILITY IN CHRONIC LYMPHOCYTIC LEUKEMIA
J. Bordini1,*, C. Lenzi2, L. Toscani1, P. Ranghetti1, E. Perotta1, L. Scarfò2, P.
Ghia2, A. Campanella2
1Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute; 2Division
of Experimental Oncology, Vita-Salute San Raffaele University, Milan, Italy
Background: Identification of new strategies to improve Chronic Lymphocytic Leukemia
(CLL) outcome is fundamental since the disease remains incurable. Malignant B-cells
show high levels of reactive oxygen species (ROS), which favors adaptation and survival
but also enhances sensitivity to pro-oxidant therapeutics. Redox homeostasis is affected
by iron balance since iron excess works as pro-oxidant, and cancer cells rearrange
iron trafficking proteins to promote iron uptake. This adaptation might be exploited
by exposing malignant cells to iron excess to cause ROS generation, lipid peroxidation,
and eventually ferroptosis. We previously demonstrated the feasibility of this strategy
in multiple myeloma and prostate cancer pre-clinical models.
Aims: We aim at exploiting high dose iron to negatively affect malignant cell survival
and increase therapeutic efficacy of current target therapies (BTK and BCL2 inhibitors).
We aim at dissecting the molecular mechanisms underlying iron toxicity and at testing
whether iron can improve immune dysfunctions.
Methods: CLL cell lines (MEC-1, MEC-2 and PCL-12), patients peripheral blood and bone
marrow mononuclear cells (PBMC and BMMC) and B lymphocytes purified from patients
PB were treated with 300 µM ferric ammonium citrate (FeAC) alone or in combination
with the BTK inhibitor ibrutinib (IBR, 5-10 µM) or the BCL2 inhibitor venetoclax (VNTX,
1.25-2.5 nM). We evaluated cell viability by annexin PI staining, lipid peroxidation
and ferroptosis by measuring malondialdehyde (MDA) levels and antioxidant gene expression
levels by qRT-PCR. We additionally evaluated T cell activation by measuring CD25 expression.
MEC-1 xenografts generated in immunodeficient RAG2-/-yc-/- mice were treated with
20 mg/Kg ferric carboximaltose.
Results:
In vitro, iron treatment impaired cell proliferation in all CLL cell lines analyzed,
inducing accumulation of MDA and cell death (p<0.01). In vivo, iron supplementation
reduced the amount of MEC-1 cells in bone marrow and spleen of xenograft mice as compared
to control saline-treated mice. In patients primary samples, iron induced cell death
of leukemic lymphocytes (CD19+CD5+) either as purified cells or within bulk PBMC (p<0.01).
Moreover, combination of iron with IBR or VNTX induced cell death at a higher extent
compared to each single drug or to iron alone (p<0.01). Leukemic cells in BMMC samples
were also iron sensitive and the effect became more evident upon combination with
IBR or VNTX (p<0.05). Neither iron nor iron-drug combinations induced cell death in
non-malignant cells. Iron also increased CD25 expression in CD8+ T cells and we are
currently exploring whether this might indicate that iron can also improve cytotoxic
mediated immune response against leukemic cells.
As single patient analysis revealed heterogeneity of response, we investigated whether
we might predict iron sensitivity by analyzing the antioxidant gene expression profile.
All CLL cell lines analyzed showed a lower basal expression of NFE2L2, HMOX1, and
GPX4 antioxidant genes than typical iron-resistant cell lines, such as prostate cancer
PC-3 cells, suggesting that differences in antioxidant gene expression levels may
mediate iron response and that this association is worth to be explored in CLL primary
samples to explain heterogeneous iron response.
Summary/Conclusion: Our pre-clinical studies suggests that exploiting iron toxicity
might be a valuable strategy to improve current target therapies in CLL. Additional
studies aimed at distinguishing iron sensitive patients from poor responders will
improve the translational potential of this therapeutic approach.
P598: EBI3/IL-27 DEPLETED MICROENVIRONMENT FAVOURS TUMOUR PROGRESSION IN CHRONIC LYMPHOCYTIC
LEUKAEMIA
I. C. Botana1,*, G. Pagano1, W. Marina1, P. M. Roessner2, B. Qu3, A. Ramsay4, B. Stamatopoulos5,
M. Seiffert2, J. Paggetti1, E. Moussay1
1Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg;
2Molecular Genetics, German Cancer Research Centre (DKFZ), Heidelberg; 3Biophysik,
Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany; 4School of
Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College
London, London, United Kingdom; 5Clinical Cellular Therapy Research Laboratory (LTCC),
Institut Jules Bordet, Brussels, Belgium
Background: Chronic Lymphocytic Leukaemia (CLL) is the most common adult leukaemia
in western countries. Unfortunately, CLL remains an incurable disease, evidencing
the urgency to develop novel effective treatments for this malignancy. CLL cells are
known to be highly dependent on interactions with non-malignant cells in their tumour
microenvironment (TME) for survival and proliferation. Such dependency highlights
the potential of immunotherapy as a therapeutic approach in CLL. Recently, several
pro-inflammatory cytokines have emerged as a powerful tool to reactivate the immune
system in a wide range of malignancies. IL-27 is a member of the IL-12 family of heterodimeric
cytokines reported to have pleiotropic functions during cancer development in different
malignancies. Based on the existing literature, we wondered whether IL-27 affects
the development and progression of CLL.
Aims: The goal of this study is to elucidate the role of IL-27 in CLL development
and progression.
Methods: We used a constitutive knockout mouse model of the EBI3 subunit of IL-27
to abrogate IL-27 expression in vivo. We performed adoptive transfer (AT) of TCL1
derived CLL cells into Ebi3-/- and WT mice, as well as generated the transgenic mouse
model Eµ-TCL1-Ebi3-/-.The splenic TME of these mice was characterized using flow cytometry.
In addition, in vivo neutralization of IL-27 using a blocking antibody was conducted.
Immunodeficient Rag2-/- mice were also injected with TCL1-derived CLL cells and either
WT or Ebi3-/- CD3+ T cells to assess the role of T-cells in leukaemia control in the
presence and absence of IL-27. Finally, the transcriptional differences among WT and
Ebi3-/- T cells were assessed using bulk-RNA sequencing, and their impact on T cell
cytotoxicity was investigated both in humans and mice using in vitro killing assays.
The serum levels of IL-27 in CLL patients and sick mice were quantified via ELISA
and compared to those of healthy controls.
Results: A strikingly enhanced CLL development and survival reduction was observed
when CLL was AT into Ebi3-/- mice, as well as in the transgenic Eµ-TCL1-Ebi3-/- mice
when compared to controls (A). Consistently, flow cytometry analysis revealed an increasingly
immunosuppressive splenic TME in the absence of EBI3, characterized by an enrichment
of highly activated and immunosuppressive Tregs and a terminally exhausted CD8+ T
cell subset. The aforementioned results were recapitulated in an IL-27 neutralization
experiment (B). Using Rag2-/- mice, we demonstrated that T cells from EBI3-/- mice
were less efficient in controlling CLL development (C). RNA sequencing revealed major
changes in CD8+ T cells transcriptional program, in particular a decreased expression
of crucial transporters (D). Finally, in vitro killing assays showed an increased
cytotoxicity of both human and murine T cells in the presence of IL-27, while serum
protein quantification showed a decrease in the levels of IL-27 in both CLL patients
(E) and sick mice (F) when compared to their healthy counterparts.
Image:
Summary/Conclusion: Overall, our results demonstrate the antitumor role of IL-27 in
CLL development and progression by promoting T-cell-mediated anti-tumor immunity,
as well as establish this cytokine as a potential immunotherapeutic agent in CLL.
P599: RARE GERMLINE VARIANTS IN ATM INFLUENCE THE PATHOGENESIS OF CLL
B. L. Lampson1, A. Gupta1, S. Tyekucheva2, Z. Wang2, N. Wojciechowska3, C. J. Shaughnessy1,
P. O. Baker1, S. M. Fernandes1, A. S. Kim3, J. R. Brown1,*
1Medical Oncology; 2Data Science, Dana-Farber Cancer Institute; 3Pathology, Brigham
and Women’s Hospital, Boston, United States of America
Background: Germline missense variants of unknown significance (VUS) are increasingly
identified in cancer-related genes by next generation sequencing. The ATM gene on
chromosome 11 is a tumor suppressor gene that is frequently deleted or mutated in
CLL, and also carries over 1,000 germline missense VUS.
Aims: We sought to evaluate whether germline ATM variants are more frequent in chronic
lymphocytic leukemia (CLL) compared to other hematologic malignancies and whether
they influence the clinical characteristics of CLL. We sought to determine whether
one of the most common VUS has an impact on ATM function.
Methods: In our hematologic malignancy clinic, we identified 3,128 patients (including
825 CLL patients), who underwent clinical-grade sequencing of the entire coding region
of ATM between 2014 and 2019. We evaluated the frequencies of germline ATM variants
in different hematologic neoplasms. In patients with CLL, we determined whether these
variants affect CLL-associated characteristics such as somatic 11q deletion. Finally
we generated an ATML2307F knock-in cell line to characterize the functional impact
of the L2307F VUS on response to etoposide treatment or irradiation.
Results: Germline ATM variants are present in 24% of patients with CLL, significantly
higher than in patients with other lymphoid malignancies (16% prevalence), myeloid
disease (15%), or no hematologic neoplasm (14%). CLL patients with germline ATM variants
are younger at diagnosis and twice as likely to have 11q deletion. The ATM variant
p.L2307F is present in 3% of CLL patients and is associated with a three-fold increase
in rates of somatic 11q-deletion. CLL in patients with two other common ATM VUS, p.S707P
and p.F858L, was also significantly enriched for ATM-aberrant disease. Cell-based
assays evaluating response to etoposide treatment or irradiation demonstrated that
ATM p.L2307F shows hypomorphic function.
Summary/Conclusion: Germline ATM variants cluster within CLL and affect the phenotype
of the CLL that develops, implying that some of these variants (such as ATML2307F)
have functional significance and should not be ignored. Further larger studies are
needed to determine whether these variants affect the response to therapy or account
for some of the inherited risk of CLL.
P600: HIGH-DEPTH RNA-SEQUENCING IDENTIFIES CD8+ STAT3 MUTATED T-LGLL PATIENTS AS A
DISTINCT BIOLOGICAL ENTITY WITHIN THE DISEASE HETEROGENEITY
G. Calabretto1,*, A. Binatti2, A. Teramo1, G. Barilà3, A. Buratin4, V. R. Gasparini1,
C. Vicenzetto1, E. Gaffo5, V. Trimarco6, L. Trentin1, M. Facco1, F. Vianello1, G.
Semenzato1, R. Zambello1, S. Bortoluzzi2
1Department of Medicine - University of Padua, Veneto Institute of Molecular Medicine;
2Department of Molecular Medicine, University of Padua, Padua; 3Hematology Unit, Ospedale
dell’Angelo, Mestre-Venezia; 4Department of Molecular Medicine and Department of Biology;
5CRIBI; 6Department of Medicine, University of Padua, Padua, Italy
Background: T-Large granular lymphocyte leukemia (T-LGLL) is a rare lymphoproliferative
disorder characterized by the clonal expansion of cytotoxic T-LGL. We recognized a
most common CD8+ subtype (CD8+ T-LGLL) and a CD4+ variant (CD4+ T-LGLL). STAT3 and
STAT5B activating mutations, the main genetic lesions, are linked to the immunophenotype
of the leukemic clone, T-LGLL clinical manifestations and disease prognosis. To schematize
the biological heterogeneity of the disease, 4 main T-LGLL subsets can be identified:
CD8+ STAT3M, CD8+ WT, CD4+ STAT5BM and CD4+ WT (M, mutated; WT, wild-type). Although
STAT3 and STAT5b may regulate the transcription of genes involved in disease severity,
the relationship between T-LGL immunophenotype (CD8+ or CD4+), STATs mutational status
and gene expression profiles (GEP), including non-coding-RNAs, has never been investigated.
Aims: The aim of the study was to disclose T-LGLL transcriptome unbalances accounting
for disease pathogenesis and clinical manifestations, by investigating GEP of leukemic
T-LGL from each disease subtype and healthy controls (CTR).
Methods: High-depth RNA-seq was performed with a HiSeq3000 (Illumina) on RNA extracted
from immuno-magnetically purified T-LGL of 20 T-LGLL patients (stratified in the 4
groups) and 5 CTR; 150 paired-end reads were generated (315 million reads/sample,
on average). RNA-seq data were analyzed for reads alignment and transcripts quantification
by CircComPara pipeline. Differentially expressed genes (DEGs) were assessed using
DESeq (adj. p<0.01) and validated by RT-qPCR.
Results: Unsupervised multi-dimensional scaling analysis of GEP clearly separated
T-LGLL from CTR samples. Of note, CD8+ STAT3M patients, characterized by a symptomatic
disease and reduced survival, resulted as a distinct biological entity with respect
to the other T-LGLL subgroups, that showed similar transcriptomic features. In detail,
we found 1612 DEGs in CD8+ STAT3M and 1707 DEGs in the other T-LGLL cases, with the
majority being down-regulated as compared to CTR. Of these, 641 genes were commonly
dysregulated in all T-LGLL subgroups, although a more marked dysregulation was observed
in presence of STAT3 mutations. 971 genes were differentially expressed uniquely in
CD8+ STAT3M cases, with 571 being differentially expressed also versus other T-LGLL
subgroups. Overall, dysregulated genes included tumor suppressors, protein kinases
and phosphatases, cytokines and receptors. Of note, two non coding-RNAs with a known
oncogenic role in other malignancies were up-regulated in LGLL samples.
Next, Gene Set Enrichment Analyses (GSEA) disclosed pathway dysregulation. In CD8+
STAT3M cases proteasome and Interferon signaling emerged among the most aberrantly
activated signaling pathways with respect to CTR, whereas the cell cycle checkpoints
pathway resulted to be activated as compared to the other T-LGLL subgroups.
Considering CD4+ STAT5BM T-LGLL, we observed the upregulation of a JAK/STAT axis negative
regulator, reported to be down-expressed in other STAT5B-related disorders characterized
by poor prognosis. This transcriptional signature might account for the peculiar indolent
clinical course of STAT5BM T-LGLL patients.
Summary/Conclusion: We identified new genetic players involved in T-LGLL pathogenesis
and disease severity, showing a peculiar GEP of CD8+ STAT3M cases, which are the most
symptomatic and treatment-requiring patients. Most importantly, the identified DEGs
are disclosing novel potential therapeutic targets to design innovative RNA-based
therapies for T-LGLL patients.
P601: TP53 ANALYSIS AND REPORTING IN CHRONIC LYMPHOCYTIC LEUKAEMIA: ARE LABORATORIES
IN COMPLIANCE WITH THE EUROPEAN RESEARCH INITIATIVE ON CLL (ERIC) RECOMMENDATIONS?
A. Cartwright1,*, S. Scott1, L. Whitby1
1UK NEQAS for Leucocyte Immunophenotyping, Sheffield Teaching Hospitals NHS Foundation
Trust, Sheffield, United Kingdom
Background: The presence of TP53 aberrations, specifically del(17p) and/or TP53 nucleotide
variants, in CLL are associated with adverse clinical outcomes and resistance to immunochemotherapy.
As such, analysis of TP53 has become part of routine clinical diagnostics prior to
initial and subsequent treatments, ensuring appropriate risk stratification and therapeutic
intervention. Deletions of TP53 are identified by fluorescent in situ hybridisation,
however, TP53 variants can be identified through various molecular techniques, including
Sanger sequencing and next-generation sequencing (NGS). ERIC recommendations outline
the requirements for testing, analysis and reporting of TP53 variants in CLL.
Aims: The aim of this study was to evaluate laboratory approaches to analysis and
reporting of TP53 variants in CLL in line with ERIC recommendations.
Methods: The UK National External Quality Assessment Service for Leucocyte Immunophenotyping
has provided external quality assessment for use of NGS gene panels in CLL since 2018.
Samples are manufactured from cell line material and one sample per annum is issued
for participating laboratories to evaluate using their in-house methodology. Between
2018-2021, three samples were formulated to contain TP53 variants and distributed.
Laboratories were requested to report methodology and variants of clinical significance,
in line with HGVS nomenclature at both the DNA and protein level. Additionally, details
of assay limit of detection (LOD), reference sequences and interpretation databases
utilised were also requested. Data returns were then evaluated relating to TP53 variant
analysis and reporting.
Results: In total, 34 (100%) participating laboratories reported the use of either
amplicon or capture based NGS methods, with assay LOD ranging from 1-15%, with a median
LOD of 5%. All participating laboratories sequenced the ‘minimum’ exons indicated
in the guidelines (exons 4-10), with 91.2% (31) laboratories sequencing ‘optimal’
exons 2-11. Furthermore, 97.1% (33) laboratories utilised the recommended Locus Reference
Genomic sequence LRG_321t1 (NM_000546), with one laboratory reporting use of LRG_321t3
(NM_001126114) variant transcript. The IARC TP53 locus-specific database for variant
interpretation was utilised by 70.6% (24) laboratories and despite recommendations,
use of dbSNP for filtering of polymorphic/neutral variants was undertaken by 76.5%
(26) laboratories. Across the 3-sample period, 97.1% (33) laboratories reported correct
DNA HGVS nomenclature and 58.8% (20) laboratories reported correct protein HGVS nomenclature.
Of the 14 laboratories reporting inaccurate protein HGVS nomenclature, 11 (78.6%)
reported minor nomenclature issues precluding full compliance.
Summary/Conclusion: In this study, there is a high level of consensus amongst laboratories
in the reporting and analysis of TP53 variants, except for protein nomenclature usage.
Guidelines serve to promote harmonisation and standardisation of processes within
laboratories and ERIC recommendations have achieved this in relation to TP53 variants
in CLL, ensuring that patients are appropriately stratified for correct therapeutic
interventions. Whilst there is currently a high level of consensus seen in analysis
and reporting, the recommendations are likely to evolve as evidence and understanding
of the genetic landscape in CLL therapeutics increases and further technological advances
are made. As such, laboratories will need to ensure continued adoption to reflect
new data reporting methods, treatments and standards of care for CLL patients.
P602: DISTINCT IMMUNE-RESPONSE PROFILE OF RICHTER TRANSFORMATION: HIGH EXPRESSION
OF IL-10, LAG-3 AND OTHER IMMUNE CHECKPOINT MOLECULES
Q. Chen1,*, A. Behdad2, S. Ma2, M. Schipma2, Y.-H. Chen2
1Pathology; 2Northwestern University, Chicago, United States of America
Background: Richter transformation (RT) is an aggressive progression of chronic lymphocytic
leukemia (CLL), often to diffuse large B cell lymphoma (DLBCL), with poor prognosis
and limited response to standard therapy. Our prior studies have shown differences
in immune checkpoint molecule expression between RT and CLL; in particular, expression
of programmed cell death 1 (PD1) in lymphoma cells and its ligands (PDL1) in background
immune cells. Significantly, we have shown high PD1 expression in RT predicts its
clonal-relatedness to CLL.
Aims: To investigate immune-response pathway dysregulation in RT by gene expression
profiling. To discover immune pathways and molecules that may serve as targets for
immune therapy. To discover biomarkers that may help identifying patients that will
benefit from checkpoint inhibitor therapy.
Methods: We studied 16 patients diagnosed with RT and 18 with CLL. We performed a
400-gene targeted NGS gene expression profiling on RT and CLL cases using the Oncomine
immune-response assay from ThermoFisher. Data analysis (RNA-seq with STAR and DESeq2)
was done by aligning quality-sufficient reads to the human genome and counting reads
for each gene. Gene normalization and differential expression were then calculated,
with adjusted p-values <0.05 determining statistically significant differentially
expressed genes. Next, a pathway analysis was performed (Metascape) to identify significant
pathways among the differentially expressed genes. We also performed PD1 and LAG3
immunohistochemical stains in all cases.
Results: Gene profiling analysis revealed significant differences in expression levels
of 89 immune-response genes between RT and CLL cases. The top upregulated genes in
RT comparing to CLL included several checkpoint pathway genes: LAG-3, HAVCR2/TIM3,
and CD274/PD-L1. Several immune modulation/immune suppressive genes were significantly
upregulated, in particular IL-10 and TGFb1. Other top upregulated genes included those
involved in innate immunity (S100A9, S100A8, IGSF6), and in cytokine signaling (CCR1,
IL-1B, IL-18). In addition to immune-response genes, MYC, TP63 and several cell proliferation
pathway genes were upregulated in RT. The top down-regulated genes in RT compared
to CLL included CD79B (B-cell receptor signaling), CD160 (checkpoint pathway), KLF2
and CD40LG (T-cell regulation), CIITA (IFNg signaling) and SELL (cell adhesion). To
further confirm these findings we performed LAG3 IHC. LAG3 was highly expressed by
large neoplastic B-cell in PD1-positive RT and only in scattered large cells in proliferation
centers of CLL, and largely negative in PD1-negative RT cases. Average positive neoplastic
cells were 67% in PD1+ RT (n=12), 2.8% in PD1- RT (n=4), and 10.8% in CLL (n=18) (P<0.05).
Summary/Conclusion: Using a targeted NGS gene profiling analysis, we demonstrate a
significant difference in expression levels of multiple immune-response genes in RT
vs CLL. In particular, we found upregulation in immune modulation genes and checkpoint
pathway genes PD1, LAG3 and TIM3, and immunosuppressive cytokine IL-10. Overexpression
of LAG3 in large neoplastic B-cells of RT, specifically in cases clonally-related
to CLL is an important finding that may serve as a diagnostic marker. Checkpoint molecules
LAG3 and TIM3 can serve as new immune-therapy targets for the treatment of RT. IL-10
blockade has been shown to enhance T-cell immunity, and may be a potential therapeutic
target.
P603: GUT MICROBIOME IN CHRONIC LYMPHOCYTIC LEUKEMIA SHOWS DEPLETION OF SHORT-CHAIN
FATTY ACIDS PRODUCING BACTERIA
T. Faitova1,*, M. Jørgensen2, R. Svanberg1, C. da Cunha-Bang1, E. E. Ilett2, C. MacPherson2,
C. Niemann1,3
1Department of Hematology; 2PERSIMUNE Centre of Excellence, Rigshospitalet, Copenhagen
University Hospital; 3Department of Clinical Medicine, University of Copenhagen, Copenhagen,
Denmark
Background: Recent studies have shown extensive crosstalk between our immune system
and gut microbiome (GM). The host immune system plays a vital role in the maintenance
of GM homeostasis by 1) establishing a balance between eliminating invading pathogens
and promoting the growth of beneficial microbes, 2) producing short-chain fatty acids
(SCFA), the main source of nutrition for the colon cells, and 3) modulating the immune
system by cytokine production. Mounting evidence shows that the GM of patients with
high rates of infection are characterized by an imbalance of bacteria, inducing proinflammatory
states and reduced capacity for SCFA synthesis. As chronic lymphocytic leukemia (CLL)
is, among others, also characterized by high rate of infectious complications and
an altered immune system, it is warranted to explore composition of the CLL microbiome.
Aims: We aim to investigate the hypothesis that deviation of the GM from homeostasis,
i.e. loss of ‘health promoting’ gut microbes and/or overgrowth of pathogenic bacteria,
distinguishes patients with CLL from the background population.
Methods: Feces samples of patients with CLL were collected, immediately fixated and
frozen within 72 h; total genomic DNA was sequenced. Feces samples of healthy controls
were chosen to match the CLL population with respect to age, demographic data, sample
collection method, and sequencing platform. Taxonomical profiling was done using an
in-house bioinformatics pipeline.
Results: A total of 61 CLL patients and 30 healthy individuals were included in the
study. We observed reduced GM alpha diversity, and depletion of bacterial members
of Lachnospiraceae and Ruminococcaceae families among the CLL patients when compared
to healthy individuals. Our data show that members of the Lachnospiraceae family (Anaerostipes
hadrus, Coprococcus comes, Blautia spp., Dorea spp.), and 3 members of Ruminococcaeae
family (Ruminococcus torques, Ruminococcus bromii, Faecalibacterium prausnitzii) were
among the most differentially abundant bacterial species between the microbiomes of
healthy individuals and CLL. Their mean proportions were shown to be significantly
higher in healthy microbiome samples. As further differentially abundant species we
observed Bacteroides sp. and Alistipes finegoldii, which both demonstrated significantly
higher mean proportions in CLL microbiomes (Fig 1).
Image:
Summary/Conclusion: To sum up, the CLL microbiomes in comparison to healthy controls
demonstrated lower enrichment of Lachnospiraceae and Ruminococcaceae families, the
major SCFAs-producing bacterial taxa reported to have a protective effect against
inflammation. This supports the notion that proinflammatory risk factors identified
in other cohorts with GM dysbiosis are also present within CLL patients. As CLL represents
an antigen driven malignancy with immune dysfunction, we hypothesize that GM dysbiosis
could both be implicated in the pathogenesis of CLL, through antigenic drive, and
contribute to the distortion of the immune system in CLL.
Notably, both immune dysfunction and treatment (e.g. antimicrobials) may influence
the CLL microbiome itself and therefore confound cross-sectional clinical studies.
We therefore plan to investigate the interaction between microbiome and CLL development
in the TCL1 mouse model of CLL. Also, identification of potential mechanistic links
between the GM and the microenvironment in CLL should be investigated, e.g. extravesicular
vesicles or cytokines released from or impacted by the GM. In addition, modulation
of the microbiome in animal models may help to establish causal connections between
the GM and CLL development.
P604: CATALASE EXPRESSION IN LEUKEMIA CELLS IS CONTROLLED BY GENETIC AND EPIGENETIC
MECHANISMS
M. Galasso1,*, E. Dalla Pozza1, R. Chignola2, S. Gambino1, C. Cavallini3, A. Pilatone1,
F. M. Quaglia4, O. Lovato3, I. Dando1, G. Malpeli5, M. Krampera4, M. Donadelli1, M.
G. Romanelli1, M. T. Scupoli1,3
1Department of Neurosciences, Biomedicine and Movement Sciences; 2Department of Biotechnology;
3Research Center LURM, Interdepartmental Laboratory of Medical Research; 4Department
of Medicine, Section of Hematology; 5Department of Surgery, Dentistry, Pediatrics,
and Gynecology, University of Verona, Verona, Italy
Background: Chronic lymphocytic leukemia (CLL) is the most prevalent form of leukemia
in Western countries. It is an incurable disease characterized by an extremely variable
clinical course and response to treatment. We have recently shown that high catalase
(CAT) expression identifies a more aggressive clinical behavior in CLL. However, molecular
mechanisms controlling catalase expression in leukemia cells are still poorly characterized.
The rs1001179 single nucleotide polymorphism (SNP) as well as DNA methylation in the
CAT promoter have been shown to regulate CAT expression in various cell types.
Aims: With the aim to characterize regulatory mechanisms underlying differential expression
of catalase in CLL, in this study we investigated the role of rs1001179 SNP and CpG
Island II methylation encompassing this SNP in the regulation of CAT expression.
Methods: Peripheral blood mononuclear cells (PBMC) from 55 CLL patients and 50 healthy
donors (HD) were used for the study. CAT mRNA levels were measured using quantitative
reverse transcription polymerase chain reaction (qRT-PCR). DNA samples were genotyped
for the rs1001179 SNP using Restriction Fragment Length Polymorphism (RFLP)-PCR. Bioinformatic
analysis was used to predict transcription factor (TF) binding to SNP sequences. Chromatin
Immunoprecipitation (ChIP)-qPCR was performed to validate predicted TF binding sites.
Methylation levels of CpG sites within the CAT promoter was determined by pyrosequencing
of bisulfite-converted DNA.
Results: The rs1001179 SNP genotyping showed that CLL cells harboring the T allele
exhibited a significantly higher catalase expression compared with cells bearing the
CC genotype. Interestingly, bioinformatic analysis predicted that the SNP provide
distinct binding sites for various TF, including ETS-1 and GR-β. ChIP assay confirmed
that CAT promoter harboring the T -but not C- allele was accessible to ETS-1 and GR-β,
but not to the other analyzed TFs. In addition, CLL cells exhibited lower methylation
levels within the CAT promoter compared with HD B cells, in line with the higher catalase
mRNA and protein levels expressed by CLL versus HD B cells. Moreover, methylation
levels negatively correlated with CAT expression in CLL cells. Remarkably, inhibition
of methyltransferase activity in leukemic cells induced a significant increase of
CAT mRNA and protein levels, thus showing that DNA methylation could control catalase
expression in CLL. Moreover, expression of DNA methyl transferase 1 (DNMT1) resulted
significantly reduced in CLL cells compared with HD B cells and inversely correlated
with CAT expression in CLL, thus suggesting that differences in methylation levels
underlying catalase expression could be driven by the DNMT1 enzyme. Finally, modeling
analysis showed that the CT/TT genotypes exhibited a lower methylation and a higher
CAT expression level, suggesting that the rs1001179 T allele and methylation cooperate
in inducing CAT gene expression.
Summary/Conclusion: Our data show that SNP as well as methylation of the catalase
promoter are involved in controlling catalase expression in CLL. The key advance of
this study is to provide new insights into the regulatory mechanisms underlying differential
expression of catalase in leukemic cells, which is of clinical relevance in CLL. Moreover,
our study may form the basis for future challenges aimed at developing combinatorial
therapies targeting catalase regulatory pathways.
P605: T-CELL PROLYMPHOCYTIC LEUKEMIA: MOLECULAR CHARACTERIZATION OF A COHORT OF 18
PATIENTS AND EVALUATION OF BORTEZOMIB AS A POSSIBLE THERAPEUTIC STRATEGY
V. R. Gasparini1,*, A. Martines2, G. Barilà3, A. Teramo1, G. Calabretto1, C. Vicenzetto1,
S. Carraro4, V. Trimarco4, L. Trentin4, M. Facco4, G. Semenzato1, L. Bonaldi2, R.
Zambello1
1Department of Medicine, University of Padova and Veneto Institute of Molecular Medicine;
2Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS,
Padova; 3Hematology Unit, Ospedale dell’Angelo, Mestre-Venezia; 4Department of Medicine,
University of Padova, Padova, Italy
Background: T-cell prolymphocytic leukemia (T-PLL) is a rare and aggressive mature
T-cell malignancy characterized by a progressive clinical course and resistance to
chemotherapy, whose pathogenesis is still largely unknown. Recurrent somatic mutations
were detected within the JAK/STAT axis (i.e., JAK3 and STAT5b genes). T-PLL patients
present with progressive lymphocytosis, splenomegaly and lymphadenopathy. New therapeutic
strategies are needed to overcome the overall survival of two years observed in T-PLL
patients even after treatment with alemtuzumab and consolidation with allogeneic stem
cell transplantation. Ex-vivo drug sensitivity and resistance tests suggested proteasome
inhibitors as novel candidates for the treatment of T-PLL.
Aims: The investigation of the molecular features of T-PLL aimed at understanding
whether JAK/STAT genetic lesions might have a role in the clinical management of patients.
Ex-vivo treatment with Bortezomib (Bz), a proteasome inhibitor, was performed to test
the affordability of this compound as a new option for T-PLL therapy.
Methods: Sanger sequencing was performed on peripheral blood mononuclear cells (PBMC)
of 18 T-PLL patients to detect the presence of somatic mutation in the hotspot regions
of JAK3 and STAT5b genes. Patients were recruited at the Padova University Hospital.
Immunophenotypic characterization of the leukemic clone was performed in all cases,
while 11 patients underwent cytogenetic analysis upon mitogen stimulation. PBMC of
10 patients were cultured for 48h with different concentrations (1.3, 2.6, 5.2 and
7.8nM) of Bz according to literature. Annexin staining and western blot analyses were
performed at both 24h and 48h to assess the efficacy of the treatment.
Results: At least one mutation within the JAK/STAT pathway was detected in 39% (7/18)
of patients. Consistent with the literature’s data, N642H variant was the most recurrent
STAT5b mutation. Conversely, the most frequent variant in JAK3 gene was N564S, which
has never been detected in T-PLL and not listed in the COSMIC database. Cytogenetic
analysis revealed a complex karyotype in 9/11 (82%) patients with specific primary
T-PLL aberrations: inv(14)(q11q32) or variant rearrangements in 78% (7/9) and t(X;14)(q28;q11)
in 2 cases. Known secondary alterations such as those involving chromosome 8 and deletion
of 11q22 were found in 89% (8/9) and 67% (6/9) of cases, respectively. Analysis of
the clinical data did not reveal a correlation with the patient’s mutational status.
Ex-vivo treatment with Bz induced apoptosis of the leukemic clone in a dose and time
dependent manner. Compared to the untreated condition (16%), Bz concentration of 5.2nM
increased the mortality of 1.7 (27%) and 4 (63.5%) times at 24h and 48h, respectively.
The apoptosis was confirmed also at protein levels by a trend of caspase 9 and PARP
cleavages upregulation at 48h. No difference in the sensitivity to treatment with
Bz was observed between mutated and wild-type patients.
Summary/Conclusion: No correlations between recurrent lesions within the JAK/STAT
axis and clinical outcome were found. This points against a pathogenetic role of these
mutations in T-PLL, conversely from other leukemias where STAT mutations are regarded
as prognostic markers. This hypothesis is further sustained by the same response to
the treatment with Bz. Since the treatment of T-PLL mostly remains an unmet clinical
need, the finding of an increased apoptosis in Bz-treated cells paves the way for
a deeper study on molecular mechanisms involved in the apoptosis of the leukemic clone.
P606: META-ANALYSIS OF LOW ALLELIC BURDEN C481 BTK-MUTATIONS CALLS FOR CAUTION IN
IBRUTINIB RESISTANCE EVALUATION
M. H. Hansen1,2,*, S. R. Veyhe1,2, N. Abildgaard1,2, C. G. Nyvold1,2, H. Frederiksen2
1Hematology-Pathology Research Laboratory, Research Unit for Hematology and Research
Unit for Pathology; 2Department of Hematology, Odense University Hospital, Odense,
Denmark
Background: Introduction of the orally administered tyrosine kinase inhibitor ibrutinib,
a drug approved for relapsed or refractory mantle cell lymphoma in late 2013 and chronic
lymphocytic leukemia in 2014, has changed treatment algoritms for B-cell malignancies
and improved progression-free survival. Ibrutinib irreversibly targets Bruton’s tyrosine
kinase (BTK), thus interrupting B-cell receptor signaling. However, it is known that
mutations in the BTK gene, located on the X chromosome, can mediate acquired resistance,
specifically modifying cysteine residue 481 to serine (C481S, X:101356176, GRCh38).
Aims: Despite this knowledge, it is still unknown how low mutational burden can contribute
to general resistance towards ibrutinib treatment. Thus, we performed a meta-analysis
based on previous cardinal studies to investigate the collective and gender-specific
distribution of variant allele frequencies.
Methods: We implemented meta-analysis of single-nucleotide variant calls from seven
studies involving whole-exome sequencing (Woyach et al. 2014 and 2017, Maddocks et
al. 2015, Ahn et al. 2017, Kanagal-Shamanna et al. 2019, Gángó et al. 2020, Bödör
et al. 2021) on mutational profiling concerning ibrutinib resistance and CLL. Four
of these studies contained gender specific data. Computational analyses, statistics,
and visualization were performed in Wolfram Language, R, and GraphPad Prism.
Results: From the available data included in these studies, we were able to base our
analyses on 226 non-BTK mutations and 170 BTK mutations within a variant allele frequency
(VAF) range of 0.0001–0.9, of which 93 mutation candidates from males (VAF < 0.9)
and 77 from females (VAF < 0.44) were observed. Extrapolative analysis of allele frequency
distribution from all other variants (excl. BTK) in the studies revealed an exponential
increase in the number of low burden variants below 10% VAF (R2>0.99). These variants
were not significantly different between genders (PMann-Whitney=0.24, nmale=130, nfemale=73).
In contrast, a significant difference was observed between male variant allele frequencies
(PMann-Whitney=0.005) and females owing to BTK hemizygosity in males. However, this
significance persisted when female BTK mutations were transformed to reflect a single
copy (VAF/2, PMann-Whitney=0.015, ΔVAF=0.05) for gender comparison and even when observations
for males were alternatively transformed to reflect two copies (2*VAF, PMann-Whitney<0.001,
ΔVAF=0.19). The previously discovered threshold of 10% was set as the detection limit.
Summary/Conclusion: Defining specific molecular contributions in drug resistance is
imperative for the correct use of targeted therapy. It has been questioned how the
low allelic burden of BTK mutations plays a role. Therefore, it is crucial to investigate
the lower limit of detection. The meta-analysis suggests that variants below a certain
threshold, here empirically 10%, may in many cases be false-positive variants, in
agreement with other reports involving WES in general, introduced by erroneous base
calling, PCR errors, etc. Although intriguing, it remains to elaborate whether the
haploinsufficiency of BTK in males skew in the degree of ibrutinib resistance.
P607: A NOVEL LYMPH NODE-MIMICKING 3D CULTURE SYSTEM DISPLAYS LONG-TERM T CELL-DEPENDENT
CLL PROLIFERATION AND SURVIVAL
M. Haselager1,2,3,4,*, E. Perelaer1, A. Kater2,3,4,5, E. Eldering1,2,3,4
1Experimental Immunology, Amsterdam University Medical Center; 2Lymhoma and Myeloma
Center Amsterdam, LYMMCARE; 3Cancer Center Amsterdam; 4Amsterdam Infection & Immunity
Institute; 5Hematology, Amsterdam University Medical Center, Amsterdam, Netherlands
Background: Primary chronic lymphocytic leukemia (CLL) cells, despite originating
from a proliferative disease, rapidly undergo apoptosis in vitro in the absence of
microenvironmental survival signals1. No current system permits long-term expansion
of CLL cells in vitro due to difficulties of mimicking a physiological microenvironment.
The lymph node (LN) is the critical site of in vivo CLL proliferation and is also
thought to play a key role in the development of resistance to targeted agents2,3.
Developing an in vitro culture system for CLL with pathophysiological relevance will
greatly benefit studies of CLL proliferation, microenvironment, clonal outgrowth and
relevant drug screening.
Aims: To design an in vitro CLL culture system that incorporates key aspects of the
CLL LN; contribution of non-CLL cells and cytokines, and induction of drug resistance
and prolonged proliferation of CLL cells.
Methods: Primary CLL cells were cultured in ultra-low attachment (ULA) plates in parallel
to standard 2D cultures. PBMCs were cultured with or without T cells (in a specific
CLL:T cell ratio to mimic the LN composition), or PBMCs were co-cultured with primary
lymph node fibroblasts. CLL cells were either stimulated directly with a B cell cocktail
(BCC) consisting of IL-2, IL-15, IL-21 and CpG and/or indirectly via a T cell stimulation
of anti-CD3/CD28.
Results: Compared to 2D cultures, 3D cultures showed several important CLL LN features.
First, significantly increased CLL proliferation independent of IGHV mutation status,
which was abrogated in the absence of T cells, or by JAK inhibitors. Notably, treatment
with BTK inhibitors significantly inhibited CLL proliferation and resulted in disintegration
of the 3D spheroid architecture. Second, co-culture with LN-derived stromal cells
further increased CLL proliferation, reaching a maximum of 8 generations. Third, 3D
cultures could be expanded approximately 3-4-fold over a course of 6 weeks using the
3D model. Fourth, when PBMCs were stimulated with BCC, spheroids developed proliferation
center-like structures after 4 weeks of culture where T cells localized together with
enrichment of Ki-67+ CLL cells. Finally, either B or T cell stimulation resulted in
an induction of venetoclax resistance, showing that T cells are able to confer drug
resistance to CLL cells in this model.
Summary/Conclusion: We present a 3D culture system that underlines the role of T cells
in sustained CLL proliferation, permitting investigation of CLL cells in the context
of a protective niche consisting of multiple cell types, thereby opening up new avenues
for clinically useful applications.
P608: DISTINCT P53 PHOSPHORYLATION PATTERNS IN CHRONIC LYMPHOCYTIC LEUKEMIA PATIENTS
ARE REFLECTED IN CIRCUMJACENT PATHWAYS’ ACTIVATION UPON DNA DAMAGE
M. Pesova1,2,*, V. Mancikova1,2, R. Helma1,2, S. Pavlova1,2, V. Hejret1, P. Taus1,
J. Hynst1, K. Plevova1,2,3, J. Kotaskova1,2,3, J. Malcikova1,2, S. Pospisilova1,2,3
1Central European Institute of Technology (CEITEC), Masaryk University; 2Department
of Internal Medicine - Hematology and Oncology, Faculty of Medicine of Masaryk University
and University Hospital; 3Institute of Medical Genetics and Genomics, Faculty of Medicine,
Masaryk University, Brno, Czechia
Background: Presence of defects in the TP53 gene represents a clinically relevant
biomarker in chronic lymphocytic leukemia (CLL). Besides gene mutations and deletions,
the functionality of p53 protein can be disrupted by other mechanisms, such as aberrant
phosphorylation.
Aims: To uncover how p53 phosphorylations affect its functions in CLL cells.
Methods: P53 phospho-patterns were analyzed in 71 TP53-intact primary CLL samples
using Zn2+-Phos-tag™ SDS-PAGE after induction of p53 protein by DNA damage (fludarabine
and doxorubicin treatments). Differences in gene expression were studied using RNA
sequencing and compared to CLL cells carrying the mutated TP53 gene. The ability of
p53 to activate transcription of its target genes (BAX, BBC3, CDKN1A, GADD45A) was
assessed by qPCR. The presence of gene mutations was analyzed using a targeted NGS
panel.
Results: While fludarabine induced a consistent phospho-pattern, two distinct p53
phosphorylation profiles were identified upon doxorubicin treatment. Profile I featured
heavily phosphorylated p53 protein, while only low phosphorylation occurred in profile
II. On the transcriptomic level, samples from the profile II were less capable of
activating p53 target genes upon doxorubicin treatment, thus resembling TP53-mutant
cells, whereas proper p53 signaling was triggered in profile I. Consistently, induction
of the studied downstream effector genes CDKN1A, BAX, BBC3 and GADD45 after doxorubicin
treatment was lower in profile II than in profile I samples but still higher than
in TP53 mutants. The differences between the two phoshoprofiles were also observed
in untreated cells: the expression of miR-34 in profile II showed an intermediate
pattern between profile I and TP53-mutant cells. Furthermore, profile II samples had
significantly higher basal p53 protein levels than profile I samples, where p53 was
generally detectable only upon DNA damage. The PROGENy analysis of the basal activity
of selected cancer-related pathways in untreated cells pointed to the different regulation
of the hypoxic pathway: the activity of hypoxic pathway in profile II samples was
in between high levels found in TP53 mutants and low levels in profile I. Finally,
we have found ATM locus/gene defects more frequently in profile II.
Summary/Conclusion: Our study suggests that wt-TP53 CLL cells with less phosphorylated
p53 show TP53-mutant-like behavior after DNA damage. Hypophosphorylation of p53 protein
and the corresponding lower ability to respond to DNA damage are linked to ATM locus
defects and to the higher basal activity of the hypoxia pathway.
The project was supported by GACR 19-15737S, MZCR-RVO 65269705, MUNI/A/1330/2021 and
AZV NU21-08-00237.
P609: CLINICOBIOLOGICAL CHARACTERISTICS AND TREATMENT EFFICACY OF NOVEL AGENTS IN
CHRONIC LYMPHOCYTIC LEUKEMIA WITH IGLV3-21R110
P. Hengeveld1,2,*, Y. E. Ertem1, J. Dubois3, C. Mellink4, A.-M. van der Kevie-Kersemaekers4,
L. Evers3, K. Heezen1, P. M. Kolijn1, O. Mook4, M. M. Motazacker4, K. Nasserinejad5,
S. Kersting6, P. Westerweel2, C. Niemann7, A. Kater3, A. Langerak1, M.-D. Levin2
1Department of Immunology, Erasmus MC, Rotterdam; 2Department of Internal Medicine,
Albert Schweizer Ziekenhuis, Dordrecht; 3Department of Hematology; 4Department of
Human Genetics, Amsterdam UMC, Amsterdam; 5Department of Hematology, Erasmus MC, Rotterdam;
6Department of Hematology, Haga Ziekenhuis, Den Haag, Netherlands; 7Department of
Hematology, Rigshospitalet, Copenhagen, Denmark
Background: The composition of the clonotypic B cell receptor (BCR) is of key importance
in chronic lymphocytic leukemia (CLL). Recently, a novel immunogenetically defined
CLL subset was described, characterized by a clonotypic BCR using the immunoglobulin
lambda (IGL) IGLV3-21*01/IGLV3-21*04 gene with a distinctive G110R somatic hypermutation
(IGLV3-21R110). IGLV3-21R110 CLL accounts for approximately 20% of all CLL and is
associated with an adverse prognosis. However, the clinicobiological profile and predictive
impact of IGLV3-21R110 CLL in the context of novel therapies remains incompletely
characterized.
Aims: To characterize the cytogenetic, immunogenetic and mutational landscape of IGLV3-21R110
CLL and to assess the predictive impact of this genotype in the context of novel therapies.
Methods: We characterized the light chain genotype, clinicobiological features and
response to therapy of patients enrolled in the HOVON-139/GIVE trial and the Dutch
sub-cohort of the HOVON-141/VIsion trial. The HOVON-139/GIVE phase-II trial evaluated
first-line minimal residual disease (MRD)-guided duration of treatment with obinutuzumab
and venetoclax in CLL patients unfit for treatment with chemoimmunotherapy. The HOVON-141/VIsion
phase-II trial evaluated MRD-guided ibrutinib and venetoclax combination treatment
in relapsed or refractory (R/R) CLL patients.
Results: The IGLV3-21R110 genotype was present in 16/65 patients (25%) in the first-line
cohort and in 32/129 patients (25%) in the R/R cohort. Loss of 13q14 and 11q22 were
enriched in IGLV3-21R110 patients, compared to other patients (del13q14: 79% vs. 57%,
P=0.009; del11q22: 34% vs. 18%, P=0.03). In contrast, the IGLV3-21R110 genotype and
trisomy 12 or loss of 17p13 were mutually exclusive (trisomy 12: 0% vs. 12%, P=0.008;
del17p13: 0% vs. 12%, P=0.01). Mutations in the SF3B1 and ATM genes were significantly
more common in IGLV3-21R110 patients, compared to other patients (SF3B1: 49% vs. 21%,
P=0.0008; ATM: 36% vs. 18%, P=0.02). IGHV and IGHD gene usage of IGLV3-21R110 patients
was markedly skewed. The IG heavy-chain complementarity determining region 3 (HCDR3)
was shorter in IGLV3-21R110 patients, compared to other patients (median HCDR3 aa
length 13 [range: 9-17] vs. 19 [range 8-30], P<0.0001). Whereas the IGHV SHM imprint
of IGLV3-21R110 patients was centered around the 98% cutoff between unmutated and
mutated IGHV, the range was wider in other patients (range 95.1%-99.7% vs. 85%-100%,
P=0.01). When comparing patients with IGLV3-21R110 CLL versus all other light chains,
were no differences regarding MRD (%uMRD<10-4 in peripheral blood, HOVON-139/GIVE
after 12 cycles: 87% vs. 98%, P=0.15; HOVON-141/VIsion after 15 cycles: 52% vs. 61%,
P=0.11) and progression-free survival (24-month PFS, HOVON-139/GIVE: 100% vs. 94%,
P=0.52; HOVON-141/VIsion: 91% vs. 92%, P=0.74) in either trial.
Summary/Conclusion: We have characterized the clinicobiological features of the largest
cohort of IGLV3-21R110 patients reported thus far. We demonstrate that CLL with IGLV3-21R110
is typified by a distinct cytogenetic, mutational and immunogenetic profile. There
was no evidence for a predictive impact of the IGLV3-21R110 genotype on the efficacy
of the novel therapies with venetoclax and ibrutinib employed in the HOVON-139/GIVE
and HOVON-141/VIsion trials. Our results suggest that novel targeted therapies may
mitigate the adverse risk profile of IGLV3-21R110 CLL. To determine whether these
patients should preferentially receive novel therapies, characterization of the predictive
impact of IGLV3-21R110 in the setting of chemoimmunotherapy is warranted.
P610: SYNERGISTIC ACTIVITY OF FTO INHIBITOR FB23-2 WITH IBRUTINIB IN XENOGRAFT MURINE
MODEL OF CHRONIC LYMPHOCYTIC LEUKEMIA
X. Hu1,*, H. Wang2, Y. Han1, X. Zhang1, Z. Tian1, Y. Zhang2, X. Wang1
1Department of Hematology, Shandong Provincial Hospital,Cheeloo College of Medicine,
Shandong University; 2Department of Hematology, Shandong Provincial Hospital Affiliated
to Shandong First Medical University, Jinan, China
Background: Several studies have documented that FB23-2, as the selective inhibitor
of RNA N6-methyladenosine (m6A) demethylase FTO, exerts crucial role in dampening
tumorigenesis. Indeed, FTO deactivation was proved to render resistant leukemia cells
sensitive to targeted agents, which suggests a potential combination therapy strategy
of FB23-2.
Aims: The present study was aimed to identify the synergistic utility of FB23-2 combination
with ibrutinib in CLL.
Methods: A disseminated leukemia xenograft murine model was established to explore
the efficacy of FB23-2 in CLL. Leukemia burden of CLL mice was quantified by luciferase
intensity using bioluminescence imaging. Bone marrow and spleen infiltration of leukemic
cells was assessed by flow cytometry. And cell survival was determined by cell counting
kit-8.
Results: To investigate the combinatory effect of FB23-2 and ibrutinib, cell viability
assay was performed firstly. FB23-2 displayed potent synergistic activity with ibrutinib
in promoting cytotoxicity in CLL cells (Figure 1A). Consistently, FTO silencing mediated
by lentivirus exhibited exquisite sensitivity to ibrutinib in CLL cells (Figure 1B).
Besides, CLL primary cells and MEC-1 cells with treatment of FB23-2 and ibrutinib
simultaneously triggered increased cell apoptosis compared to single agent (Figure
1C). Further underlying cooperative mechanism investigation demonstrated an aberrant
activation of DNA damage pathway after serial dilution of FB23-2 treatment. Hence,
enhanced expression of p-ATM, p-chk2 and p-H2AX was detected in FTO knockdown and
ibrutinib co-treatment group, indicating FTO inhibition inducing ibrutinib-sensitivity
via regulating DNA damage (Figure 1D).
In addition, we investigated the efficacy of FB23-2 alone and corporation in CLL xenograft
murine model. Compared to ibrutinib alone, combination treatment with FB23-2 significantly
prolonged the lifespan of CLL mice (median survival time 20 versus 22.5 days, respectively,
p=0.02, Figure 1E). FB23-2+ibrutinib group showed an obviously declined proportion
of CLL cells compared to ibrutinib-treated alone both in bone marrow(20.93%±2.95%
versus 37.17%±4.22%, p=0.02) and spleen (23.58%±6.03% and 54.58%±9.01%, p=0.03, Figure
1F-G). Similar results were obtained with three treated group relative to control
group (all p<0.05). Moreover, FB23-2 combined with ibrutinib diminished leukemia burden
and splenomegaly in CLL mice (Figure 1H-I). As shown in Figure 1J, leukemia involvement
and malignant proliferation were more evident in control group, confirming the potent
anti-leukemic activity of FB23-2.
Image:
Summary/Conclusion: Taken together, our investigation provides pre-clinical evidence
for the utility of FB23-2 in CLL, especially combinatory benefit with ibrutinib. FB23-2
represents a promising strategy to novel treatment optimum in CLL.
P611: NEXT-CLL: A NEW NGS-BASED METHOD FOR RAPID ASSESSMENT OF IGHV MUTATIONAL STATUS
IN CHRONIC LYMPHOID LEUKEMIA
E. Bourbon1, K. Chabane2, A.-S. Michallet3, E. Ferrant1, I. Mosnier2, S. Poulain4,
M. Giraud5, A. Bouvard2, H. Ghesquieres6, S. Hayette2, P. Sujobert2, S. Huet2,*
1Hematology clinics; 2Laboratory of hematology, Hospices Civils de Lyon, pierre-benite;
3Hematology clinics, Centre Leon Berard, Lyon; 4Laboratory of hematology; 5Bio-informatics,
CHRU de Lille, Lille; 6Hematology clinics, Hospices Civils de Lyon, Lyon, France
Background: Chronic Lymphocytic Leukemia (CLL) is a highly heterogeneous disease both
in terms of biological landscape and clinical course. Current guidelines recommend
IGHV mutation status (MS) determination prior to treatment initiation in order to
guide the first-line therapeutic choice between chemoimmunotherapy and novel agents.
Currently, the IGHV MS is determined in most routine laboratories by low-throughput
Sanger sequencing that allows for the detection of the dominant clone in the majority
of CLL cases. However, this technic remains technically challenging despite best practice
guidelines, does not provide any insight into subclonal architecture and intraclonal
diversity, and has a general failure rate ranging from 10 to 20%. Besides, commercially
available NGS solutions have drawbacks (costs) and constraints (as they necessitate
mandatorily a MiSeq sequencing machine and are not feasible on other devices) that
restrict their use by any laboratory in routine practice.
Aims: In this study, we aimed at providing a ready-to-use and rapid strategy to evaluate
the IGHV gene MS using NGS in routine practice. We present a new method called Next-CLL,
allowing rapid assessment on any NGS device available in routine diagnostic laboratories.
Methods: Peripheral blood samples were obtained from 80 patients with typical CLL
at diagnosis. Genomic DNA (gDNA) and complementary DNA (cDNA) were used. In order
to fully cover the IGHV rearranged gene sequence, 4 mutliplex PCR reactions were designed:
[LEADER-FR2R/JHc], [FR1c-JHc], [FR1-JHc] and [FR2-JHc], producing overlapping amplicons.
Final products from the 4 PCR reactions were mixed and purified, subjected to A-tailing
and ligated to Illumina-single indexed adapters. The library was then sequenced (2x150 bp)
on a NextSeq 500 (Illumina). A reassembly of IGHV overlapping amplicons was performed
using the BBTools Suite. The outputs were then uploaded on the Vidjil plateform for
IGHV clonal analysis. The MS was calculate using the IMGT web interactive tool following
ERIC recommendations. IgHV MS was assessed in parallel by Sanger sequencing or the
commercial NGS assay IdentiClone® Assay (Invivoscribe) as reference. Gene usage, %
identity to the germline and stereotype subsets were compared with the results obtained
from Next-CLL.
Results: Next-CLL identified a productive clone in 100% of cases (n=80), compared
to PCR with Sanger sequencing (15% of failure). All cases that failed using the Sanger
sequencing were subjected to a second NGS technique (Invivoscribe) that confirmed
the results from Next-CLL, ruling out any possibility of false positive cases. Next-CLL
showed 100% concordance with the reference techniques for IGHV gene identification
and allowed assessment of the MS from the leader sequence, as recommended by the ERIC
consortium. Only 2 cases showed discordant IGHV MS, close to the 98% identity cut-off,
between Next-CLL and Sanger sequencing (the latter being unable to capture the leader
sequences), thus providing 97% of concordance between the 2 techniques (Figure 1).
Moreover, 100% concordant results were obtained between gDNA and cDNA (n=32 cases
analysed for both) using Next-CLL.
Image:
Summary/Conclusion: Our new method Next-CLL allows assessment of the IGHV MS by NGS
with an easy and rapid workflow, providing clinically useful information from the
IGHV leader sequence and allowing analysis of intra-clonal diversity (not possible
with the standard Sanger sequencing). Importantly, it might be used both on gDNA and
cDNA and on any sequencing machine, on the contrary to existing commercially available
kits.
P612: CHRONIC LYMPHOCYTIC LEUKEMIA (CLL)-DERIVED T-CELLS HAVE DISTURBED FATTY ACID
METABOLISM, POSSIBLY CONTRIBUTING TO T-CELL DYSFUNCTION
C. F. Jacobs1,2,3,4,*, H. Simon-Molas1,2,3,4, N. Zelcer5, A. P. Kater2,3,4,6, F. S.
Peters1,2,3,4
1Experimental Immuno-Hematology; 2Hematology, Amsterdam University Medical Center;
3Cancer Center Amsterdam; 4Amsterdam institute for Infection & Immunity; 5Medical
Biochemistry, Amsterdam University Medical Center; 6Lymphoma and Myeloma Center Amsterdam,
Amsterdam, Netherlands
Background: T-cells from CLL patients are dysfunctional and display reduced activation,
proliferation and cytotoxicity upon in-vitro activation. Whilst the role of glucose
metabolism in activated T cells is extensively studied, the contribution of other
energy sources is less clear. Fatty acid (FA) and lipid uptake in T cells involves
the scavenger membrane transporter CD36. CD36-mediated lipid transport has been identified
to (metabolically) modulate intratumoral T cells (both Treg and cytotoxic CD8+ T cells).
However, lipid metabolism and a potential role for CD36 has not been studied in the
context of T cell dysfunction in CLL
Aims: To examine the role of FA metabolism and the fatty acid transporter CD36 in
acquired T-cell dysfunction in CLL.
Methods: Untreated CLL patients with a white blood cell count (WBC) greater than 20×109
cells/L and, as control, age-matched healthy donors (HD) were studied. T cells were
analyzed by flow cytometry, confocal microscopy or extracellular flux analysis (Seahorse),
either directly after thawing or after a 2 day culture with or without αCD3/αCD28
antibodies.
Results: Analysis of CD36+ vs CD36- CD4 and CD8 T-cells from HD revealed that CD36+
cells had higher long-chain fatty acid (LCFA) and glucose uptake, and higher expression
of the glucose transporter GLUT-1 and the rate-limiting mitochondrial FA transporter
CPT1α (Fig. 1A). The cells expressing CD36 were also more likely to increase their
CD25 expression and mitochondrial mass upon stimulation, indicating a metabolic and
functional advantage potentially conferred by the surface expression of CD36. The
absolute amount and percentage of T cells (CD4 and CD8) expressing FA transporter
CD36 was lower in CLL T-cells as compared to HD (Fig. 1B). The levels of LCFA uptake
and expression of CPT1α were comparable, indicating that the lower amount of CD36
might successfully cater to the FA demands of resting CLL T cells(Fig. 1C). However,
while stimulation of HD T-cells markedly upregulated CD36 abundance, LCFA uptake and
CPT1α, concomitantly with the activation marker CD25, CLL-derived T-cells failed to
mount a similar response (Fig. 1D). This lack of induction of CD36 expression was
also observed in CLL-derived T cells that increased CD25, which implies that this
response is independent of T-cell activation. The transcription factors peroxisome
proliferator-activated receptors (PPAR) α and γ are important regulators of FA metabolism.
We observed reduced expression of these transcription factors in resting CLL T cells.
However, distinct from CD36, CLL T cells were indistinguishable from HD T-cells in
their ability to increase levels of both PPARα and PPARγ upon stimulation, indicating
that alternative mechanisms might be implicated in the regulation of CD36 and FA metabolism
in CLL T cells.
Image:
Summary/Conclusion: Collectively, our results show that disturbances in FA metabolism
are present on multiple levels in CLL T cells and that lack of LCFA uptake into CLL
T-cells could represent a limiting step in FA metabolism. We also highlight CD36+
cells as a metabolically distinct T-cell subtype that is largely absent in CLL T-cells,
independently of their activation status. We are currently investigating strategies
to increase CD36 expression and FA metabolism in general in CLL T cells, which could
ameliorate CLL T cell dysfunction and provide targets to improve autologous T cell
therapies.
P613: TLR9 SIGNALLING IS A POTENTIAL TUMOUR ESCAPE MECHANISM FOLLOWING BTKI THERAPY
FOR THE TREATMENT OF CHRONIC LYMPHOCYTIC LEUKAEMIA.
E. Kennedy1,*, S. Mitchell1, T. A. Burley1, T. Liloglou2, R. Johnston3, I. Ashworth1,
D. Bak-Blaz3, K. Chamberlain3, E. Ladikou1, S. Mackay4, C. Pepper1, A. G. Pepper1
1Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School,
Falmer; 2Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and
Integrative Biology, University of Liverpool, Liverpool; 3Brighton and Sussex University
Hospitals NHS Trust, Brighton; 4Cell Biology and Drug Discovery and Design Groups,
Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde,
Glasgow, United Kingdom
Background: CLL cell trafficking is fundamental to CLL progression and an important
focus when identifying novel therapeutic targets. Toll-like receptor 9 (TLR9) is an
intracellular pattern recognition receptor, and a potential contributor to CLL cell
chemotaxis and tumour maintenance. TLR9 recognises unmethylated CpG motifs within
bacterial/viral/mitochondrial DNA, and TLR9 activation promotes an NFkB and STAT3-driven
activation/migratory phenotype in primary CLL cells. We have previously shown cell-free
unmethylated DNA to be up to 28-fold higher in CLL patient plasma, relative to healthy
controls.
Aims: To investigate TLR9 activation as a potential mechanism of resistance to current
B-cell receptor (BCR)-targeted therapeutics.
Methods: Migration assays were performed using primary CLL cells stimulated -/+ 1μM
ODN 2006 (TLR9 agonist), -/+ 1μM ibrutinib (BTK-inhibitor), 2μM ODN INH-18 (TLR9 antagonist)
or -/+ 2.5-10μM CW15337 (NIK inhibitor). CLL cells migrated through 5μM pore membranes
towards a CXCL12-gradient.
Results: TLR9 activation induced a dichotomous migratory response in CLL patient samples.
Following stimulation with ODN 2006, 25/42 (60%) patient samples showed an
increase
in CLL cell migration (‘Responders’) and 17/42 (40%) showed either
no change
or a
decrease
in CLL cell migration (‘Non/Reverse Responders). Interestingly, IGHV-mutated (M-CLL)
samples were almost exclusively ‘Responders’ (i.e., 14/17 [82%]) and IGHV-unmutated
(U-CLL) samples were equally likely to be ‘Responders’ or ‘Non/Reverse Responders’
(i.e., 11/25 [44%] vs 14/25 [56%] respectively).
Whilst there was no difference in the expression levels of TLR9 in M-CLL vs U-CLL
cells, U-CLL samples expressed significantly higher basal levels of CD69 (B-cell activation
marker), and their TLR9-induced migratory response negatively correlated with their
basal migration. We therefore hypothesised that U-CLL ‘Non/Reverse Responders’ may
have reached maximal stimulatory capacity through BCR-signalling alone, rendering
them unresponsive to further activation.
To test this hypothesis, we simulated BCR and TLR9 stimulation in M-CLL and U-CLL
cells using mathematical modelling. Our simulations showed M-CLL cells (with low basal
BCR-activation) to induce NFkB signalling in response to TLR9 activation and U-CLL
cells (with high basal BCR-activation) to be unresponsive. Importantly, when simulating
treatment with a Bruton’s Tyrosine Kinase inhibitor (BTKi), U-CLL cells gained sensitivity
to TLR9 activation. These results have since been verified in vitro using BTKi-treated
patient samples, where we found a subset of U-CLL ‘Non/Reverse Responders’ to become
‘Sensitised’ to TLR9 activation in the presence of ibrutinib. In the ‘Responder’ subgroup,
ODN INH-18 (TLR9 antagonist) and ibrutinib inhibited CLL cell migration synergistically.
Together, these data implicate TLR9 signalling as a tumour escape mechanism following
BTKi therapy. Since both the BCR and TLR9 signalling pathways culminate in NFkB activation,
we are currently investigating components of NFκB signalling as potential novel therapeutic
targets. Preliminary data using the NIK selective inhibitor ‘CW15337’ shows CLL cell
migration to be inhibited with an IC50 of <1μM.
Summary/Conclusion: CLL patient plasma contains high levels of TLR9 ligand (unmethylated
DNA), which may promote CLL cell trafficking and BTKi resistance in select subgroups.
We believe that targeting downstream NFκB signalling has the potential to inhibit
both BCR and TLR9 signalling, and to increase treatment efficacy in TLR9 ‘Responder’
and BTKi ‘Sensitised’ patients.
P614: SUBCLONAL ARCHITECTURE OF CHROMOSOMES REVEALED BY SINGLE-CELL ANALYSIS OF GENE
EXPRESSION IN A PATIENT WITH CLONAL EVOLUTION OF RELAPSING/REFRACTORY CLL
J. Kotaskova1,2,3,*, T. Kurucova2, K. Reblova1,2,3, M. Valisova1,2, K. Zavacka1,2,
A. Mareckova1, M. Bohunova1, S. Pavlova1,2, J. Malcikova1,2, V. Navrkalova1,2, M.
Jarosova1,3, M. Doubek1,2,3, K. Plevova1,2,3, S. Pospisilova1,2,3
1Department of Internal Medicine – Hematology and Oncology, University Hospital Brno
and Faculty of Medicine, Masaryk University; 2Central European Institute of Technology,
Masaryk University; 3Institute of Medical Genetics and Genomics, University Hospital
Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
Background: Chronic lymphocytic leukemia (CLL) manifests by remarkable intraclonal
heterogeneity of genomic defects, especially in patients with relapsed/refractory
disease, often connected with complex karyotype. In cancer cells, chromosomal changes
are often multiple and complex. Aberrations can be present only in subclones and often
underlie disease refractoriness. Using the bulk analysis such as WGS, WES, or genomic
array to precisely determine the co-occurrence of aberrations in the individual cell
provides limited information.
Aims: To perform an in-depth, single-cell RNA sequencing (scRNAseq) study of subclonal
changes associated with relapsed/refractory CLL.
Methods: A bulk analysis of DNA from separated malignant cells using comprehensive
NGS panel LYNX (PMID 34082072) was performed on a representative CLL case at three
time-points (TPs): at dg (TP1, month 0, Rai II), before first therapy (TP2, month
19) and during relapse (TP3, month 59). Simultaneously, the transcriptome was analyzed
using scRNAseq (Chromium system, 10x Genomics) in 2330 cells (TP1), 2397 cells (TP2),
and 1902 cells (TP3). A detailed data analysis was carried out with the Seurat R package.
In addition, we used the InferCNV tool to detect the chromosomal aberrations in every
tested cell. We identified multiple chromosomal changes and compared the findings
with the results of bulk DNA analysis using a genomic array (ThermoFisher Scientific)
and FISH.
Results: We detected a number of aberrations that were already present at TP1, including
del13q, del11q, mutations in NOTCH1, RPS15, ZMYM3, and minor mutations in TP53 and
ATM. At TP2, additional changes comprised mutations in PIM1, XPO1, and del(3q). Between
TP2 and TP3, the patient underwent several treatment lines (FCR, BR, COP) without
an apparent therapeutic effect. The cells from TP3 carried complex changes, including
additional mutations in BIRC3 and RB1. Unsupervised clustering of scRNAseq gene expression
data defined major clones bearing aberrations (losses on chromosomes 1, 9, 13, 17,
18). Moreover, these clones were composed of cells with additional aberrations defining
a number of separate subclones. The proportion of subclones was typically under the
detection limit of the genomic array and showed extreme intraclonal heterogeneity
of the relapsed/refractory CLL case.
Summary/Conclusion: We showcase how genomic alterations influence the expression of
affected genes. Such fact can be exploited to reconstruct chromosomal disruptions
in individual cells using scRNAseq and to uncover the subclonal architecture of the
disease.
Grants: MH-CZ_AZV_NU20-08-00314, MH-CZ_AZV_NV19-03-00091, MEYS-CZ_MUNI/A/1330/2021,
MH-CZ_RVO_65269705.
P615: BCL2 RESISTANCE MUTATIONS IN A REAL-WORLD COHORT OF PATIENTS WITH VENETOCLAX-TREATED
CHRONIC LYMPHOCYTIC LEUKAEMIA
L. Kotmayer1,*, T. László1, R. Kiss1, L. L. Hegyi1, G. Mikala2, P. Farkas3, A. Balogh3,
T. Masszi3, J. Demeter4, J. Weisinger3, H. Alizadeh5, L. Gergely6, A. Sulák7, M. Egyed8,
M. Plander9, P. Pettendi10, D. Lévai11, T. Schneider11, Z. Pauker12, A. Masszi11,
R. Szász6, C. Bödör1, D. Alpár1
1Department of Pathology and Experimental Cancer Research, Semmelweis University;
2South-Pest Central Hospital – National Institute of Hematology and Infectious Diseases;
3Department of Internal Medicine and Hematology; 4Department of Internal Medicine
and Oncology, Semmelweis University, Budapest; 51st Department of Internal Medicine,
Clinical Centre, University of Pécs, Pécs; 6Division of Hematology, Department of
Internal Medicine, University of Debrecen, Debrecen; 72nd Department of Internal Medicine
and Cardiology Center, University of Szeged, Szeged; 8Kaposi Mór University Teaching
Hospital of County Somogy, Kaposvár; 9Markusovszky University Teaching Hospital, Szombathely;
10Hetényi Géza Hospital and Clinic of County Jász-Nagykun-Szolnok, Szolnok; 11National
Institute of Oncology, Budapest; 12Borsod-Abaúj-Zemplén County Hospital and University
Teaching Hospital, Miskolc, Hungary
Background: The Bcl-2 inhibitor venetoclax has transformed the therapeutic landscape
of therapy refractory and treatment naïve chronic lymphocytic leukaemia (CLL). Despite
the remarkable response rates in both patient groups, a subset of CLL patients receiving
venetoclax treatment experience disease progression, frequently associated with pathogenic
variants of the BCL2 gene. Apart from the most common hotspot G101V and D103Y mutations,
pathogenic variants affecting several different loci of the coding region of BCL2
have been described, however data and international recommendations on the sensitive
detection and monitoring of these mutations are still absent to date.
Aims: Our aim was to develop a droplet digital PCR (ddPCR) based assay for the detection
of BCL2 G101V and D103Y mutations and to reveal further resistance mutations emerging
at relapse by performing targeted ultra-deep next-generation sequencing (NGS).
Methods: Peripheral blood samples from 71 patients treated with venetoclax-rituximab
or first line venetoclax-obinutuzumab combination were collected from 11 Hungarian
oncohematological centres. Genomic DNA was extracted from peripheral blood mononuclear
cells and leukemic cell purity was assessed by flow cytometry. Fractional abundance
of the most common hotspot BCL2 mutations G101V and D103Y was assessed using the QX200
digital droplet PCR system. Following ddPCR analyses, targeted ultradeep NGS was performed
on samples obtained at relapse using a custom-designed panel covering the whole coding
regions of BCL2, BTK, PLCG2 and TP53 genes. Fractional abundance of BCL2 mutations
identified by NGS was assessed retrospectively in serial peripheral blood samples
by ddPCR using custom-designed, allele-specific assays.
Results: Lower quantitative limit of detection of the BCL2 ddPCR assays was tested
and could ubiquitously be established as 0.01%. With a median follow up of 14 months,
BCL2 G101V and D103Y mutations were detected in 8.5% (6/71) and 9.9% (7/71) of the
patients, respectively, with 90% (9/10) of them experiencing relapse during the follow-up
period. Three patients harbored both resistance mutations as detected by ddPCR. BCL2
G101V and/or D103Y were observed in 43.5% (10/23) of the cases showing signs of disease
progression. In four cases, emergence of the mutations predated the first clinical
signs of relapse with a median 4 months. In samples obtained at relapse, targeted
ultradeep NGS uncovered two additional BCL2variants (V156D and A113G), which were
successfully backtracked and detected by ddPCR. Both V156D and A113G were detected
in patients previously found to harbor G101V or D103Y, with no further BCL2 resistance
mutations identified in G101V and D103Y wild type cases.
Summary/Conclusion: In a real-world cohort of CLL patients receiving venetoclax in
combination therapy, comprehensive and sensitive screening of BCL2 mutations can identify
molecular mechanisms of venetoclax resistance in nearly half of the patients experiencing
relapse. In patients harboring multiple BCL2 mutations, convergent evolution of the
CLL subclones may contribute to the driver mechanisms of resistance, justifying the
comprehensive approach for the detection of these variants. In secondary venetoclax
resistant cases displaying wild type BCL2, further molecular screening methods are
required to reveal alternative genetic or non-genetic reasons for disease progression.
P616: THE LOSS OF IΚBΕ INHIBITOR ACCELERATES DISEASE DEVELOPMENT IN CHRONIC LYMPHOCYTIC
LEUKEMIA
C. Lenzi1,*, J. Bordini2, A. Pseftogkas1, A. Morabito2, G. Tsiolas3, F. E. Psomopoulos3,
A. Campanella1, M. Frenquelli2, P. Ghia1
1Division of Experimental Oncology, Università Vita-Salute San Raffaele; 2Division
of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy; 3Center
for Research and Technology Hellas-CERTH, Thessaloniki, Greece
Background: Chronic lymphocytic leukemia (CLL) is an indolent B cell malignancy in
which B lymphocytes accumulate in peripheral blood (PB), bone marrow (BM) and spleen
(SP), as a result of a complex intertwining between microenvironmental stimuli and
genetic defects. Molecular studies revealed recurrent mutations in a large number
of genes, each present with distinct frequency in different subgroups of patients.
Among these genes, NFKBIE, a negative regulator of the NF-κB pathway, has been found
mutated in a relevant proportion of patients with CLL, mainly those belonging to the
clinically aggressive subset #1, suggesting that a reduced expression of NFKBIE may
contribute to CLL aggressiveness through constitutive NF-κB activation independent
of BcR signaling.
Aims: We aimed at studying the effects of NFKBIE in the pathogenesis of CLL using
functional inactivation of the gene both in vitro and in vivo.
Methods: We engineered the MEC-1 CLL cell line using CRISPR/Cas9 technology to generate
NFKBIE- knock-out (KO) cell lines. We analyzed the effect of NFKBIE ablation studying
in vitro cell proliferation and migration, gene expression, BcR signalling pathway,
and response to treatment with the BTK inhibitor Ibrutinib. We also evaluated the
relevance of NFKBIE manipulation in vivo by injecting the engineered cells into RAG2-/-
γc-/- immunodeficient mice. In addition, we generated NFKBIE-/wt mice and crossed
them with Em-TCL1 mice, a validated CLL mouse model, to generate immunocompetent Em-TCL1-NFKBIE-/wt.
We analyzed disease expansion by flow cytometry, measuring the amount of malignant
B cells in the PB, SP and BM.
Results: MEC-1 cells carrying NFKBIE ablation showed faster proliferation in vitro
as compared to controls (p<0.05). RNAseq analyses indicated de-regulation of migratory
pathway that was functionally confirmed by migration experiments with the specific
chemokines (i.e. CCL19, CCL21 and SDF1). In particular, the percentage of migrated
KO cells was significantly higher than that of control cells, either in the presence
or the absence of each of the three chemokines. As predicted by the function of the
gene, biochemical analyses in KO cells revealed the increase of cRel and p50 protein
levels and the increase of p65 phosphorylation, all NF-κB transcription factors indicating
unabated cellular activation. Interestingly, in vitro culture of the cells in the
presence of Ibrutinib showed that NFKBIE KO cells are more resistant to apoptosis
induction than control cells (p<0.05). The effect of NFKBIE manipulation was also
more evident in vivo. Xenografts generated with MEC-1 NFKBIE KO cells in RAG2-/- γc-/-
immunodeficient mice showed an increased disease expansion in the SP and BM (p<0.05)
and a shorter survival (p<0.05) than mice injected with control cells. In immunocompetent
Em-TCL1 mice, which develop the CLL-like disease with aging, preliminary analysis
at 3, 6 and 9 months of age showed higher malignant cells expansion in PB of Em-TCL1-NFKBIE-/wt
compared with control Em-TCL1 mice.
Summary/Conclusion: The loss of NFKBIE function appears to be associated with a more
aggressive disease in all CLL models tested, both in vitro and in vivo, accelerating
disease development under CLL pro-tumorigenic conditions. In addition, it appears
to be potentially involved in the onset of resistance to the BTK inhibitor Ibrutinib,
thus providing an alternative mechanism that may explain drug failure in the resistant
patients who do not carry BTK/PLCG2 mutations.
P617: FACING LIMITED DATASET IN HEMATOLOGICAL MALIGNANCIES: OPTIMIZATION OF MACHINE
LEARNING MODELS WITH THE GENERATION OF NEW TRAINING VARIABLES AND HYPER-PARAMETERS
SELECTION.
R. BIZOÏ1,*, L. Mauvieux2,3
1DBSYS; 2Laboratoire d’hematologie, Hopital Hautepierre; 3IRFAC, INSERM U1113, Strasbourg,
France
Background: There is a growing interest in the application of Machine-Learning (ML)
techniques in medical research. However, the datasets in this field concern a limited
number of individuals. Our goal was to implement technologies and measures for estimating
learning performance in a small dataset of B-cell chronic lymphoid malignancies with
extreme individuals (outliers) and a modality imbalance for the target variable.
Aims: We postulated that a limited set of antigen expression could decipher the different
diagnosis of chronic B-cell malignancies (CBCM). In this aim we retrospectively studied
the flow cytometry results obtained at the diagnosis for 673 patients explored in
our center: 212 CLL/lymphocytic lymphoma, 160 lymphoplasmacytic lymphoma (LPL, including
Waldenstrom Macroglobulinemia), 76 mantle cell lymphoma (MCL), 169 marginal zone lymphoma
(MZL), 35 Hairy cell lymphoma (HCL) and 27 follicular lymphoma. We focused on median
fluorescence intensity (MFI) values of CD148, CD180 and CD200 and three categorical
“positive or negative” markers (CD5, CD23, FMC7).
Methods: We used the Scikit-Learn python library and LightGBM to test several ML models
and develop techniques for generating new variables and selecting hyper-parameters.
The treatments on the dataset were the following:
1. The three quantitative dimensions were normalized using Box-Cox transformation
for LightGBM, LogisticRegression, RandomForest, KNN and GaussianNaiveBayes algorithms.
2. 70% of the data for training and 30% for testing were randomly separated.
3. Outlier removal and diagnostic dimension, modality balancing treatments were performed
only on the learning sample using different unsupervised methods: localOutlierFactor,
IsolationForest, OneClassSVM and EllipticEnvelope.
4. In order to avoid overfitting, the number of individuals of the least frequent
modalities were balanced using (Synthetic Minority Over-sampling Technique) SMOTE
algorithm.
5. For a better accuracy of classification algorithms, a discretization was set on
the quantitative dimensions (CD148, CD180 and CD200 MFI). Thus all the values of the
three dimensions were merged into one and this dimension was discretized into 96 quantiles.
6. New variables were created, that allowed a better learning for the LightGBM and
RandomForest classification algorithms. Polynomial transformation and a principal
component analysis were used for Logistic Regression.
7. Algorithms hyperparameters of ML were optimized in order to improve performance
using GridSearchCV. The selection of the best ML models was done using cross-validation.
Results: The models were used for the prediction of CLL, LPL, MCL and MZL diagnosis.
The results for each of the classifiers are summarized in Table 1. The global accuracy
of the prediction using only 6 antigenic markers was very stable, ranging from 80
to 85% for the different algorithms. The F1-score, that represents the harmonic mean
of the precision and recall of prediction (TP/(TP+ ½* (FP + FN)) was the best for
CLL (f1 = 91,4%) and the lowest values were obtained for MCL and LPL.
Image:
Summary/Conclusion: In conclusion, the methodology presented here allows an optimization
of machine learning algorithms for limited set of data. Using only the results of
six flow cytometry stainings, that can be analyzed in a single tube, high diagnosis
prediction rates were obtained. These results, that need to be confirmed in different
series, open the way to innovating diagnosis assistance tools in the field of hematological
malignancies. These methods can also be applied to other diagnosis issues.
P618: SECOND-GENERATION CD19 TARGETING TRI-SPECIFIC KILLER ENGAGER DRIVES ROBUST NK
CELL FUNCTION AGAINST B CELL MALIGNANCIES
J. Miller1,*, B. Kodal1, P. Hinderlie1, D. Vallera2, G. Berk3, V. Bachanova1, M. Felices1
1Medicine; 2Radiation Oncology, University of Minnesota, Minneapolis; 3GT Biopharma,
Brisbane, United States of America
Background: Chronic Lymphocytic Leukemia (CLL) involves uncontrollable clonal expansion
of CD5+/CD19+ B lymphocytes. While patients can sometimes coexist with CLL for years,
eventually there is progression to bulky adenopathy and pancytopenia from marrow suppression
requiring therapy. In recent years, targeted therapies, including utilization of the
inhibitors for B cell receptor (BCR) signaling and for B cell lymphoma 2 (Bcl-2) protein,
have been effective treatment options. However, these treatments are associated with
limitations such as development of drug resistance and the lower treatment efficacy
ratio in high-risk patients. Meanwhile, CAR-T cell immunotherapy failed to produce
effective treatment outcomes against CLL, mostly due to defects in the effector T
cells leading to product failures, as well as being associated with high levels of
off-target cytotoxicity.
Aims: Thus, approaches to CLL treatment are necessary and NK cell immunotherapy presents
a viable and non-toxic option to this problem.
Methods: Our group previously demonstrated the immunotherapeutic potential of utilizing
first-generation CD19-targeting 161519 Tri-Specific Killer Engager (TriKE®) to drive
natural killer (NK) cell cytotoxicity against B-cell malignancies. We have now produced
a novel second-generation CD19 TriKE, ‘CAM161519’, utilizing a humanized camelid anti-CD16
VHH single domain antibody (sdAb) ‘CAM16’, a wild-type IL-15 component, and anti-CD19
tumor antigen scFv all linked via short peptide linkers. The TriKE primarily works
by generating a cytolytic bridge between NK cells and CD19 expressing B cell malignancies
while also providing an expansion and survival signal to the NK cells. Using flow-based
assays and imaging based cytolytic assays we explored the capability of CAM161519
to target a variety of B cell lines (Raji, Daudi, and Nalm6) and primary B cell malignancies
from patients with CLL and B cell acute lymphoblastic leukemia (B-ALL). Expanded NK
cells were incubated with primary CLL targets from 6 different patients and no treatment
(NT), rhIL-15 (30 nM), Rituximab (10 ug/mL), or CAM16159 (30 nM). NK cell degranulation
and IFNg production was evaluated after 5 hours of co-culture (Figure). CLL killing
was evaluated in a similar co-culture, but with a staining mix containing CD5 and
CD19, to identify CLL tumor cells in order to determine the percent CLL targets killed
normalized to incubation of CLL cells with NK cells alone (Figure Image).
Results: The CAM161519 robustly activated NK cell degranulation (CD107a) and inflammatory
cytokine production (IFNg) against B cell lines. The activation was specific as it
was strongly reduced against CD19 knockout Nalm6 cells. The TriKE also induced specific
NK cell proliferation. IncuCyte imaging based cytolytic assays demonstrated strong
killing activity against Raji targets. When evaluating activity against primary targets,
CAM161519 induced enhanced NK cell degranulation, cytokine production, and cytotoxicity
using flow cytometry-based assays against primary CLL (Figure Image) and B-ALL targets
when compared to IL-15 treatment. CAM161519 also rescued functionality of NK cells
from CLL patients against their autologous CLL cells.
Image:
Summary/Conclusion: Taken together, these data are indicative of the potential for
clinical translation of the CAM161519 TriKE in the CLL and B-ALL settings alone or
in combination with cellular products.
P619: TARGETING TRANSLATION BY DISRUPTING THE NEWLY IDENTIFIED PROHIBITIN-EIF4F COMPLEX
AS A NOVEL THERAPEUTIC STRATEGY IN CHRONIC LYMPHOCYTIC LEUKEMIA
A. Largeot1, V. Klapp1, S. Gonder1,*, E. Viry1, M. Szpakowska2, D. Perez Hernandez2,
G. Dittmar2, A. Chevigné2, L. Ysebaert3, B. Stamatopoulos4, L. Desaubry5, J. Paggetti1,
E. Moussay1
1Department of Cancer Research, Luxembourg Institute of Health, Luxembourg; 2Department
of Infection and Immunity, Luxembourg Institute of Health, Esch sur Alzette, Luxembourg;
3Service d’Hématologie, IUCT-Oncopole, Toulouse, France; 4Laboratory of Clinical Cell
Therapy, ULB-Research Cancer Center (U-CRC), Jules Bordet Institute Université Libre
de Bruxelles, Brussels, Belgium; 5Regenerative Nanomedicine Laboratory (UMR1260),
Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France
Background: CLL is the most common type of leukemia in adult, and despite great advance
in the standard of care in the last decades, there is still no cure available. CLL
cells are dependent on their microenvironment for proliferation and survival. Microenvironmental
stimuli are associated with an increase in translation globally but also at the level
of specific transcripts, including Myc (Yeomans et al., 2016, Blood).
Aims: Here, we tested the targeting of translation initiation in CLL as a novel therapeutic
strategy and identified prohibitin as partner of the translation initiation machinery.
Methods: In order to target translation, we used the FL3 molecule, a synthetic flavagline,
which is a known inhibitor of translation initiation in other types of cancers (Boussemart
et al., 2014, Nature). This drug binds to the scaffold proteins prohibitins (PHBs),
but the mechanism for translation inhibition is still unclear. PHBs are crucial partners
for the activation of the Ras-Raf-MEK-ERK pathway, ultimately leading to eIF4E (a
factor of the eIF4F translation initiation machinery) phosphorylation and to activation
of translation.
Results: We confirmed the increase in translation in human primary CLL cells upon
stimulation (A). In addition, we showed that in the Eµ-TCL1 murine model, CLL cells
have a higher translation rate compared to normal B cells and T cells. In vitro treatment
of human CLL cells (either cell lines or primary patient samples) with FL3 leads to
a decrease in translation rate. It is associated with a strong decrease of cell viability
(B) and apoptosis induction, even at a low nanomolar dose. By western blot, we showed
that in CLL, contrary to other cancer types, FL3 does not prevent RAF1 and ERK phosphorylation.
However, eIF4e phosphorylation is impaired. This observation strongly suggests a direct
impact of the drug through its molecular target PHBs on the translation initiation
machinery. By co-immunoprecipitation, proximity-ligation assay (C) and Nanoluciferase
experiments, we demonstrated the interaction between PHBs and the members of the eIF4F
complex. In addition, gene silencing of PHB by shRNA leads to a decrease in translation.
This indicates for the first time a direct role of PHBs in translation, and allows
a better understanding of FL3’s mechanism of action. We thus propose that PHB is necessary
for the correct assembly of the eIF4F complex. Interestingly, we showed that the loss
of eIF4e phosphorylation is neither responsible for the impairment of translation
nor for the decrease in cell proliferation. By pulse SILAC experiments, we determined
the proteins that are subjected to increased translation upon TLR stimulation, and
to decreased translation upon FL3 treatment. Among others, oncogenes such as c-MYC
or ETS-1 (D) have been identified. In vivo treatment of mice with FL3 after TCL1 adoptive
transfer leads to a significant delay in CLL progression and an increased survival
(E-F), demonstrating the relevance and possible impact of this molecule. Finally,
high expression of translation initiation-related genes correlated with poor survival
and unfavorable clinical parameters in CLL patients (G).
Image:
Summary/Conclusion: To conclude, we identified translation initiation as a potential
therapeutic target in CLL. The use of the inhibitor of translation FL3, efficiently
impairs proliferation of CLL cells in vitro and controls CLL development in vivo.
Moreover, we start to unveil the mechanism of action of this molecule, by the identification
of a novel interactor of the translation initiation machinery, namely prohibitins.
P620: DYNAMICS OF GENOMIC ABERRATIONS IN RELATION TO DISEASE ACTIVITY IN UNTREATED
PATIENTS WITH CHRONIC LYMPHOCYTIC LEUKEMIA
V. Navrkalova1,2,*, J. Kotaskova1,2, K. Plevova1,2, T. Reigl2, M. Valisova1, M. Bohunova1,
M. Zenatova1, L. Radova2, A. Panovska1, S. Pospisilova1,2
1Internal Medicine - Hematology and Oncology, University Hospital Brno and Masaryk
University; 2Molecular Medicine, Central European Institute of Technology, Masaryk
University, Brno, Czechia
Background: Chronic lymphocytic leukemia (CLL) manifests remarkable clinical variability
linked with extensive heterogeneity of genomic defects. Despite the progress with
novel therapeutic agents, some patients still face relapse and disease persistence
due to subclonal populations, which harbor various somatic defects, growth dynamics,
and therapy sensitivity. Clonal evolution of genomic defects was observed mainly in
treated CLL patients, whereas progression and therapy need occur at some point during
the disease course suggesting clonal dynamics even in untreated patients.
Aims: To describe impact of clonal dynamics in gene mutations and chromosomal defects
on clinical course of the disease in paired samples of untreated CLL patients.
Methods: 200 samples from 100 CLL patients were analyzed by well-established comprehensive
NGS panel LYNX (PMID 34082072) in two time points; at the diagnosis and before first
therapy. Variants in 70 genes associated with B-cell malignancies, copy-number alterations
(CNAs) and copy-neutral loss of heterozygosity (CN-LOH) across the whole genome were
evaluated by a dedicated in-house bioinformatic pipeline. Clinical data were correlated
with obtained results.
Results: The surveillance period median between two sampling points was 36 months
(range 7-174). One third of patients was still untreated during the follow-up (median
86 months, range 21-271), and the time to first treatment among 68 patients was 38
months (range 11-141). At the baseline, the most frequent defects were mutations in
NOTCH1 (15 %), SF3B1 (15 %), ATM (12 %) genes, and 13q- (56 %), 11q- (19 %) and tri12
(15 %). We observed common clonal and subclonal co-occurrence of genomic defects in
the pretreatment period. In sequential samples, we detected a change in the composition
of genomic aberrations. Most alterations were stable (59 % mutations, 70 % chromosomal
defects), but we observed dynamic changes in the rest with the predominance of growing
allelic frequency and occurrence of new defects. Unsupervised clustering according
to the presence of individual genomic defects with the emphasis on their dynamic changes
and CLL driver attribute enabled to distinguish three clusters. The correlation with
clinical data showed significant differences among clusters in IGHV status, CLL activity
(categorized according to therapy need), and an increase of absolute lymphocyte count
(ALC) per month. Obviously, the most adverse was cluster 1 with unmutated IGHV, rapid
increase in ALC, and dynamics in CLL activity. Cluster 2 seemed to be intermediate
and cluster 3 indolent. Groups also significantly differed in the presence and evolution
of 11q-, 13q-, ATM mutations, tri12, chr13 LOH, 3p-, 2p+, chr9 LOH. Importantly, stable
defects of chr13 were the most abundant in cluster 3, stable tri12 and expansions
of 13q- in cluster 2, dynamics defects including 11q-, ATM mutations, 3p-, 2p+ and
LOH 9 in cluster 1.
Summary/Conclusion: In untreated CLL patients, clonal stable genomic defects prevail.
However, common subclonal alterations and dynamic changes were observed in a considerable
proportion of patients. After unsupervised clustering, a group of patients showing
the most adverse clinical disease course harbored ATM defects, 3p-, 2p+, and chr9
LOH, possibly standing behind fast progression and therapy need. Hence, detailed monitoring
of clonal evolution can help understand the heterogeneity in the CLL activity in the
pretreatment period.
Supported by AZV NV19-03-00091, MZ-CR RVO 65269705, MUNI/A/1330/2021.
P621: SUBCLONAL EVOLUTIONARY TRAJECTORIES IN IGLV3-21R110 CHRONIC LYMPHOCYTIC LEUKEMIA
L. Paschold1,*, D. Simnica1, R. Benitez Brito2, C. Schultheiss1, T. Zhang1, C. Dierks1,
M. Binder1
1Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University
Halle-Wittenberg, Halle, Germany; 2Department of Hematology, Inselspital, University
of Bern, Bern, Switzerland
Background: There is a large body of evidence supporting the importance of antigenic
interactions of the B cell receptor (BCR) with self or non-self antigens that make
this receptor and its signaling machinery one of the most important drivers in many
types of non-Hodgkin lymphoma. This has led to a new pathobiological understanding
of these diseases as well as novel therapeutic strategies.
Chronic lymphocytic leukemia (CLL) is a prototype disease in which BCR-BCR self-interactions
induce autonomous signaling. While virtually all CLL BCR are prone to autonomous signaling
induced by such contacts, the IGLV3-21R110 molecular interaction found in 10% of CLL
patients defines a distinct clinical subset of patients that show a very aggressive
clinical course despite novel treatments. In these cases, a single amino acid replacement
from G to R located at the boundary of variable and constant region of the BCR is
an important tumor promotor driving neoplastic B cell growth.
Aims: It has been unclear how the R110 point mutation is generated. We set out to
investigate the trajectory of the malignant CLL B cell clone and subclonal heterogeneity
in IGLV3-21R110 cases in order to gain insight into the molecular pathogenesis of
this unique amino acid exchange.
Methods: We screened an initial CLL cohort of 127 cases by flow cytometry using an
anti-IGLV3-21R110 antibody. Positive cases were subjected to immunosequencing of the
light chain locus via amplicon based next-generation sequencing and characterized
for typical chromosomal deletions and driver gene mutations.
Results: We identified twelve CLL cases with IGLV3-21R110. Of these, we found a higher
frequency of mutations in ATM (42%) and SF3B1 (25%) than in unselected CLL cohorts,
which is in line with findings reported by others. As expected, the repertoire of
healthy B cells in the blood of IGLV3-21R110 CLL patients contained IGLV3-21 light
chains with the wild-type G110 residue, indicating that the R110 exchange is of acquired
nature. Interestingly, in half of the IGLV3-21R110 CLL cases we found additional clones
with IGLV3-21 and R110, including cases with divergent J gene cassettes originating
from different pre-B cells. This indicates that the R110 exchange may occur multiple
times within independent B cell clones in the same patient. Due to the unique location
at the variable-constant boundary and the high correlation of the IGLV3-21R110 patient
subset with mutations of genes involved in DNA repair (ATM/SF3B1), R110 is likely
generated as a result of incorrect V-J recombination facilitated by the error-prone-environment.
Summary/Conclusion: Our work indicates that the G110R point mutation results from
incorrect V-J rearrangements in the error-prone environment that may be induced by
SF3B1 and ATM mutations associated with this CLL subset. We found evidence for a branching
light chain evolution that leads to a striking convergence of separate clones on the
identical light chain amino acid exchange in the same patient. This strongly emphasizes
the role of this mutation as a tumor promotor involved in malignant transformation
of this subset of CLL.
P622: DOUBLE-HIT TRAF3 DELETION AND MUTATION IDENTIFIED BY HIGH-THROUGHPUT SEQUENCING
DEFINE A NEW INDEPENDENT PROGNOSTIC FACTOR IN CHRONIC LYMPHOCYTIC LEUKEMIA
C. Perez-Carretero1,2,*, M. Hernández-Sánchez1,2, M. Quijada-Álamo1,2, T. González1,2,
A. Rodríguez-Sánchez1,2, M. Martín-Izquierdo1,2, J. Matías-Martín1, A. Rubio3, J.
Dávila4, A. García de Coca5, J. Galende6, P. Jimenez-Montero7, J.-A. Queizán7, I.
González-Gascón y Marín8, J.-Á. Hernández-Rivas8, R. Benito1,2, J.-M. Hernández-Rivas1,2,
A.-E. Rodríguez-Vicente1,2
1Department of hematology, Hospital Universitario de Salamanca; 2Universidad de Salamanca,
IBSAL, Centro de Investigación del Cáncer, IBMCC-CSIC, Salamanca; 3Department of hematology,
Hospital Miguel Servet, Zaragoza; 4Department of hematology, Hospital Nuestra Señora
de Sonsoles, Ávila; 5Department of hematology, Hospital Clínico, Valladolid; 6Department
of hematology, Hospital El Bierzo, Ponferrada; 7Department of hematology, Hospital
General, Segovia; 8Department of hematology, Hospital Universitario Infanta Leonor,
Madrid, Spain
Background: Chromosome 14q abnormalities involving the IGH gene are present in 5-15%
of chronic lymphocytic leukemia (CLL) patients. Besides IGH translocations, interstitial
14q32.33/IGH deletions have been reported at lower frequencies (2%), and their clinical
implications and underlying molecular mechanisms remain unknown. In other B-cell malignancies
such as multiple myeloma, TRAF3 gene, proximal to the IGH locus, may be encompassed
in the deletion, with mutations in the remaining allele. However, the molecular status
of TRAF3 in CLL and their impact in prognosis have not been studied in depth.
Aims: To analyze the genetic landscape of CLL patients with IGH deletion and evaluate
the molecular status of TRAF3 to elucidate their impact in CLL pathogenesis and prognosis.
Methods: A total of 897 CLLs were analyzed by FISH with the IGH break-apart probe
and the 4-probe CLL panel (ATM, CEP12, D13S319 and TP53) (Vysis). 324 untreated CLLs
(54 with IGH deletion (del-3’IGH) and 270 as the control group) were sequenced by
targeted-NGS, with a custom panel including 54 CLL-related genes (Nextseq platform,
Illumina). Copy number variations (CNVs) of TRAF3 were assessed by a NGS approach
and validated by SNP Array 6.0.
Results: FISH analyses identified a total of 54 out of 897 CLLs (6%) with a 300 kb
deletion of the centromeric side of IGH (del-3’IGH CLLs). Moreover, our results showed
that 76% of del-3’IGH CLLs had additional FISH abnormalities, being the most frequent
the loss of 13q (42%), +12 (22%), del(11q) (11%) and 17p- (9%). NGS analyses showed
that the most recurrently mutated genes in del-3’IGH CLL were NOTCH1 (30%), ATM (20%)
and TRAF3 (13%). Notably, the mutational frequency of TRAF3 mutations was significantly
higher in del-3’IGH CLLs than in the control group (13% vs. 0.5%, p<0.001).
Given the surprisingly incidence of TRAF3 mutations in this subgroup, we further assessed
its CNV status, noticing that TRAF3 loss was enriched in del-3’IGH CLLs when compared
to the control group (11/54, 20% vs. 4/270, 1.5%, p<0.001). By integrating mutational
and CNV information, we observed that 7 out of 11 patients with a TRAF3 loss harbored
mutations in this gene. The deletion was present in a higher percentage of cells than
the mutation (mean: 70.5% vs. 15.6%), which may suggest that TRAF3 mutation appears
as a subclonal event, leading to a biallelic inactivation of this gene. Our results
showed a novel biallelic alteration in CLL, reminding the double null event that occurs
in other gene drivers such as TP53 and ATM.
Of note, this double-hit on TRAF3 contributed to a shorter time to first treatment
(TFT) in del-3’IGH CLL patients (5 vs. 54 months, p<0.001). Furthermore, TRAF3 biallelic
inactivation had a similar TFT than TP53 or ATM-altered patients with an adverse prognosis
(median TRAF3: 5 vs. 59 months, p<0.001, ATM: 2 vs 59, p<0.001 and TP53: 12 vs 59,
p=0.002), being also an independent risk factor in the multivariate analyses in the
CLL cohort (n=324) (HR=0.21, 95% CI=0.05-0.85, p=0.029) (Table 1).
Image:
Summary/Conclusion: Our work demonstrates that TRAF3 mutations and deletions are highly
enriched in CLL patients with del-3’IGH, resulting in the biallelic inactivation of
this gene. The presence of this double-hit on TRAF3 worsen the prognosis of CLL patients,
being and independent prognostic factor with similar impact on TFT than the biallelic
loss of TP53.
Funding: PI21/00983; FI19/00191 (CPC); CD19/00222 (MHS); FEHH (MQÁ); USAL (RBS)
P623: IBRUTINIB REGULATES MIR-181A/B VIA C-FOS IN CHRONIC LYMPHOCYTIC LEUKEMIA
A. Ramassone1,*, F. Palmiero1, A. Tomasso2, I. Innocenti2, L. Laurenti2, R. Visone3
1Center for Advanced Studies and Technology, “G. d’Annunzio” University, Chieti; 2Fondazione
Policlinico Universitario A. Gemelli IRCCS, Roma; 3Department of Medicine, Dentistry
and Biotechnology, “G. d’Annunzio” University, Chieti, Italy
Background: B-cell receptor (BCR) signalling controls B-cell survival and proliferation,
and its deregulation has a pathogenic role in Chronic Lymphocytic Leukemia (CLL),
a B-cell malignancy. Currently, several BCR signalling inhibitors are used in therapy,
among them the Ibrutinib. We previously reported that the BCR-signalling inhibition
mediated by Ibrutinib leads to miR-181a/b activation; those miRNAs are significant
biomarkers of CLL progression, and their downregulation is associated to a poor prognosis;
furthermore, low levels of miR-181a/b impair several anti-apoptotic proteins, contributing
to cell survival. BCR signalling also controls c-Fos/AP-1 pathway in pre-B cells.
Recently, we suggested the involvement of c-Fos in miR-181b expression, however the
mechanism by which this regulation is exerted is still not explored. Moreover, the
involvement of c-Fos in miR-181a/b transcription during Ibrutinib is still not clear.
Aims: Assess the impact of Ibrutinib on c-Fos expression; assess the involvement of
the transcription factor c-Fos in miR-181a/b transcription; assess the impact of miR-181a/b
in CLL cells survival during Ibrutinib treatment.
Methods: To assess the impact of Ibrutinb on c-Fos transcription, we evaluated its
expression in CLL cells treated with Ibrutinib or vehicle. To assess the involvement
of c-Fos in miR-181a/b transcription, we evaluated i) the expression of the miRs and
of their primary transcripts in CLL cells overexpressing c-Fos, and ii) the ability
of c-Fos to bind miR-181a/b host gene promoter by luciferase, ChIP and EMSA assays.
To assess the impact of the miRs on CLL cells survival during Ibrutinib treatment,
we performed MTT assay in CLL cells after miR-181a/b silencing and Ibrutinib treatment.
Results: Considering that c-Fos was found to be controlled by BCR signalling in pre-B
cells, we evaluated whether BCR signalling affect c-Fos also in CLL cells: we found
that c-Fos expression increases in primary CLL cells after Ibrutinib treatment. Since
miR-181a/b was up-regulated after Ibrutinib treatment in vitro and in vivo in CLL
cells, and considering that a possible involvement of c-Fos in miR-181b expression
was already suggested, we sought to identify if c-Fos was a direct regulator of miR-181a/b
expression. We confirmed that c-Fos overexpression affects miR-181b levels in CLL
patients, but not miR-181a; however, the dysregulation of the miR shows heterogeneity
among patients. Indeed miR-181b expression increases in 3 and decreases in 2 of 7
patients. To better understand how c-Fos impact on miRs expression, we analysed both
miR-181a and miR-181b at the transcriptional level in CLL and lymphoblastoid cell
lines, finding that c-Fos negatively regulates the pri-miR-181a/b. To clarify the
mechanism by which c-Fos regulates miR-181a/b transcription, we analysed the miRs
host gene (MIR181A2HG) promoter. Since MIR181A2HG promoter shows two consensus sequences
for the transcription factor c-Fos, we studied whether c-Fos binds these regions.
We found that c-Fos binds at MIR181A2HG promoter by recognizing the two consensus
sequences. Accordingly, after their deletion, the ability of c-Fos to recognize the
promoter was overcome. Finally, since miR-181a and miR-181b are involved in cell death,
we assessed whether those miRNAs participate in CLL cells death induced by Ibrutinib:
we found that the silencing of both miRNAs improves CLL cell survival during Ibrutinib
treatment.
Summary/Conclusion: c-Fos is a downstream target of BCR signalling and a direct regulator
of miR-181a/b transcription in CLL. MiR-181a/b are effectors of Ibrutinib-induced
CLL cells death.
P624: CHROMOTHRIPSIS IN PATIENTS WITH CLL AND COMPLEX KARYOTYPE: PATTERNS OF ABERRATIONS
AND PROGNOSTIC VALUE
S. Ramos-Campoy1,2,*, A. Puiggros1,2, J. Kamaso1,2, S. Beà3, S. Bougeon4, M. J. Larráyoz5,
D. Costa3, H. Parker6, G. M. Rigolin7, M. L. Blanco8, R. Collado9, I. Ancín10, R.
Salgado11, M. Moro12, T. Baumann3, E. Gimeno1,13, C. Moreno8,14, M. J. Calasanz5,
A. Cuneo7, J. C. Strefford6, F. Nguyen-Khac15, D. Oscier16, C. Haferlach17, J. Schoumans4,
B. Espinet1,2
1Molecular Cytogenetics Laboratory, Pathology Department, Hospital del Mar; 2Translational
Research on Hematological Neoplasms Group, Cancer Research Program, Institut Hospital
del Mar d’Investigacions Mèdiques (IMIM); 3Hematopathology Unit, Hospital Clínic,
Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBERONC, Barcelona,
Spain; 4Oncogenomic Laboratory, Hematology Service, Lausanne University Hospital,
Lausanne, Switzerland; 5Department of Genetics, Universidad de Navarra, Pamplona,
Spain; 6Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton,
United Kingdom; 7Hematology Section, St. Anna University Hospital, Ferrara, Italy;
8Department of Hematology, Hospital Universitari de la Santa Creu i Sant Pau, Barcelona;
9Department of Hematology, Consorcio Hospital General Universitario, Valencia; 10Department
of Hematology and Hemotherapy, Hospital Universitario Cruces, Bilbao; 11Department
of Hematology, Fundación Jiménez Díaz, Madrid; 12Department of Hematology, Hospital
Universitario Central de Asturias, Oviedo; 13Applied Clinical Research in Hematological
Malignances, Cancer Research Program, Institut Hospital del Mar d’Investigacions Mèdiques
(IMIM), Barcelona; 14Josep Carreras Leukemia Research Institute, Badalona, Spain;
15Hematology Department and Sorbonne University, Hôpital Pitié-Salpêtrière, APHP,
INSERM U1138, Paris, France; 16Department of Molecular Pathology, Royal Bournemouth
Hospital, Bournemouth, United Kingdom; 17MLL Munich Leukemia Laboratory, Munich, Germany
Background: Chromothripsis is a unique catastrophic event found in 1-5% of chronic
lymphocytic leukemia (CLL) patients, associated with poor prognostic factors and short
survival (Edelmann et al, 2012; Puente et al, 2015; Leeksma et al, 2021). It is characterized
by the presence of multiple genomic rearrangements in one chromosome, resulting in
oscillating switches between 2-3 copy number states by chromosomal microarrays (CMA)
or next-generation sequencing (Stephens et al, 2011). Optical genome mapping (OGM)
is a new method based on the imaging of long DNA molecules (>250kb) labelled at specific
motifs, generating a unique pattern. This allows mapping of the genomic location of
each molecule and detection of structural and numerical chromosomal abnormalities
with high resolution and sensitivity.
Aims: 1. To analyze the genome complexity associated with chromothripsis and the patterns
of rearrangements detected by OGM; 2. To determine the prognostic impact of chromothripsis
in a cohort of CLL patients enriched in complex karyotypes (CK).
Methods: A total of 33 patients with chromothripsis detected by CMA were included:
11 with chromothripsis-like patterns (≥7 switches), 22 with classical chromothripsis
(≥10 switches). Nine of the cases were studied with OGM (Bionano Genomics). Clinico-biological
features and time to first treatment (TTFT) were analyzed. Results were compared with
a cohort of 129 patients showing CK without chromothripsis (control group) (Ramos-Campoy
et al, 2021).
Results: All patients with chromothripsis showed a high genomic complexity (30/33
with CK by chromosome banding analysis and 3/33 classified as complex by CMA [range:
10-24 CNV, being at least three of them >5Mb (range: 3-6)]. Forty-six chromothriptic
events were identified. These affected 1-4 chr/patient, but a single chromosome was
involved in 25/33 patients (76%). Chromothripsis was distributed throughout all chromosomes,
most frequently affecting 3 (n=5), 6 (n=5) and 13 (n=5). The events were mainly multiple
losses (25/46) or alternated gains and losses (19/46). Size and coordinates of CNVs
detected by OGM and CMA were highly concordant. OGM detected multiple rearrangements
cryptic by CMA (median: 9/chromothriptic event). Two patterns of chromothripsis-related
rearrangements were identified: one clustered in the chromothriptic region (3/9) and
the other involving both chromothriptic and non-chromothriptic chromosomes (range:
1-6 chr) (6/9). One patient showed 36 rearrangements throughout the genome, suggesting
the coexistence of chromothripsis and chromoplexy, characterized by the presence of
multiple chained translocations (Figure). Patients with chromothripsis showed a higher
frequency of TP53
del/mut (70% vs 39%; p=0.001) and a shorter TTFT (15m vs 2m; p=0.013) than the control
group. No differences were observed for IGHV and del(11q) status. When stratifying
patients based on TP53 status, the presence of chromothripsis was not associated with
shorter TTFT. In the multivariate analysis including TP53, genomic complexity by CMA
and chromothripsis, only TP53 was statistically significant (HR=1.6; p=0.029).
Image:
Summary/Conclusion: 1. Chromothriptic events involve multiple loci in CLL patients;
2. OGM allows not only the detection of CNV but also the identification of two rearrangement
patterns associated with chromothripsis (clustered or involving non-chromothriptic
chromosomes) whose clinical impact should be further explored. 3. In CLL patients
with CK, the adverse prognosis of chromothripsis is associated with a higher frequency
of TP53
del/mut.
P625: SINGLE-CELL RNA SEQUENCING REVEALS THE HETEROGENEITY OF TUMOR CELLS AND IMMUNE
MICROENVIRONMENT IN RICHTER SYNDROME AND ACCELERATED CLL
Y. Sha1,2,*, Y. Miao1,2, S. Qin1,2, R. Jiang1,2, W. Wu1,2, Y. Xia1,2, L. Wang1,2,
L. Fan1,2, W. Xu1,2, H. Jin1,2, H. Zhu1,2, J. Li1,2
1Department of Hematology, The First Affiliated Hospital of Nanjing Medical University;
2Department of Hematology, Pukou CLL Center, Pukou division of Jiangsu Province Hospital,
Nanjing, China
Background: Richter Syndrome (RS) is defined as development of aggressive lymphoma
arising from chronic lymphocytic leukemia/Small lymphocytic leukemia (CLL/SLL). Accelerated
CLL/SLL (aCLL/SLL) is pathologically defined as a highly proliferative form of CLL/SLL
(either expanded proliferation center or Ki-67>40%). The underlying mechanism behind
transformation from indolent CLL/SLL to aggressive DLBCL remains largely unknown.
The biological difference between RS DLBCL and aCLL/SLL has not been explored yet.
Aims: To explore intra- and inter-tumor and immune microenvironment (TME) heterogeneity
of RS and aCLL/SLL. To disclose the molecular mechanism underlying transformation
from CLL/SLL to DLBCL.
Methods: Single-cell RNA sequencing (scRNA-seq) of paired lymph node (LN) and peripheral
blood (PB) was done among 2 RS (P1, P2) and 2 aCLL/SLL (P3, P4) patients (pathologically
diagnosed in LN, Fig. A) and bioinformatics analysis was conducted.
Results: Identification of cell type in 8 samples showed great heterogeneity (Fig.
B). Single-cell trajectory analysis was done using “monocle” inferred tumor development
and differentiation of each patient. Monoclonal B cells with high Ki-67 expression
and high proportion of G2M phase cells were enriched in the end of pseudotime trajectories
(Fig. C). Further analysis of CD5, CD20 and CD23 expression alongside pseudotime trajectories
were also conducted, indicating that our trajectories could depict the transformation
from indolent CLL/SLL to aggressive DLBCL. Therefore, cell states were clustered according
to the trajectories. P1 and P2 were clustered as seven groups respectively (P1A-G,
P2A-G) and P3 were clustered as three groups (P3A-G). Notably, although minor proportion
of proliferating cells were enriched in the end of P4 pseudotime trajectories, clusters
could not be split in P4 patients. All cell states groups were mapped to UMAP analysis
and the proportion of G2M phase was highest in state G of RS patients. Single-sample
GSEA (ssGSEA) analysis was done to disclose the pathway activation in transformation
process and found that pathways including cell cycle, TCA cycle, spliceosome, MYC
targets, unfolded protein response, epigenetics, DNA repair, glycolysis and oxidative
phosphorylation were gradually activated alongside trajectories from state A-G in
RS. Moreover, pathways including NOTCH, P53, PI3K-AKT, Antigen processing and presentation,
apoptosis, hypoxia and etc. were found significantly upregulated in RS (Fig. E). Further
analysis of immune microenvironment of RS and aCLL/SLL showed that exhaustion T cells
were significantly enriched in RS lymph node compared with aCLL/SLL, indicating the
immune surveillance escape mediated by exhaustion T cells might prompt DLBCL transformation.
PDCD1 (PD-1), CTLA4, LAG3, HAVCR2 (TIM3), TIGIT expression were significantly upregulated
in exhaustion T cells of RS patients, suggesting potential treatment target of RS
for immune checkpoint inhibitors (Fig. F).
Image:
Summary/Conclusion: Our study was the first one using scRNA-seq to disclose the tumor
heterogeneity and immune microenvironment of RS and aCLL/SLL in single-cell dimension.
Evolution trajectories from indolent CLL/SLL to aggressive DLBCL were constructed
and pathway activation in the process of transformation were further explored. Moreover,
immune microenvironment analysis of RS and aCLL/SLL showed that exhaustion T cells
were significantly enriched in RS lymph node and immune checkpoints including PDCD1
(PD-1), CTLA4, LAG3, HAVCR2 (TIM3), TIGIT were significantly upregulated in exhaustion
T cells of RS patients, suggesting potential treatment target of RS.
P626: FUNCTIONAL SCREENING OF PI3K INHIBITORS STRATIFIES RESPONDERS TO IDELALISIB
AND INDICATES DRUG CLASS ACTIVITY IN IDELALISIB-REFRACTORY CLL
S. Skånland1,*, Y. Yanping1, P. Athanasiadis1, L. Karlsen1, A. Urban1, I. Murali2,
S. Fernandes2, A. Hilli3, K. Taskén1, F. Bertoni4, A. Mato5, E. Normant6, J. Brown2,
G. Tjønnfjord1, T. Aittokallio1
1Oslo University Hospital, Oslo, Norway; 2Dana-Farber Cancer Institute, Boston, United
States of America; 3Diakonhjemmet Hospital, Oslo, Norway; 4Università della svizzera
italiana, Bellinzona, Switzerland; 5Memorial Sloan Kettering Cancer Center; 6TG Therapeutics,
New York, United States of America
Background: The phosphatidylinositol 3-kinase inhibitors (PI3Ki) idelalisib and duvelisib
are approved for relapsed chronic lymphocytic leukemia (CLL). While patients may show
an initial response to these therapies, development of treatment resistance or intolerance
remains clinical challenges. Prediction of individual treatment responses based on
clinically actionable biomarkers is needed to overcome these challenges.
Aims: 1. To characterize functional responses to 10 PI3Ki in CLL
2. To study PI3Ki drug class activity in idelalisib-refractory CLL
3. To investigate whether ex vivo drug sensitivity can predict in vivo treatment responses
Methods: CLL cells from patients that were treatment naïve (n=7), idelalisib refractory
(n=9), or on idelalisib treatment (longitudinal samples from n=6 patients) were screened
against 10 PI3Ki (buparlisib, compound 7n, copanlisib, duvelisib, idelalisib, nemiralisib,
pictilisib, pilaralisib, umbralisib, ZSTK474), both alone and in combination with
the B-cell lymphoma 2 (Bcl-2) antagonist venetoclax. Two idelalisib-resistant lymphoma
cell lines were used as reference. Treatment effects on cell signaling were profiled
using phospho flow and effects on cell viability were monitored with detection of
cleaved caspase-3 and using the CellTiter-Glo assay.
Results: Pan-PI3Ki were most effective in inhibiting PI3K signaling both after 30 min
and 24h exposure. Inhibition of cell signaling correlated with induction of apoptosis.
Treatment with a PI3Ki induced cell death in 72h CellTiter-Glo assays, although to
varying extent. The pan-PI3Ki were overall most potent in inhibiting CLL cell viability.
Interestingly, pan-PI3Ki exhibited sustained activity both in idelalisib-resistant
lymphoma cell lines and in CLL cells from idelalisib-refractory patients. Combination
treatment with a PI3Ki and venetoclax resulted in synergistic induction of apoptosis.
Notably, CLL cells from idelalisib-refractory patients remained sensitive to idelalisib+venetoclax
combination. Ex vivo drug testing on CLL cells from a patient who presented with relapsed
disease after sequential treatment with FCR, ibrutinib, idelalisib and venetoclax
revealed sensitivity to PI3Ki+venetoclax treatment. The patient consequently started
treatment with idelalisib+venetoclax. She obtained an initial partial response, but
then suffered intolerable adverse effects and the therapy was stopped. Protein profiling
of the patient’s CLL cells collected at 5 time-points before, during and after the
combination regimen showed that B-cell signaling was reduced by the treatment, but
again significantly increased when the therapy was stopped. Increased phosphorylation
of Bruton’s tyrosine kinase (BTK) and extracellular signal-regulated kinase (ERK)
at the latest time-point was mirrored by increased sensitivity to venetoclax combined
either with a BTK or MEK inhibitor. Unfortunately, the patient passed away and the
efficacy of these alternative therapies could not be tested. To more systematically
assess the predictive value of ex vivo drug sensitivity data, we analyzed drug responses
to 73 combinations on CLL cells from patients treated with idelalisib. CLL cells collected
at baseline from patients who obtained a long-term response to idelalisib showed significantly
higher drug sensitivity scores than CLL cells from patients who obtained a short-term
response.
Summary/Conclusion: Our findings indicate PI3Ki drug class activity in idelalisib-refractory
CLL, and suggest that ex vivo drug sensitivity may guide precision medicine and predict
treatment responses. These results warrant further testing in larger cohorts and in
clinical trials.
P627: COMBINED DRUG AND CRISPR/CAS9 SCREENING REVEALS SPECIFIC TARGETS FOR SF3B1-
AND NOTCH1-MUTATED CLL CELLS
L. Dostálová1,2, A. Ladungová1,3, D. Škrnová1, N. Varma Gottumukkala1,2, Y. Lodhi1,2,
M. Smida1,4,*
1CEITEC Masaryk University; 2Department of Biology, Faculty of medicine, Masaryk university;
3National Centre for Biomolecular Research, Faculty of Science, Masaryk university;
4Department of internal medicine - Oncology and hematology, University hospital Brno
and Medical faculty of Masaryk university, Brno, Czechia
Background: Chronic lymphocytic leukemia (CLL) is genetically heterogeneous disease
with a plethora of mutations occurring in relatively low fraction of patients. Mutations
in SF3B1 and NOTCH1 genes are among the most frequent recurrent mutations found in
10-20% CLL cases. The presence of these mutations is considered as a negative prognostic
factor, yet, there is no specific treatment available to achieve more prominent response.
Aims: The aim of this study was to use the combination of CRISPR/Cas9 screening and
high-throughput drug screening on cells with introduced SF3B1 or NOTCH1 mutations
in order to reveal unique vulnerabilities specific to the cells carrying these frequent
CLL mutations.
Methods: Using CRIPSR/Cas9-based homology directed repair, we have introduced specific
mutations most recurrent within the hotspots of SF3B1 (K700E) and NOTCH1 (P2514fs)
into CLL-derived cell line HG3. These two knock-in cell line variants were then subjected
to a drug screening with a library of 859 approved drugs, enriched with oncology-indicated
drugs. Cell viability in response to 10 uM library concentration upon 72h was assessed
by Cell-Titer Glo and evaluated against the wild-type counterparts. Simultaneously,
genome-wide CRISPR/Cas9 knockout screen was performed in these cell lines to identify
genes whose deletion is selectively lethal to SF3B1 or NOTCH1-mutated cells.
Results: The CRISPR/Cas9 dropout screen has revealed several interesting genes that
are in synthetic lethal relationship with the introduced point mutations. In particular,
SPDYE1, a gene involved in cell cycle regulation, and LUC7L3, involved in splicing
and cAMP regulatory element binding, were found to be synthetically lethal with the
NOTCH1 mutation. On contrary, SNUPN, responsible for transport of spliceosomal snRNPs,
and UQCRC1, involved in cytochrome c regulation and oxidative phosphorylation, were
found to be essential for SF3B1-mutated cells. Drug screening using library of approved
drugs demonstrated general hypersensitivity of both the mutant cell lines and the
wild-type controls to the inhibitors of proteasome and HDAC enzymes. In addition,
exquisite sensitivity of NOTCH1-mutant and SF3B1-mutant cell lines, contrary to their
wild-type counterparts, was revealed towards the inhibitors of various hormone receptors
or inhibitors of 20S proteasome.
Summary/Conclusion: The combination of diverse screening approaches may reveal novel
vulnerabilities specific for individual mutations frequently recurring in CLL patients.
Thorough validations of the identified hits is ongoing, together with the analyses
into the possible mechanisms behind these unique targets.
This research was partly financed by the grant MUNI/A/1330/2021, MUNI/IGA/1516/2020
and MUNI/A/1419/2021.
P628: HIGH THROUGHPUT IMMUNOGENETIC EVIDENCE FOR EXTENSIVE IN VIVO CLASS SWITCH RECOMBINATION
EVENTS IN CLL
E. Sofou1,2,*, A. Agathangelidis3, A.-M. Antoniadou1, E. Georgiou2, M. Papaioannou4,
N. Pechlivanis1, F. Psomopoulos1, K. Stamatopoulos1, A. Chatzidimitriou1
1Institute of Applied Biosciences, Centre for Research and Technology Hellas; 2Laboratory
of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki,
Thessaloniki; 3Department of Biology, School of Science, National and Kapodistrian
University of Athens, Athens; 41st Department of Internal Medicine, School of Medicine,
Aristotle University of Thessaloniki, Thessaloniki, Greece
Background:
In vivo Class Switch recombination (CSR) has previously been reported for a fraction
of patients with chronic lymphocytic leukemia (CLL) expressing unmutated IGHV genes
(U-CLL) and high mRNA levels of activation-induced cytidine deaminase compared to
patients expressing mutated IGHV genes (M-CLL). This observation is certainly intriguing,
however inherently limited due to the low-sensitive, Sanger-based approach used for
the immunogenetic characterization.
Aims: Here, we sought to overcome the aforementioned limitation and obtain more insight
into in vivo CSR in CLL using a highly-sensitive, next generation sequencing (NGS)
methodology.
Methods: We studied 73 patients (48 U-CLL, 25 M-CLL) sampled at diagnosis. Case selection
was based on the dominant expression of the IgM isotype, as determined by flow cytometry.
IGHV-IGHD-IGHJ gene rearrangements were RT-PCR amplified for the μ, γ, and α transcripts
utilizing IGHV Leader and IGHC-specific primers. All obtained PCR products (n=215)
were subjected to paired-end NGS. High-quality, productive IGHV-IGHD-IGHJ gene rearrangements
(n=23,217,561; median= 97,932/sample) were annotated with IMGT/HighV-QUEST. Clonotypes
were defined as rearrangement sequences expressing the same IGHV gene and identical
complementarity-determining region 3 (VH CDR3) amino-acid (AA) sequence. Clonotype
variants were defined as different nucleotide sequences clustered in the same clonotype.
Results: The IGH gene repertoire of μ transcripts was clearly monoclonal in all cases,
showing a dominant CLL clonotype (DCC) with a median frequency of 93.5% for either
U-CLL or M-CLL. Subsequently, the IGH gene repertoires of γ and α transcripts were
scanned for the presence of the DCC. In U-CLL, 46/48 cases expressed switched transcripts
with median frequencies of 14% (range 0.01-97.2%) and 21.3% (range 0.1-95.3%) for
γ and α transcripts, respectively. In M-CLL, 24/25 patients expressed switched transcripts
albeit at significantly lower levels than U-CLL (p=0.01) (median frequencies: 0.2%
and 13.6%; ranges 0.03-94% and 0.04-91.2% for the γ and α transcripts, respectively).
Interestingly, within U-CLL, cases belonging to stereotyped subset #1 (n=21) and its
satellite subset #99 (n=2) exhibited significantly higher levels of switched transcripts
(median frequencies: 37.9% and 37.6% for the γ and α transcripts, respectively) than
all other cases (p=0.04 for γ transcripts, p=0.002 for α transcripts). Analysis of
somatic hypermutation (SHM) in the DCC within the repertoires of μ, γ and α transcripts
of U-CLL cases revealed the presence of shared SHMs (even among cases with 100% IGHV
gene identity) but also distinct SHMs shared by the γ and α clonotypic transcripts
but not the IgM, indicative of common selection pressure post-CSR. Interestingly,
M-CLL patients displayed less intraclonal diversity in the γ and α clonotypic transcripts,
despite the significant burden of SHM within the IgM DCC.
Summary/Conclusion: In conclusion, our data reveal that the B cell receptor IGH gene
repertoire is highly dynamic and interconnected in both U-CLL and M-CLL, albeit considerably
more pronounced in the former. This is especially the case for patients assigned to
the clinically aggressive stereotyped subsets #1 and #99, prompting speculations regarding
the precise nature of the respective affinity maturation process. Shared SHMs between
clonotype variants only among clonotypic γ and α transcripts rather than the μ transcripts
indicate that CSR likely precedes SHM, particularly in U-CLL.
P630: TARGETING NEGATIVE REGULATION OF MAPK SIGNALING IN CHRONIC LYMPHOCYTIC LEUKEMIA
M. Stumpf1,*, V. Ecker1, P. Giansanti2, J. Lu3, T. Zenz4, I. Ringshausen5, B. Küster6,
J. Ruland1, M. Buchner1
1TranslaTUM - Central Institute for Translational Cancer Research, Technische Universität
München, München; 2Bavarian Biomolecular Mass Spectrometry Center (BayBioMS), Technical
University of Munich, Freising; 3European Molecular Biology Laboratory (EMBL), Heidelberg,
Germany; 4Department of Medical Oncology and Hematology, University Hospital and University
of Zurich, Zurich, Switzerland; 5Wellcome Trust/MRC Cambridge Stem Cell Institute
and Department of Haematology, Jeffrey Cheah Biomedical Centre, University of Cambridge,
Cambridge, United Kingdom; 6Chair of Proteomics and Bioanalytics, Technical University
of Munich (TUM), Freising, Germany
Background: The MAPK-ERK pathway is a key signaling cascade for cellular homeostasis.
Following MAPK activation, feedback loops increase the production of pathway inhibitors,
such as dual specificity phosphatases (DUSPs), to restrict MAPK signal. DUSPs can
inhibit tumor growth by preventing ERK activation and may act as oncogenes by helping
tumor cells in adjusting to elevated MAPK levels. Despite mounting evidence that MAPK
signaling is critical for CLL formation and progression, little is known about the
role of MAPK negative feedback via the DUSP family in CLL.
Aims: In this study, we investigated the significance of DUSP1 and DUSP6’s negative
regulation of MAPK in CLL and evaluated their usage as possible therapeutic targets
in drug resistant disease.
Methods: We examined the expression levels of DUSP1 and DUSP6 in primary CLL patients
using RNAseq and immunoblot analysis. To determine their functional relevance, we
employed the small molecule inhibitor BCI on primary murine and human (drug resistant)
CLL cells in vitro and in vivo and generated genetic knockout MEC-1 CLL cells, to
validate the inhibitor effects. In addition, we investigated the downstream effects
of DUSP1/6 inhibition on CLL signaling events using global phosphoproteomics in primary
CLL and MEC-1 cells. Finally, using particular inhibitors in in vitro experiments,
we confirmed the importance of discovered pathway activations.
Results: To evaluate a potential role of negative feedback regulation of MAPK signaling
in CLL, we first analyzed mRNA expression levels of DUSP1 and DUSP6 in CLL samples
(n=210). Both negative regulators were expressed in all of the CLL samples studied,
with DUSP6 levels being upregulated in cases with activating mutations in the KRAS
or BRAF genes (12.1±17 RNAseq counts for BRAF/KRASwt cases vs. 46.7±19 RNAseq counts
for BRAF/KRAS mutated cases). Furthermore, after microenvironmental stimulation, DUSP
levels increased in CLL lymph nodes compared to peripheral blood (DUSP1: log2-fold
change of 2.6, p=0.0094; DUSP6: log2-fold change of 1.8, p=2.2*10e-7; Herishanu et
al, 2011) and stromal coculture. Notably, increased DUSP6 mRNA expression was linked
to a poor clinical outcome, as evidenced by a shorter time to treatment (p=0.0109;
Log-rank Test) and overall survival (p=0.0075; Log-rank Test). We next used the dual-specific
DUSP1/6 inhibitor BCI to block the phosphatase activity and found that this causes
cell death in CLL cells, but not or only mildly affected the survival of healthy donor
B cells or other T- and B-cell lines. In the TCL1-driven CLL mouse model, administration
of the DUSP1/6 inhibitor is effective in lowering CLL cell progression (>50% reduction
of total CLL cell count in the spleen; n=5; p=0.0029). We generated genetic knockout
lines of DUSP1 or DUSP6, which inhibited CLL cell expansion, to confirm the inhibitor’s
specificity. We next used phosphoproteomics to explore the signaling consequences
of DUSP1/6 inhibition in CLL, and discovered that DUSP1/6 inhibition caused acute
stimulation of BCR/MAPK signaling. Subsequently, the DNA damage response (DDR) pathway
is activated, resulting in the induction of apoptotic cell death. Finally, we tested
whether DUSP1/6 inhibition is efficacious in drug-resistant CLL by using BCI to treat
therapy-naive and ibrutinib refractory CLL samples, and we discovered that DUSP1/6
inhibition is extremely successful in targeting treatment-resistant CLL.
Summary/Conclusion: Conclusion: Based on these results, we propose transient DUSP1/6
inhibition as a novel and unexpected concept for eliminating drug-resistant CLL cells.
P631: A PCR-BASED ASSAY FOR IGLV3-21R110 SCREENING CONFIRMS ITS PROGNOSTIC VALUE IN
AN INDEPENDENT COHORT OF 613 PATIENTS WITH CHRONIC LYMPHOCYTIC LEUKEMIA.
C. Syrykh1,2,*, N. Russiñol1, T. Baumann3,4, M. Kulis1,5, M. Alcoceba5,6, M. González5,6,
E. Colado7, Á. R. Payer7, M. Aymerich1,3,5, M. J. Terol8, S. Ruiz-Gaspà1, A. López-Guillermo1,3,5,9,
J. I. Martín-Subero1,5,9,10, D. Colomer1,3,5,9, J. Delgado1,3,5,9, E. Campo1,3,5,9,
F. Nadeu1,5
1Institute d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain;
2Department of Pathology, Institut Universitaire du Cancer, CHU de Toulouse, Toulouse,
France; 3Hospital Clinic of Barcelona, Barcelona; 4Hospital Universitario 12 de Octubre;
5Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid; 6Biología
Molecular e Histocompatibilidad, IBSAL-Hospital Universitario, Centro de Investigación
del Cáncer-IBMCC (USAL-CSIC), Salamanca; 7Servicio de Hematología y Hemoterapia, Hospital
Universitario Central de Asturias, Oviedo; 8Servicio de Hematología, Hospital Clínico
Universitario, INCLIVA, Universidad de Valencia, Valencia; 9Universitat de Barcelona;
10Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
Background: Recent studies have shown that expression of the immunoglobulin lambda
light chain IGLV3-21 gene carrying a point mutation in the amino acid 110 (named IGLV3-21R110)
defines a subset of chronic lymphocytic leukemia (CLL) with an intermediate epigenetic
subtype and poor prognosis (Maity et al. PNAS, 2020; Nadeu et al. Blood, 2021). This
mutation is found in up to 6.5% of patients with CLL at diagnosis and up to 18% of
cases enrolled in clinical trials. In addition, IGLV3-21R110 seems to have prognostic
value independently of the IGHV gene somatic hypermutation (SHM) status, stereotype
subset #2, and disease stage, which makes it a promising prognostic biomarker. However,
further studies on independent cohorts are needed to support its application in clinical
practice.
Aims: To develop a rapid detection method of the IGLV3-21R110 mutation and to assess
its prognostic significance in a cohort of 613 previously unpublished CLL patients.
Methods: A multiplex IGLV3-21R110 mutation-specific polymerase chain reaction (msPCR)
assay was established in a cohort of 12 patients (including 6 IGLV3-21R110 mutated)
and validated in 159 cases (including 7 mutated; Nadeu et al. Blood, 2021). This msPCR
assay was applied on an independent cohort of 613 CLL patients (575 CLL, 38 monoclonal
B-cell lymphocytosis). Clinical analyses were performed for time to first treatment
(TTFT) and overall survival (OS) from time of diagnosis considering only cases diagnosed
as CLL.
Results: A msPCR approach was designed integrating two forward primers aligning to
distinct regions of the IGLV3-21 gene and two R110-specific reverse primers matching
the IGLJ1 and IGLJ2/3 genes, respectively. A third pair of primers targeting exon
9 of FBXW7 was used as an internal control. PCR conditions were set up on a cohort
of 12 cases and subsequently validated on 159 previously published cases with 100%
concordance. We then applied this msPCR assay to a cohort of 613 previously unpublished
CLL patients and identified the R110 mutation in 22 (3.6%) cases, including 75% mutated
IGHV (M-CLL) and 25% unmutated IGHV (U-CLL). Moreover, 84.6% of IGLV3-21R110 cases
carried non-stereotyped immunoglobulin genes while the remaining stereotyped IGLV3-21R110
cases were subset #2. Ten of the IGLV3-21R110 cases were classified according to the
epigenetic subtypes and all belonged to the intermediate CLL (i-CLL) subgroup. Clinically,
M-CLL patients carrying the IGLV3-21R110 as well as i-CLL with IGLV3-21R110 had a
shorter TTFT compared to M-CLL and i-CLL lacking IGLV3-21R110, respectively, and similar
to U-CLL/naïve-like CLL (p<0.005). Multivariable analyses including IGLV3-21R110,
disease stage, and IGHV gene SHM status or epigenetic subtypes confirmed the independent
prognostic value of IGLV3-21R110 on TTFT (p<0.005). IGLV3-21R110 had also a prognostic
impact on OS since M-CLL carrying the IGLV3-21R110 had shorter OS similar to that
of U-CLL (Figure). Finally, we combined these novel data with those of our previously
published 489 CLL cohort (Nadeu et al. Blood, 2021) (N total=1102). Clinical analyses
of the whole cohort confirmed the independent prognostic value of IGLV3-21R110.
Image:
Summary/Conclusion: We have developed a reliable, easy-to-use msPCR assay suitable
for IGLV3-21R110 screening in large cohorts. By applying this msPCR on a cohort of
613 CLL patients, our results corroborate the relevance of IGLV3-21R110 testing to
improve the risk stratification of CLL patients in the clinical practice, especially
within M-CLL and i-CLL subtypes.
P632: ORAL AND GUT MICROBIAL DIVERSITY CORRELATES WITH PROGNOSTIC FEATURES IN CHRONIC
LYMPHOCYTIC LEUKEMIA
M. Szelest1,*, M. Paziewska1, M. Kiełbus1, M. Masternak1,2, J. Zaleska1, E. Wawrzyniak3,
E. Kalicińska4, P. Jabłonowska4, T. Wróbel4, A. Wolska-Washer5,6, J. Z. Błoński3,6,
K. Giannopoulos1,2
1Department of Experimental Hematooncology, Medical University of Lublin; 2Department
of Hematology, St John’s Cancer Centre, Lublin; 3Department of Hematology, Medical
University of Lodz, Lodz; 4Department of Haematology, Blood Neoplasms and Bone Marrow
Transplantation, Wroclaw Medical University, Wroclaw; 5Department of Experimental
Hematology, Medical University of Lodz; 6Copernicus Memorial Hospital, Lodz, Poland
Background: For the last years it has been speculated that immunologic and inflammatory
factors, including antigen stimulation, could be involved in the pathogenesis of chronic
lymphocytic leukemia (CLL). With increasing evidence that the activation of cellular
proinflammatory signaling pathways driven by somatic mutations and an increased release
of proinflammatory cytokines is associated with CLL development, it seems relevant
to investigate the role of chronic inflammation in the pathogenesis of this disease.
Recent studies have revealed that the intestinal microbiota could modulate the activity
of immune system through creating an imbalance between cell proliferation and apoptosis.
Thus, the relationship of microbial dysbiosis and specific taxon abundances with CLL
biology and clinical outcome should be examined.
Aims: As it was reported that the activity of human microbiome is involved in cancer
development and host response against anticancer therapy, the aim of this study was
to investigate the stool and oral microbiome alterations in CLL patients with respect
to selected prognostic and predictive markers.
Methods: The study included 92 patients with untreated CLL. Microbiota composition
of 85 stool samples and 76 oral samples from these cases were determined by 16S rRNA
next-generation sequencing and analyzed with respect to the Binet stage, IGHV and
TP53 mutation status, CD38 and ZAP-70 expression, and cytogenetic aberrations. The
species richness and evenness were analyzed using Shannon and Chao1 indexes, respectively.
The DADA2 tool was used to classify sequences into appropriate taxonomic groups. The
differences in taxonomic profiles and species diversity in oral and stool samples
with distinct prognostic features were investigated using the Wilcoxon or Kruskall-Wallis
tests.
Results: Alpha-diversity analysis indicates that there were no significant differences
in richness and evenness between probes with distinct CLL features in both fecal and
oral samples. However, at phylum level, an increase in Proteobacteria, a common feature
of gut dysbiosis, was observed in stool samples from patients with stage Binet C compared
to stage Binet A (p=0.042), and in oral samples with del11q compared to samples with
no del11q (p=0.021). The abundance of Bacteroidota was higher in fecal samples collected
from patients with stage Binet A in comparison to stage Binet B (p=0.051) and C (p=0.009).
Moreover, Bacteroidota was relatively more abundant in oral samples from patients
with stage Binet A in comparison to stage Binet C (p=0.008). Bacteroidota was significantly
less abundant in stool samples from patients with unmutated IGHV compared to samples
with mutated IGHV (p=0.029). Stool samples from patients with CD38+ exhibited decreased
abundance of Bacteroidota and increased abundance of Firmicutes in comparison to CD38-
cases (p=0.031 and p=0.022, respectively). Furthermore, fecal samples from patients
with del6q showed a higher abundance of Firmicutes and lower abundance of Bacteroidota
compared to samples with no del6q (p=0.006 and p=0.018, respectively). Additionally,
a relative increase in Actinobacteriota and Firmicutes was found in oral samples with
tri12 in comparison to samples with no tri12 (p=0.039 and p=0.016, respectively).
Image:
Summary/Conclusion: Our findings provide new insights into oral and gut microbial
diversity which is related to distinct prognostic features in CLL and might point
to the inflammatory-related modulation of the clinical course of disease.
P633: ACTIVE CHROMATIN REGULATORY LANDSCAPE OF STEREOTYPED SUBSETS IN CHRONIC LYMPHOCYTIC
LEUKEMIA REVEALS A DISTINCTIVE SIGNATURE IN SUBSET #8
M. Tsagiopoulou1,2,*, V. Chapaprieta3, N. Russiñol3, N. Pechlivanis1, N. Papakonstantinou1,
N. Stavroyianni4, F. Psomopoulos1, E. Campo3, K. Stamatopoulos1, J. I. Martin-Subero3
1Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki,
Greece; 2CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science
and Technology (BIST), Barcelona, Spain; 3Institut d’Investigacions Biomèdiques August
Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain; 4Hematology Department
and HCT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
Background: Chronic lymphocytic leukemia (CLL) patients with stereotyped B cell receptors
have been characterized from the perspective of genetic and DNA methylation alterations.
Recent studies have characterized the chromatin landscape of non-stereotyped CLLs
and normal B cells (Beekman, et al. Nat Med. 2018).
Aims: To characterize the active chromatin regulatory landscape of 4 major stereotyped
subsets as compared to IGHV status-matched non-stereotyped CLLs.
Methods: We used chromatin-immunoprecipitation followed by sequencing (ChIP-Seq) with
an antibody for the H3K27ac (a bona fide histone mark for regulatory element activation)
in sorted CLL cells from 15 normal B cell subpopulations and 44 CLL, including 18
cases from stereotyped subsets #1, #2, #4, and #8. To understand their functional
impact, we integrated our data with 16 RNA-seq data considering the 3D chromatin configuration
(Hi-C data: GM12878 and a representative U-CLL case) (Figure 1)
Results: Unsupervised principal component analysis (PCA) of H3K27ac data revealed
different profiles in CLL as a whole, regardless of stereotypy status, vs normal B
cells (PC1, 25%). Of note, PC2 (14%) revealed a clear separation between subset #8,
a highly aggressive CLL subtype with the highest risk of Richter’s transformation
amongst all CLL, vs other CLLs or normal B cells, suggesting that subset #8 may have
a specific acetylation pattern. Indeed, supervised differential acetylation analysis
(FDR<0.01) uncovered a signature of 903 Differentially Acetylated Regions (DAR) in
subset #8 vs U-CLL (Figure 1) and no differences for the other subsets. This signature
was unrelated to +12, highly prevalent (~60%) in subset #8, or IgG expression, a hallmark
of subset #8. Hierarchical clustering analysis of this signature revealed 6 clusters
of regions with different chromatin dynamics highlighting the de novo active cluster1
(n=197 regions) in subset #8 vs other CLLs or normal B cells. These differentially
acetylated regions (DARs) were enriched (FDR<0.05) in binding sites of several differentially
expressed TFs in #8 vs U-CLL, e.g. members of SOX, GATA, and CEBP families as well
as MEF2A and HNF1A, which showed the highest enrichment. Next, we used the topologically
associating domains (TAD) resulting from Hi-C data and RNAseq data to identify the
potential target genes of the 197 de novo DARs in subset #8. We identified the target
genes in 78/197 (39.6%) DARs that were linked to 112 genes, as some active regulatory
regions were associated with more than one gene. Among them, the DAR located in chr9:137085994-137086793 bp
was related to the expression levels of 8 genes (LCNL1, CLIC3, FBXW5, AJM1, MAMDC4,
CCDC183, TRAF2 and an un-annotated transcript) within the same TAD. This region also
contained 6 additional H3K27ac peaks with higher signal in subset #8, though not reaching
significance. This finding suggests deregulation of a broad region in chromosome 9
leading to the coordinated upregulation of 8 transcripts. Concerning the activated
genes, TRAF2 is a remarkable example since TRAF family members (TRAF6, -4, -1) are
found overexpressed in U-CLL and here we report an active region in subset #8 leading
to TRAF2 overexpression.
Image:
Summary/Conclusion: We report a subset #8-specific chromatin acetylation signature
containing de novo activated regions, linked to the upregulation of target genes,
in some cases including blocks of adjacent genes. This study further supports the
distinct biological nature of subset #8 and may help to understand the aggressive
clinical behavior of this distrinctive CLL variant.
P634: MHC-BASED LARGE-SCALE SCREENING FOR ANTI-TUMOR T CELLS IN CHRONIC LYMPHOCYTIC
LEUKEMIA REVEALS CD8+ T CELLS WITH SPECIFICITY AGAINST THE CLONOTYPIC B-CELL RECEPTOR
IMMUNOGLOBULIN
A. Vardi1,2,*, Y. Basavaraju3, A. Agathangelidis2,4, N. Wulff Pedersen3, M. Karipidoy2,
A.-L. Schaap-Johansen3, A. Fylaktou5, N. Stavroyianni1, M. Iskas1, A. Anagnostopoulos1,
A. Chatzidimitriou2, P. Marcatili3, S. R. Hadrup3, K. Stamatopoulos2
1Hematology Department & HCT Unit, G.Papanikolaou Hospital; 2Institute of Applied
Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece; 3Department
of Health Technology, Technical University of Denmark, Lyngby, Denmark; 4Department
of Biology, School of Science, National and Kapodistrian University of Athens, Athens;
5Department of Immunology, National Peripheral Histocompatibility Center, Thessaloniki,
Greece
Background: Chronic lymphocytic leukemia (CLL) is currently incurable, indicating
a need for novel strategies towards disease eradication, including reinvigoration
of anti-tumor immune responses. T cells in CLL appear selected by restricted antigens,
with evidence suggesting that the selecting epitopes may lie within the clonotypic
B-cell receptor immunoglobulins (BcR IG).
Aims: We previously performed ad hoc prediction of putative T-cell class I neoepitopes
contained within the clonotypic BcR IG of CLL patients. Here, we performed major histocompatibility
complex (MHC)-based large-scale screening to identify autologous CD8+ T cells recognizing
the predicted neoepitopes.
Methods: We evaluated 653 peptides derived from the clonotypic BcR IG of 25 CLL patients
(median 21 predicted neoepitopes/patient, across 13 MHC-I alleles). Considering the
MHC-I typing of each patient, we constructed patient-specific peptide-MHC multimers
labeled with a unique DNA barcode plus a fluorochrome (PE). We generated MHC-specific
multimer libraries which we then mixed with respective autologous T-cell enriched
peripheral blood mononuclear cells (PBMCs). Duplicate samples/patient were analyzed.
In addition, known viral peptide-MHC multimers labeled with a different fluorochrome
(APC) as well as three MHC-matched healthy donor PBMCs were used as controls. PE-
and APC-positive multimer-binding CD8+ T cells were sorted for each cell sample; subsequently,
DNA barcode amplification and sequencing was performed. Sequencing data were processed
(Barracoda software) to obtain the number of clonally reduced barcode reads assigned
to a given sample and peptide-MHC specificity. The number of clonally reduced reads
for a given pMHC specificity was used to estimate the frequency of T cells specific
for a given epitope, based on the average number of T cell receptor–MHC multimer interactions
detected in the total MHC multimer-binding T cell pool in a given cell sample. Peptides
with a log fold change >2 were considered as significant [log fold change: (number
of DNA barcode reads associated with a specific pMHC)/3x (number of total barcode
reads derived from the same sample)].
Results: Overall, 3 peptide-MHC multimers were recognized by CD8+ T cells: (i) VTVADTAVYY
(peptide A) and (ii) INLNPSLKRR (peptide B), both within the context of the A03*01
allele, and (iii) YSFTSYWINW (peptide C) within the context of the A24*02 allele.
Peptide A derived from the IGHV4-34 FR3 region of a somatically hypermutated clonotypic
BcR IG, containing a single A to V somatic hypermutation (SHM) at position 96. Peptide
B derived from the IGHV4-39 CDR2-FR3 junction of a somatically hypermutated clonotypic
BcR IG, containing 3 SHMs (T to I at position 65, Y to L at position 67 and S to R
at position 74). Peptide C derived from the IGHV5-10-1 CDR1-FR2 junction of a clonotypic
BcR IG assigned to stereotyped subset #1, containing the sole SHM of the IG (S to
N at position 40). The immunogenicity of these peptides was further corroborated by
the fact that they were recognized not only by the autologous T cells of the patient
from whom the peptide derived, but also by T cells of other patients as well as a
healthy donor sharing the respective MHC allele.
Summary/Conclusion: In conclusion, we offer proof of concept that the targeted SHM
which shapes the CLL BcR IG repertoire may produce idiotypic targets for T cell-based
therapy or peptide vaccine design. Characterization of the epitope-binding T cells
is currently underway by our group, aiming to provide further insight on how these
cells could be recruited into effective anti-tumor responses.
P635: TARGETING THE ONE-CARBON METABOLISM AS NOVEL THERAPEUTIC STRATEGY IN CHRONIC
LYMPHOCYTIC LEUKEMIA
E. Viry1,*, A. Largeot1, S. Gonder1, S. Chemlal1, N. Kiweler2, J. Meiser2, T. Helleday3,
E. Moussay1, J. Paggetti1
1Tumor Stroma Interactions, Department of Cancer Research; 2Cancer Metabolism Group,
Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg;
3Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute,
Solna, Sweden
Background: Chronic Lymphocytic Leukemia (CLL), the most common type of leukemia in
adults, is characterized by the clonal expansion of CD5+ CD19+ B cells. CLL progression
is highly dependent on complex interactions with non-malignant cells of the tumor
microenvironment. Despite great advance in the standard of care in the last decades,
there are still unmet medical needs (long-life treatment, resistance…). Altered cellular
metabolism has emerged as a hallmark of cancer by sustaining the uncontrolled growth
of cancer cells. The one-carbon pathway is a major driver for tumor proliferation,
providing building blocks for biosynthesis of nucleotides through pyrimidines (dTTP)
and purines synthesis, redox metabolism (GSH), and energy balance by suppling ATP
and NADPH to cells (A). Understanding the metabolic reprogramming occurring in cancer,
notably in CLL, may provide insights to support the development of novel therapies.
Aims: We aim to get insights into one-carbon metabolism regulation in CLL, identify
new therapeutic targets and find new treatment options for CLL patients.
Methods: Through a collaboration with a biotech company that develops inhibitors of
the one-carbon metabolism, we tested new MTHFD1/2 inhibitors, which we believe, are
promising in the context of CLL. The cytotoxic activity of the inhibitors was evaluated
on a panel of murine and human CLL cell lines and primary cells, and also on others
B cell malignancies such as Mantle Cell Lymphoma (MCL), Diffuse Large B-Cell Lymphoma
(DLBCL), and Multiple Myeloma (MM). The molecular mechanisms sustaining the cytotoxic
activity were evaluated by performing metabolic tracing, rescue experiments and CRISPR/Cas9
KO.
Results: We showed a strong expression of both MTHFD1 and 2 in all the cancer cell
lines tested and in stimulated primary human CLL cells. A higher expression of both
enzymes was also observed in primary CLL cells, isolated from the Eµ-TCL1 murine model
of CLL compared to normal B cells. In vitro treatment with the inhibitors efficiently
reduced cell viability of CLL cell lines and primary cells, as for MCL and DLBCL cell
lines, at low nanomolar dose while no effect was observed on MM cell lines (B). This
effect is associated with a blockade of cell proliferation. Using 13C-serine isotope
tracing, we could showed that the inhibitors significantly reduced ATP production
from serine in CLL cells, probably resulting in altered nucleotides biosynthesis.
By performing rescue experiments with thymidine or hypoxanthine, we confirmed that
the cytotoxic effect of the inhibitors on CLL cells is mediated through a defect in
pyrimidine synthesis (C). Interestingly, we demonstrated that the resistant MM cell
lines overexpressed the SHMT1 enzyme, and could be sensitized to MTHFD1/2 inhibition
by combination therapy with SHMT1/2 inhibitor (D). Taken together these results suggest
that SHMT1 may be a key player in the response to MTHFD1/2 inhibition. Furthermore,
we have previously reported that CLL induces a dramatic remodeling of tumor microenvironment,
notably leading to the accumulation of highly immunosuppressive Tregs (Wierz et al.,
Blood, 2018). We showed that both enzymes are overexpressed in those highly immunosuppressive
Tregs, and that MTHD1/2 inhibitors are able to reduce the viability of ex vivo polarized
Tregs.
Image:
Summary/Conclusion: MTHFD1/2 inhibitors display a high cytotoxic activity in CLL and
other B-cell malignancies by impairing cell proliferation through a defect in pyrimidine
synthesis. These inhibitors also exhibit cytotoxic effect on activated Treg cells
ex vivo, reinforcing its therapeutic potential for CLL treatment.
P636: REAL WORLD OUTCOME WITH IBRUTINIB IN PATIENTS WITH CHRONIC LYMPHOCYTIC LEUKEMIA
FROM THE GERMAN REALITY STUDY
M. Welslau1,*, R. Schlag2, B. Heinrich3, L. Vornholz4, S. Buchholz4, B. Marquard4,
S. Dörfel5, G. Anke6
1Oncology and Hematology, MVZ at Medical Clinic Aschaffenburg-Alzenau, Aschaffenburg;
2Oncology and Hematology, Oncological Practice, Würzburg; 3Oncology and Hematology,
Oncological Practice, Augsburg; 4Medical & Scientific Affairs, Janssen-Cilag GmbH,
Neuss; 5Oncology and Hematology, Onco Center Dresden, Dresden; 6Hematological and
Oncological Diseases, MVZ Potsdam, Potsdam, Germany
Background: Chronic lymphocytic leukemia (CLL) is one of the most common forms of
leukemia, characterized by abnormal survival and proliferation of mature B cells in
peripheral blood, bone marrow, and lymph nodes. Ibrutinib is a once-daily Bruton’s
tyrosine kinase inhibitor (BTKi) with a significant benefit in both progression free
survival (PFS) and overall survival (OS) demonstrated in multiple phase 3 trials for
patients (pts) with previously untreated (1L) or relapsed/refractory (R/R) CLL. In
2014, ibrutinib was approved by the European Medicines Agency and made available to
CLL pts in Germany. Prior to the REALITY study, limited data were available on the
effectiveness of ibrutinib-treated pts with CLL in routine clinical practice in Germany.
Aims: REALITY was a prospective, multicenter, non-interventional study to evaluate
the effectiveness of ibrutinib as the 1st, 2nd, or ≥3rd line of treatment (cohorts
C1, C2, and C3 respectively) in pts with CLL during the first 2 years of observation
period in a real-world setting.
Methods: 302 pts were enrolled in the three cohorts (C1: 104 pts; C2: 90 pts; C3:
108 pts) and treated in 57 study sites in Germany from January 2017 to July 2021.
Written informed consent was obtained from all pts. Key study endpoints include retention
rate (defined as ratio of pts on ibrutinib treatment to the number of pts at risk),
start of next therapy, PFS, OS, and safety. The results of this study are descriptive,
and no formal hypotheses was pre-specified.
Results: Median age at time of ibrutinib treatment initiation was 74.0 years for the
1L pts in C1, and 72.5 and 73.0 years in the R/R cohorts of C2 and C3, respectively.
In cohort C1 (1L), pts with genetic high risk features del(17p) and TP53 mutation
were more frequent compared to C2 and C3. In C1 del(17p) was reported in 34/51 (66.7%)
pts and TP53 mutations in 31/49 (63.3%) pts. In the R/R cohorts C2 and C3 del(17p)
was reported in 11/57 (19.3%) pts and 13/65 (20.0%) pts, respectively. TP53 mutations
were reported in 14/48 (29.2%) pts in C2 and 21/40 (52.5%) pts in C3.
The primary study endpoint was retention rate, 77.9% pts continued to be on treatment
after 1 year in C1 (1L). In C2 and C3 (R/R), the 1-year retention rates were 74.4%
and 58.3% respectively.
In C1, with median follow-up of 30.6 months, median PFS was not reached (NR). The
1-year PFS and OS in the C1 cohort (1L) were 91.3% and 96.2%, respectively. Only 8
pts initiated next line treatment in C1 after 1 year.
In C2, with a median follow-up of 31.5 months, median PFS was NR. The 1-year PFS and
OS were 85.6% and 93.3%, respectively. 11 pts initiated next line treatment in C2
after 1 year.
In C3 median follow-up was 30.9 months and median PFS was NR. The 1-year PFS and OS
were 83.3% and 93.5%, respectively. 26 pts initiated next line treatment in C3 after
1 year.
Detailed results after 1 and 2 years of observation are summarized in Table 1.
No new safety signals for ibrutinib were observed.
Image:
Summary/Conclusion: This first, comprehensive data from clinical practice of ibrutinib-treated
pts with CLL in Germany showed that ibrutinib is a highly effective treatment option,
especially as a 1st line treatment. The effectiveness and safety profile were consistent
with clinical trials.
P637: FRONTLINE THERAPY CLL WITHOUT 17P DEL/P53 ABERRATIONS: SYSTEMATIC REVIEW AND
BAYESIAN NETWORK META-ANALYSIS
G. Caridà1, D. Ciliberto1, G. Pelaia2, N. Staropoli1, M. A. Siciliano3, N. D. Calandruccio1,
R. Toscano4, M. Rossi5,6,*
1Oncology Unit; 2Pediatric Unit, “Magna Graecia” University, Catanzaro; 3Oncology
Unit, “S. Giovanni di Dio” Hospital, Crotone; 4Oncology Unit, AO “S. Francesco di
Paola”, Paola (CS); 5Department of Experimental and Clinical Medicine, Magna Græcia
University, Catanzaro, Italy, “Magna Graecia” University; 6Hematology Unit, AO “Pugliese
Ciaccio”, Catanzaro, Italy
Background: Chronic Lymphocytic Leukemia (CLL) is the most common adult leukemia,
with a median age at diagnosis of 72 years. IgVh unmutated CLL without 17p del/p53
aberrations (UN-only CLL) still carry a poorer prognosis as compared to IgVh mutated
(MU-only) cases. Although many treatment options are currently available for the front-line
therapy, the best choice and treatment sequencing stratified on efficacy and tolerability
for UN/MU-only CLL remains to be defined.
Aims: We performed a systematic review and Bayesian network meta-analysis to define
best frontline therapy and treatment sequence for UN/MU-only CLL.
Methods: Analysis of the literature was performed through the main databases. Due
to different therapeutic approaches to front-line therapy of CLL, we chose to conduct
a Bayesian Network Meta-Analysis (NMA), allowing to rank (from best to worst) multiple
treatments in a single analysis. We generated a rank for Progression Free Survival
(PFS), Overall Response Rate (ORR), Minimal Residual Disease (MRD), assessed by flow
cytometry on peripheral blood, and tolerability. PFS was evaluated in unselected population
for IgVh, and in UN/MU-only CLL patients.
Results: Fifteen clinical trials, with a total of 7958 patients, have been included,
while 16 different treatments were considered to build four different network meta-analyses
to identify the best treatment in PFS, ORR, MRD, and tolerability: Acalabrutinib (ACA);
ACA + Obinutuzumab (ACA-O); Alemtuzumab; Bendamustine + Rituximab; Chlorambucil; Chlorambucil
+ O; Chlorambucil + Ofatumumab; Chlorambucil + Rituximab; Fludarabine + Cyclophosphamide
+ Rituximab; Fludarabine + Cyclophosphamide; Ibrutinib (IBR); IBR + O; IBR + Rituximab;
Lenalidomide, Venetoclax + IBR (VEN-IBR), Venetoclax + Obinutuzumab + Ibrutinib (VEN-O-IBR),
Venetoclax + O (VEN-O). ACA-O scored the best in PFS in an unselected population (SUCRA
100%, probable the best 99.9%), UN- and MU- only CLL patients. Our analysis showed
that VEN-O ranked the first for ORR (SUCRA value 100%, probably the best 100%). VEN-O
ranked the first also for MRD (SUCRA 90.4%, probably best 36.2%) versus the second
VEN-O-IBR (SUCRA 84.0%, probably best 35.9); in the direct comparison, the difference
between VEN-O and VEN-O-IBR is not statistically significant (Odds Ratio 1.44, CrI
0.00,563.57). Finally, ACA monotherapy is the most tolerable therapy (SUCRA value
100%, probably the best 100%), whereas IBR-O turned to be the most toxic treatment
(SUCRA value 0%, probably the best 1%). Interestingly, IBR monotherapy ranked the
9th in the network, whereas all combination regimens including Obinutuzumab carried
a high risk of toxicity.
Image:
Summary/Conclusion: To our knowledge, this is the first network meta-analysis that
compares at the same time PFS, tolerability, ORR, and MRD. Our PFS analysis showed
that ACA-O was the best treatment in both MU/UN-only patients. Furthermore, ACA turned
out to be the most tolerated regimen, whereas the BTKis-O based regimens significantly
carried a high toxicity profile as compared to BTKis alone, counterbalancing the advantage
of O-addition in terms of ORRs, where O-based regimens achieved the higher rank, when
combined either with VEN or BTKis. ACA-O was the best treatment for PFS, although
with a worse toxicity profile as compared to ACA monotherapy. VEN-O scores as the
best in ORR and MRD. The combination ACA-O has the highest probability to be the best
treatment for prolonging the PFS in untreated, UN/MU-only CLL patients. ACA monotherapy
is a valuable alternative considering both the safety and efficacy profile.
P638: THE RISK OF SECOND PRIMARY MALIGNANCIES AND CAUSE-SPECIFIC MORTALITY AMONG PATIENTS
WITH T-CELL PROLYMPHOCYTIC LEUKEMIA IN THE UNITED STATES: A POPULATION-BASED STUDY.
K. Chamarti1,*, M. Mannem1, B. Bakhati1
1Department of Internal Medicine, Texas Tech University Health Sciences Center at
Permian Basin, Odessa, United States of America
Background: T cell prolymphocytic leukemia (T-PLL) is a rare lymphoid malignancy,
compromising 2% of mature lymphocytic leukemias. The T-PLL follows an aggressive clinical
course with median survival of 7.5 months with conventional chemotherpay. In the era
of targeted therapy with anti-CD52 antibody for patients with T-PLL, there is data
lacking on the spectrum of second primary malignancies (SPMs) and cause-specific mortality.
Aims: In this current study we aim to identify the risk of SPM and cause specific
mortality in patients with T-PLL in the era of targeted therapies with anti-CD52 antibodies
using a national registry from the United States.
Methods: The incidence of SPMs and cause-specific mortality among two-month survivors
of T-PLL, was estimated using the National Cancer Institute’s Surveillance, Epidemiology,
and End Results (SEER)-13 registries. We included patients from 1992-2018 with ICD-O-3
code 9834/3 from the registry. We estimated the risk of SPMs using standardized incidence
ratios (SIRs) and cause-specific mortality by standardized mortality ratios (SMRs)
and absolute excess risk (AER)/10,000 population.
Results: We included a total of 314 patients with T-PLL in this study. The median
age at diagnosis was 68.5 years (range 1-85 years). The median follow-up duration
was 12 months (range 0-196 months). A total of 13 (4%) patients developed 14 SPMs
at the last follow-up. The SPM risk for the cohort was significantly elevated (SIR
2.54, 95% confidence interval [CI] 1.39-4.27) compared with general population. The
median latency period for SPM development was 28 months (range 8-108 months). The
SIR was significant among males (SIR 2.96, 95% CI 1.42-5.44), but not in females (SIR
1.88, 95% CI 0.51-4.82). The SIR according to age groups was significant in patients
with age < 60 years (SIR 3.84, 95% CI 1.25-8.97), but not in age > 60 years (SIR 2.14,
95% CI 0.98-4.06). Specific SPMs with the highest risk included nasopharyngeal cancers
(SIR 200.87, 95% CI 5.09-1,119.20) and multiple myeloma (SIR 33.39, 95% CI 6.89-97.58).
At last follow-up, 155 (49%) patients had died, of which 89% were due to malignant
neoplasms. There was statistically significant risk of mortality due to all malignant
cancers combined (SMR 58.35, 95% CI 49.03-68.94), largely due to mortality from lymphocytic
leukemia (SMR 2587, 95% CI 2021-3264, AER 1757), non-Hodgkin lymphoma (SMR 100, 95%
CI 45.88-190.45, AER 220), stomach and duodenal ulcers (SMR 73.74, 95% CI 1.87-410.84,
AER 24.43), uterine cancer (SMR 66.55, 95% CI 1.68-370.78, AER 24.39), and pneumonia
and influenza (SMR 11.88, 95% CI 2.45-34.71, AER 68.04). The risk of death from lymphocytic
leukemia was highest within the first year of diagnosis of T-PLL and continued to
decline slowly thereafter, although it remained statistically significantly elevated
more than other causes of death (SMR 1yr=3405, AER 2666; SMR 1-5 yrs=2623, AER 1715;
SMR 5-10 yrs=1015, AER 516).
Summary/Conclusion: Our study reveals that the risk of developing SPMs remains elevated
in patients with T-PLL compared to the general population. Additionally, men are at
an increased risk for SPMs compared to women. Despite therapeutic advances in T-PLL,
hematologic neoplasms (including lymphocytic leukemia) have remained the leading cause
of death in the last three decades. Further studies are needed to better define the
healthcare needs of T-PLL patients beyond initial treatment to reduce the burden of
mortality from T-PLL and other SPMs.
P639: OUTCOMES FOR PATIENTS WITH CHRONIC LYMPHOCYTIC LEUKEMIA PREVIOUSLY TREATED WITH
A COVALENT BTK AND BCL2 INHIBITOR IN THE UNITED STATES: A REAL-WORLD DATABASE STUDY
A. R. Mato1,*, Y. Chen2, A. C. Girvan2, L. M. Hess2, L. Bowman2, P. Abada2, H. Konig2,
R. A. Walgren2
1Memorial Sloan Kettering Cancer Center, New York; 2Eli Lilly and Company, Indianapolis,
United States of America
Background: Covalent BTK inhibitors (cBTK) and B-cell lymphoma 2 inhibitors (BCL2i)
have transformed outcomes in patients with chronic lymphocytic leukemia (CLL). Although
these agents are highly effective, they are not curative, and a proportion of patients
will require additional therapy. No prospective randomized clinical trials have evaluated
the efficacy of therapies following treatment with these two modern drug classes.
Aims: We therefore evaluated treatment outcomes in patients with CLL following treatment
with at least these two drug classes using a real-world dataset from the United States
(US).
Methods: De-identified patient-level electronic medical record data linked to claims
data in the US from ConcertAI were utilized for this study. Eligible patients were
≥18 years old, diagnosed with CLL between December 2011 and October 2020. To ensure
a minimum of 1 year follow-up for all patients, data were available through July 2021.
Patients were required to have completed treatment with at least one cBTKi and a BCL2i
during the 1st-3rd lines of therapy. Time-to-event analyses utilized the Kaplan-Meier
method.
Results: 382 patients (median age 73.4 years, male 64.4%) met eligibility criteria.
Of these, 228 (59.7%) received subsequent therapy following BTK inhibitor and BCL2
inhibitor regimens, while the remaining 154 (40.3%) did not receive further treatment.
Among those who received further treatment, the most common therapies included retreatment
with a venetoclax-containing regimen (n=112, 49.1%). Of note, only 2.6% (n=6) of patients
received immediate post-dual failure therapy with a PI3K inhibitor in this setting.
Among those who received subsequent therapy (n=228), the median time from the start
of the immediate post-cBTKi/BCL2i therapy to discontinuation or death was 5.5 months
(95% CI: 3.5-6.9). For the entire cohort of patients with CLL who discontinued BTKi
and BCL2i regimens (n=382), the median time from the end of the last of BTKi/BCL2i
therapy to discontinuation of subsequent therapy or death was 5.6 months (95% CI:
4.3-6.0).
Summary/Conclusion: Patients with CLL who have been previously treated with both a
cBTKi and BCL2i experience poor outcomes as observed by duration of treatment and
time to treatment discontinuation of subsequent therapy. This population represents
a growing area of unmet medical need for patients with CLL.
P640: PIRTOBRUTINIB, A HIGHLY SELECTIVE, NON-COVALENT (REVERSIBLE) BTK INHIBITOR IN
COMBINATION WITH VENETOCLAX±RITUXIMAB IN RELAPSED/REFRACTORY CLL: RESULTS FROM THE
BRUIN PHASE 1B STUDY
L. E. Roeker1,*, A. R. Mato1, J. R. Brown2, C. C. Coombs3, N. N. Shah4, W. G. Wierda5,
M. R. Patel6, K. L. Lewis7, M. Balbas8, J. Zhao8, N. C. Ku8, J. F. Kherani8, D. E.
Tsai8, B. Nair8, C. Y. Cheah7
1Memorial Sloan Kettering Cancer Center, New York; 2Dana-Farber Cancer Institute and
Harvard Medical School, Boston; 3University of North Carolina at Chapel Hill, Chapel
Hill; 4Medical College of Wisconsin, Milwaukee; 5MD Anderson Cancer Center, Houston;
6Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, United States
of America; 7Linear Clinical Research and Sir Charles Gairdner Hospital, Perth, Australia;
8Loxo Oncology at Lilly, Stamford, United States of America
Background: Covalent Bruton tyrosine kinase inhibitors (BTKi) have transformed the
management of chronic lymphocytic leukemia (CLL), but patients (pts) discontinue these
agents due to resistance or intolerance. Pirtobrutinib is an oral, highly selective,
non-covalent (reversible) BTKi with promising efficacy and safety in heavily pretreated
relapsed/refractory (R/R) CLL pts, regardless of BTK C481 mutation status. Recent
clinical studies reported on the safety and efficacy of time-limited venetoclax and
covalent BTKi combination regimens.
Aims: We evaluated the safety and efficacy of pirtobrutinib combined with venetoclax
± rituximab in pts with R/R CLL.
Methods: BRUIN is a phase 1/2 global, multicenter study (NCT03740529) of pirtobrutinib
in pts with advanced B-cell malignancies. The phase 1b portion evaluated the safety
of pirtobrutinib at a continuous dose of 200 mg QD from Cycle 1, Day 1 plus venetoclax
starting on Cycle 2, Day 1 with a standard 5-week dose ramp to 400 mg QD (PV) and
PV plus rituximab at 375 mg/m2 on Cycle 1, Day 1, then 500 mg/m2 on Day 1 of Cycles
2-6 (PVR). Prior BTKi was allowed; prior venetoclax was not permitted. Objectives
included safety and overall response rate (ORR) of each combination.
Results: As of 27 SEP 2021, 15 pts received PV and 10 pts received PVR. Median age
was 66 years (range, 39-78). Median prior lines of therapy was 2 (range, 1-4). The
majority of pts in both cohorts had received prior chemotherapy (56%, n=14), CD20
monoclonal antibody (72%, n=18), and/or covalent BTKi (68%, n=17). No dose-limiting
toxicities were reported. Safety profiles were generally similar across both cohorts.
The most common treatment-emergent adverse events (TEAE) of any grade, regardless
of attribution, were neutrophil count decrease (36%), nausea (32%), fatigue (32%),
diarrhea (28%), and constipation (24%). The only Grade ≥3 TEAE to occur in more than
2 pts was neutrophil count decrease (36%, n=9). One pt experienced Grade 4 clinical
tumor lysis syndrome with resultant acute kidney injury during venetoclax dose escalation,
which resolved with supportive measures. No pts discontinued treatment due to AEs.
For the 22 pts with efficacy data available as of 03 NOV 2021, median duration of
follow-up was 9 months (range, 3.9-15) and the ORR was 95.5% (95% CI, 77-100). All
responding pts except 1 remain on therapy (PVR responder discontinued due to death
unrelated to study treatment). As all responses were ongoing and early, and MRD analysis
was not yet performed.
Summary/Conclusion: Pirtobrutinib combined with venetoclax ± rituximab was well tolerated
and had a safety profile consistent with known drug class findings and no clear additive
toxicities in pts with R/R CLL. Early results demonstrate promising efficacy with
combination therapy.
P641: RETREATMENT WITH VENETOCLAX AFTER VENETOCLAX, OBINUTUZUMAB +/- IBRUTINIB: POOLED
ANALYSIS OF 13 PATIENTS WITH CHRONIC LYMPHOCYTIC LEUKEMIA (CLL) TREATED IN GCLLSG
TRIALS
P. Cramer1,*, M. Fürstenau1, A. Giza1, S. Robrecht1, E. Tausch2, C. Schneider2, C.-M.
Wendtner3, M. Hoechstetter3, J. Schetelig4, S. Böttcher5, P. Dreger6, A.-M. Fink1,
P. Langerbeins1, O. Al-Sawaf1, K. Fischer1, S. Stilgenbauer2, B. Eichhorst1, M. Hallek1
1Department I of Internal Medicine and German CLL Study Group; Center for Integrated
Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Cologne;
2Department III of Internal Medicine, University Hospital Ulm, Ulm; 3Department of
Hematology, Oncology, Immunology, Palliative Care, Infectious Diseases and Tropical
Medicine, Klinikum Schwabing, Munich; 4Department I of Internal Medicine, University
Hospital Carl Gustav Carus, Dresden; 5Department III of Internal Medicine, University
Hospital Rostock, Rostock; 6Department V of Internal Medicine, University Hospital
Heidelberg, Heidelberg, Germany
Background: Fixed duration venetoclax (V)-containing therapies are commonly used in
CLL, either combined with obinutuzumab (Ven-G) for treatment-naïve or with rituximab
for relapsed/refractory disease. Current clinical trials also combine Ven-G with ibrutinib
(GIVe) or acalabrutinib (GAVe) to increase efficacy and possibly shorten treatment
duration with minimal residual disease (MRD)-guided discontinuation strategies.
Aims: There is only little evidence regarding the best sequence of currently available
therapies and if time-limited therapies can be repeated at progression. Therefore,
we pooled the patients (pts) with two consecutive V-containing treatment lines from
GCLLSG trials, to study efficacy and safety of a re-treatment with V-based regimens.
Methods: Thirteen pts with two sequential V-containing lines (V1 and V2) were identified
within the following 5 multicenter phase-II or -III trials: 3 pts each received V1
in CLL2-BAG and CLL2-GIVe, 2 Ven-G in CLL13 (excluded from time-to event analyses
due to unavailable data) and 1 in CLL14; all were subsequently treated in CLL2-BAAG
(V2). Four additional pts received V1 and V2 in CLL2-BAG.
Results: At the start of V1, the median age was 58 (range 49-65) years and 11 pts
(92%) had a high/very high CLL-IPI, all 13 pts had an unmutated IGHV, 8 (62%) a del(17p)
and/or TP53mut and 6 (46%) a complex karyotype. Four pts had already received a median
of 2.5 (range 1-4) prior treatment lines (chemo(immuno)therapies only).
V1 was Ven-G in 10 pts, including 4 pts with a bendamustine debulking, and GIVe in
3 pts. Median duration of V1 was 13 (range 11-29) months for all pts and 14 (range
13-26) months in the 9 pts with a MRD-guided discontinuation strategy who achieved
uMRD, one pt failed to achieve uMRD in bone marrow and had to stop after approximately
2.5 years as per protocol. Three pts received V1 with a fixed duration of approximately
one year.
V2 was GAVe in 9 pts, including 2 with a bendamustine debulking, and Ven-G or Ven
in 2 each, all with a MRD-guided discontinuation. Thus far, 5 pts have stopped V2:
two pts due to uMRD and one each due to adverse events (allergic skin reactions),
planned allogeneic stem cell transplantation and end of study treatment after approximately
2.5 years. Median treatment duration was 16 (range 8-30) months for V2, which is already
longer than with V1 (see above) but 8 pts still continue V2.
All pts experienced adverse events (AEs) during V1 and V2, AEs CTC °III/IV occurred
in 11 and 8 pts during V1 and V2, respectively and serious AEs in 6 and 4, respectively.
Hematological AEs °III/IV occurred in 6 and 2 pts during V1 and V2, tumor lysis syndromes
°III in 2 and 3 pts and infections °III/IV in 2 each.
All pts responded to V1 and V2, uMRD in PB was achieved at the end of V1 in 12/13
pts (92%) and thus far with V2 in 9/13 pts (69%). Median time between end of V1 and
start of V2 was 29 (range 15-55) months. Median progression-free survival from start
of V1 was 42 months and after V2 no progressions occurred so far at a median follow
up time of 19 (range 8-33) months. All pts are alive, see also Fig. 1.
Image:
Summary/Conclusion: In this pooled analysis of 13 pts with two consecutive time-limited
V-containing therapies, which includes mainly pts with adverse risk factors and a
short remission duration, V-based re-treatment appeared to be safe and efficacious:
no increased rate of AEs was seen so far and all pts responded with at least 2/3 even
achieving uMRD again.
P642: IMPACT OF TYPE 2 DIABETES ON SURVIVAL AND TREATMENT IN PATIENTS WITH CHRONIC
LYMPHOCYTIC LEUKEMIA
E. Curovic Rotbain1,2,3,4,*, K. Rostgaard2,4, M. A. Andersen1,4,5, N. Vainer1, C.
Da Cunha-Bang1, H. Hjalgrim1,2,4,6, H. Frederiksen3,7,8, S. Slager9, C. Utoft Niemann1,6
1Department of Hematology, Rigshospitalet; 2Hematology Group, Danish Cancer Society
Research Center, Copenhagen; 3Department of Hematology, Odense University Hospital,
Odense; 4Department of Epidemiology, Statens Seruminstitut; 5Department of Clinical
pharmacology, Bispebjerg and Frederiksberg Hospital; 6Department of Clinical Medicine,
Copenhagen University, Copenhagen; 7Departemnt of Clinical Research, University of
Southern Denmark; 8Academy of Geriatric Cancer Research (AgeCare), Odense University
Hospital, Odense, Denmark; 9Department of Health Sciences Research, Mayo Clinic, Rochester,
United States of America
Background: Because of a generally advanced age at diagnosis of chronic lymphocytic
leukemia (CLL), this group of patients is often burdened by comorbid conditions. Comorbid
conditions are associated with increased mortality in CLL, due to causes both related
and unrelated to CLL. Diabetes is one of the most common comorbid conditions with
a prevalence of 8-21% at time of CLL-diagnosis, and the prevalence of diabetes is
projected to double in the general population within the next two decades. While many
comorbid conditions may have limited potential for improvement of management, treatment
for type 2 diabetes (T2D) is rapidly evolving with new treatment combinations displaying
survival benefits.
Aims: To assess the impact of T2D on time to treatment and mortality for patients
with CLL from time of diagnosis and from time of first-line treatment.
Methods: In Denmark, all citizens have access to free health care services. Contacts
with the health care system are registered by the unique personal identification number,
enabling linkage of data on person level across national registers. We followed all
patients registered with newly diagnosed CLL in the Danish CLL Register or the Danish
Cancer Register in the period 2002-2018 from 1 month after CLL-diagnosis until death
or end of follow-up. Data on causes of death were obtained through the Danish National
Register of Causes of Death. T2D was defined as having an International Classification
of Diseases-10 diagnosis of T2D (E11) in the Danish National Patient Register or ≥1
prescription for non-insulin anti-diabetic drugs (ATC code A10B) in the Danish Prescription
Register. Fine-Gray and Cox proportional hazards regressions were fitted, and corresponding
Kaplan-Meier curves computed.
Results: In total, 7 446 patients with CLL were identified, of whom 802 (11%) had
T2D at CLL diagnosis. T2D was associated with longer time to first treatment (hazard
ratio (HR) 0.81, 95% confidence interval (CI) [0.67-0.99]) and higher mortality (HR
1.58, 95% CI [1.42-1.76]) from time of CLL diagnosis. Patients with T2D had an increased
mortality due to causes both related (HR 1.15, 95% CI [0.99-1.34]) and unrelated (HR
2.07, 95% CI [1.67-2.55]) to CLL. Among 1 487 patients receiving first-line treatment,
164 (11%) had T2D. Upon first-line treatment, patients with T2D had shorter event-free
survival (HR 1.40, 95% CI [1.07-1.79]) and higher mortality (HR 1.22, 95% CI [0.95-1.56])
when compared with patients with CLL without T2D. Treatment with fludarabine, cyclophosphamide,
and rituximab (FCR) was not associated with a superior survival compared with bendamustine
and rituximab (BR) in patients with T2D (HR 2.58, 95% CI [0.61-10.86]).
Summary/Conclusion: T2D is associated with an inferior prognosis in CLL. CLL patients
with T2D had a lower probability of receiving CLL-treatment yet higher mortality.
While the latter may be related to surveillance bias, further investigation in to
causes is required. Though not statistically significant, analyses suggest that among
patients with CLL and T2D, BCR treatment might be superior to FCR.
P643: RISK FACTORS FOR UNFAVORABLE OUTCOME OF HOSPITALIZED PANTIENTS WITH CONCURENT
CHRONIC LYMPHOCYTIC LEUKEMIA AND COVID-19 – EXPERIENCE OF THREE SERBIAN UNIVERSITY
CENTERS
Z. Cvetkovic1,2,*, O. Markovic2,3, K. Markovic4, A. Novkovic1, A. Divac3, N. Vukosavljevic4,
M. Tanasijevic1, T. Bibic1
1Department of Hematology, Clinical Hospital Center Zemun; 2University of Belgrade,
Medical Faculty; 3Department of Hematology, Clinica Hospital Center Bezanijska kosa;
4Department of Hematology, Clinical Hospital Center Zvezdara, Belgrade, Serbia
Background: Vulnerability of patients (pts) with chronic lymphocytic leukemia (CLL)
and their susceptibility to Covid-19 infection is documented in several studies with
reported case fatality rates (CFRs) up to 40%, but there is still paucity of data
on identifying risk factors of their adverse outcome.
Aims: To evaluate demographic, patient-related, CLL-related and Covid-19 related risk
factors in hospitalized pts with concurrent CLL and Covid-19.
Methods: Total of 81 CLL pts were identified in medical records of three University
centers in Belgrade: Clinical Hospital Center (CHC) Zemun, CHC Bezanijska kosa and
CHC Zvezdara dedicated to treatment of Covid-19 pts during pandemic (from 15 March
2020 to 31 December 2021).
Results: For all 81 pts CFR was 32.1%. Age (median age 68 yrs;range 45-90 yrs) and
sex (apparent male prevalence: 61 male and 20 female; M:F=3.05) had no influence on
outcome. Pts with Charlson comorbidity index >4 (29/81;35.8%) had significantly higher
CFR (38% vs 9.5%, p=0,025). Concerning CLL-directed treatment: 26/81(32.1%) pts were
on active treatment (5 pts were on Bruton tyrosine kinase inhibitor, 21pts receiving
imunochemotherapy), 11/81(13.6%) pts were in remission on previous lines of therapy,
while 44/81(54.3%) pts were treatment naive. CLL treatment history had no impact on
CFR, as well as anemia (Hb<100g/l) that was present in 29/81(35.8%)pts, hipogammaglobulinemia
(21/81;26%pts) and hiperferritinemia>450ng/mL (50/81;61.7%pts). Of evaluated laboratory
parameters, high levels of lactate-dehydrogenase (LDH>2xUNL:6/81;7.4%pts), D-dimer
(>1000ng/mL:36/81;44.4%pts), and C-reactive protein (CRP>100mg/L: 31/81;38.3%pts)
proved to be associated with adverse outcome; p-values 0.002, 0.039 and <0.001, respectively.
According to Covid-19 clinical course, the severe Covid-19 score had 35(43,2%)pts,
and critical 19(23.5%)pts. Covid-19 infection was treated according to current National
guidelines. Corticosteroids were administrated to 81.5% of pts, antiviral agents to
38.3%, IL-6 receptor inhibitor to 11.1%, antiviral monoclonal antibodies to 7.4% and
intravenous immunoglobulin to 19.8% of pts. None of listed therapeutic approaches
had impact on CFRs. Antibiotics were administrated to 43/81 (53.1%) of pts with documented
or highly suspected concomitant bacterial infection (procaltitonin level>0.5ng/mL
and/or chest X-Ray image corresponding to bacterial pneumonia), and the bacterial
coinfection had adverse impact on CFR (51.2% vs.10.2%; p<0.001). Significantly higher
mortality was documented in pts who needed supplemental oxygen (58/81;71%) (CFR 43.1
vs.4.3%; p<0.001), and intensive care unit (ICU) admission (25/81-30.9%; 19/25 needed
mechanical ventilation) (CFR 88% vs.7.1%;p<0.001). In multivariate analysis, bacterial
coinfection and ICU admission proved to be the most significant adverse parameters
influencing outcome (p=0.012).
Summary/Conclusion: Our study proved the dismal outcome of CLL pts with concurrent
Covid-19. That could be mainly attributed to the high proportion of bacterial coinfections
reflecting their frailty and sucessibility to both viral and bacterial infections.
P644: THE LIFE EXPECTANCY OF PATIENTS WITH HAIRY CELL LEUKEMIA VERGES UPON THE LIFE
EXPECTANCY OF THE GENERAL POPULATION: A POPULATION-BASED STUDY IN THE NETHERLANDS
A. Dinmohamed1,2,3,*, C. Maas1,2, O. Visser4, J. Doorduijn5, R. Mous6, R. Raymakers6,
A. Kater3, W. Posthuma1,7,8
1Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht;
2Public Health, Erasmus MC, Rotterdam; 3Hematology, Amsterdam UMC, Amsterdam; 4Registration,
Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht; 5Hematology, Erasmus
MC Cancer Institute, Rotterdam; 6Hematology, UMC Utrecht, Utrecht; 7Internal Medicine,
Reinier de Graaf Gasthuis, Delft; 8Hematology, Leiden University Medical Center, Leiden,
Netherlands
Background: At present, no other therapeutic strategy has substantially outperformed
purine nucleoside analogues (PNAs)—introduced around the late 1980s and early 1990s—to
manage hairy cell leukemia (HCL). The most recent population-based study in HCL showed
that 10-year relative survival of HCL patients diagnosed in the Netherlands during
2001-2015 was 97%, 95%, and 83% in the age groups <60, 60-69, and ≥70 years, respectively
(Dinmohamed AG et al. Blood; 2018). Given the improved longevity of HCL patients,
relative survival rates fall short to inform on longevity across the entire patients’
life span.
Aims: Studies estimating the life expectancy of HCL patients are hitherto lacking.
Therefore, we assessed trends in the life expectancy of HCL patients from a historical
and contemporary perspective.
Methods: We selected all HCL patients diagnosed between 1989 and 2019—with follow-up
for survival up to January 1, 2021—from the Netherlands Cancer Registry (N=1,828;
median age, 60 years; interquartile range, 50-70 years; 78% males). We estimated the
loss in expectation of life (LEL)—i.e., the difference between the life expectancy
of patients and an age-, sex-, and period-matched group from the general population—using
flexible parametric relative survival models. The LEL is interpreted as the average
number of life years lost due to an HCL diagnosis. The LEL can vary markedly across
ages because life expectancy is age-dependent. Therefore, the proportional LEL (PLEL)
was estimated. These survival measures were presented by year of diagnosis for four
ages at diagnosis (i.e., 40, 50, 60, and 70 years), stratified by sex.
Results: The life expectancy of HCL patients increased gradually across all ages between
1989-2019, irrespective of sex (Fig A). It is noteworthy that HCL patients diagnosed
in 1990 lost comparatively few life-years due to their diagnosis, ranging from a LEL
of 1.7 to 3.9 life-years lost depending on age and sex (Fig B). Estimates for patients
diagnosed in 2019 ranged from 0.8 to 1.9 life-years lost (Fig B).
Over time, the decrease in LEL was most pronounced for younger patients (Fig B). For
example, a 50-year-old male diagnosed with HCL in 1990, on average, has a LEL of 3.8
years (95% confidence interval [CI]: 2.1-5.5), whereas a male with a HCL diagnosis
in 2019, on average, has a LEL of 1.0 years (95% CI, 0.3-1.7). The corresponding estimates
for a 70-year-old male HCL patient were 2.3 (95% CI, 1.4-3.1) and 1.1 (95% CI, 0.6-1.7),
respectively.
The PLEL estimates also portrays that outcomes in HCL patients improved over time
across all ages (Fig C). Nevertheless, there was a persistent age differential in
the PLEL over time. More specifically, younger patients consistently have more remaining
life-years than older patients, reflected in lower PLEL estimates in younger patients.
Of note, the life expectancy estimates of female patients should be interpreted with
caution by considering the wideness of the 95% CIs due to the comparative rarity of
HCL in females to estimate the life expectancy accurately.
Image:
Summary/Conclusion: The life expectancy of HCL patients verges upon the life expectancy
of the general population. This encouraging finding was already objectified around
the early-1990s when PNA therapy was introduced for HCL management. Thereafter, the
life expectancy gradually increased over time. Novel therapeutic strategies may reduce
the minimal excess mortality encountered in contemporary diagnosed patients, particularly
among the elderly.
P645: THE INFLUENCE OF ACTIVE CARDIAC MONITORING WITH REMOTE CONTROL ON THE SURVIVAL
OF PATIENTS WITH CHRONIC LYMPHOCYTIC LEUKEMIA, RECEIVING IBRUTINIB.
E. Emelina1,*, G. Gendlin1, I. Nikitin1
1Hospital Therapy №2, Pirogov Russian National Research Medical University, Moscow,
Russia
Background: Survival of chronic lymphocytic leukemia (CLL) patients receiving ibrutinib
(Ib) is influenced by many factors, among which cardiovascular disease, both in history
and during treatment with Ib, is of great importance.
Aims: To assess the impact on 5-year overall survival (5-OS) of patients CLL treated
with Ib two options for dynamic monitoring by a cardiologist: standard cardiac monitoring
(SC) and active cardiac monitoring with remote control (ACM).
Methods: We observed in dynamics for 5 years’ period 217 patients with CLL, constantly
receiving therapy with Ib 420 mg/day. In the ACM group (n=89), in addition to the
standard examination, every week or more often using messengers, we did active medical
monitoring of the patient’s symptoms and well-being, assessment of blood pressure
and heart rate, control of cardioprotective medicines taken, correction of the therapy,
calling patients for examination and additional examination. The remaining 128 patients
were examined in dynamics, but did not support remote control, constituting the SC
group. The age of patients in the ACM group and in the SC group did not differ and
was 66.0 (60.0–70.0) years and 66.0 (59.0–74.0) years, respectively (p = 0,39). There
were slightly more men in the SC group (68.8%) than in the active cardiac monitoring
group (53.9%), p = 0.026. The proportion of patients with CLL with the number of pretreatment
lines from 0 to 2 (median -1) in the ACM group was 52.7%, in the SC group it was 58.1%;
with the number of lines from 3 to 12 (median - 4) in the AKM group - 47.3%, in the
SC group - 41.9% (p = 0.53). The AKM and SC groups did not differ in the results of
the Geriatric 8 scale, Charlson index, and echocardiography parameters at visit 1.
In the ACM group, there were more patients with cardiac problems: with arterial hypertension
(AH) (p < 0.0001) and atrial fibrillation (p < 0.0001), receiving anticoagulants (p
< 0.0001), a comparable number of patients with coronary artery disease.
Results: In the ACM group, 70 out of 77 (90.9%) patients with CLL and AH achieved
a stable level of target blood pressure values, in contrast to the SC group - 26 out
of 66 (39.9%), p < 0.0001. Significantly more events requiring cardiac surgery (stenting
of the coronary arteries, installation of a pacemaker) were detected in the ACM group
– 12, versus 0 in the SC group (p = 0.0004). In the ACM group, despite a more pronounced
cardiac comorbidity, 5-OS was significantly better, than in the SC group in both men
(p < 0.0001) and women (p < 0.0001) with CLL and in patients older than 70 years old
(p = 0.0004). 5-OS was also better in the ACM group than in the SC group in patients
with CLL with a median number of previous lines of therapy equal to 1 (p<0.0001) and
in patients with a median number of chemotherapy lines equal to 4 (p<0.0001), in patients
with genetic abnormalities (p=0.004) and pretreated with fludarabine and/or anthracyclines
(p < 0.0001).
Summary/Conclusion: Early detection and correction of cardiovascular complications/events,
achievement of stable target blood pressure values, constant monitoring of cardioprotective
treatment in the ACM group explain the statistically highly significant differences
in 5-OS in patients with CLL who are on constant Ib therapy. Conducting active cardiomonitoring
with remote control makes it possible to achieve higher rates of total 5-OS in patients
with CLL receiving Ib.
P646: EFFECTIVENESS AND SAFETY OF VENETOCLAX IN COMBINATION WITH RITUXIMAB (VENR)
IN R/R CLL PATIENTS WITH OR WITHOUT RISK-ASSOCIATED GENETIC MARKERS – DATA FROM THE
OBSERVATIONAL VERVE STUDY
I. Schwaner1,*, H. Hebart2, C. Losem3, T. Wolff4, K. Famulla5, J. Huelsenbeck5, B.
Schmidt6, T. Nösslinger7, D. Rossi8
1Onkologie Kurfürstendamm, Berlin; 2Kliniken Ostalb, Mutlangen; 3MVZ Onkologie und
Hämatologie, Neuss; 4Onkologie Lerchenfeld, Hamburg; 5AbbVie Deutschland GmbH & Co
KG, Wiesbaden; 6Hämatologisch-Onkologische Gemeinschaftspraxis, München, Germany;
7Medizinische Abteilung für Hämatologie und Onkologie, Hanusch Krankenhaus, Wien,
Austria; 8Institute of Oncology Research, Bellinzona, Switzerland
Background: Treatment with Venetoclax in combination with Rituximab (VenR) has shown
promising efficacy and good tolerability in clinical trials1 However, there is limited
real-world data on VenR in CLL patients with or without genetic markers, known to
be associated with unfavorable outcomes.
Aims: The non-interventional observational study VeRVe aims to investigate safety
and effectiveness of VenR for treatment of CLL under real-world conditions in Germany,
Austria, and Switzerland. In this analysis we focus on safety and effectiveness of
VenR in CLL patients with or without TP53 mut and/or del(17p) as well as IGHV status.
Methods: CLL patients included in this study required therapy and were eligible for
VenR treatment according to local label2. Study documentation is possible at baseline,
weekly during ramp-up, monthly until the end of month 6 and quarterly afterwards up
to a maximum of 3 years. Response assessment can be documented at end of ramp-up,
after 3, 12, 24 and 36 months.
Results: At the time of analysis (November 4th, 2021), 100 patients were enrolled
in the VenR arm of the VeRVe study and received at least one dose of Ven. 86 of them
did also receive at least one dose of rituximab. 28 patients were female, 32 had aberrant
TP53 (TP53 mut and/or del(17p)) and 40 were IGHV unmutated. TP53, del(17p) and IGHV
status was unknown for 20, 18 and 42 patients, respectively. The median age was 72
y in the population with TP53 aberration and 71 y in the population with wild-type
(wt) TP53. Within the TP53 aberrant vs the TP53 wt group 48% vs 67% had received 1,
48% vs 33% had received 2 or more prior lines of therapy. At least 1 comorbidity was
reported for 85% of TP53 aberrant and 81% of TP53 wt patients.
After 12 months of therapy, the best ORR was 88% in the total, 90% in the TP53 aberrant
and 91% in the TP53 wt population. Thereby, CR/CRi was achieved in 54% of TP53 aberrant
and 44% of TP53 wt patients. When subdivided according to IGHV status, the best ORR
was 88% in IGHV mutated and 91% in IGHV unmutated patients. 29% of IGHV mutated and
62% of IGHV unmutated patients reached CR/CRi as best ORR within 12 months. However
IGHV status was not documented for 42 patients.
Estimated PFS-rates after 1 year were 88% in the total population and 86% and 90%
in the TP53 aberrant and TP53 wt population, respectively (figure). Estimated OS-rates
after 1 year were 90% in the total population, 93% in the TP53 aberrant population
and 90% in the TP53 wt population.
After a median observation time of 451 days (range 1-1016), CTCAE grade ≥3 AEs were
reported in 57% (n=57) of all patients, 61% (n=20) in TP53 aberrant patients and 64%
(n=27) in the TP53 wt patients. SAEs were reported in 36% (n=36) of all, 42% (n=14)
in TP53 aberrant and 40% (n=17) in the TP53 wt patients. TLS occurred in 10 patients
(10%) of the overall population. 2 of them where clinical TLS (both in the group TP53
wt).
Image:
Summary/Conclusion: In conclusion, patients with and without TP53 aberrations were
comparable regarding age and burden of comorbidities, but in the group with TP53 aberrations
slightly more patients had a high number of prior therapy lines. However, both populations
responded to VenR treatment with comparably high best ORR, estimated PFS-rates and
estimated OS-rates. This was also the case for IGHV mutated vs unmutated patients,
however patient number was low for this analysis, because the IGHV status was often
not documented in this real-world study. VenR treatment was well tolerated in the
overall population and all subgroups and no new safety signals were detected.
P647: SAFETY AND EFFECTIVENESS OF VENETOCLAX MONOTHERAPY IN R/R CLL PATIENTS WITH
OR WITHOUT RISK-ASSOCIATED GENETIC MARKERS – DATA FROM THE OBSERVATIONAL VERVE STUDY
I. Schwaner1,*, H. Hebart2, C. Losem3, T. Wolff4, K. Famulla5, J. Huelsenbeck5, B.
Schmidt6, T. Nösslinger7, D. Rossi8
1Onkologie Kurfürstendamm, Berlin; 2Kliniken Ostalb, Mutlangen; 3MVZ Onkologie und
Hämatologie, Neuss; 4Onkologie Lerchenfeld, Hamburg; 5AbbVie Deutschland GmbH & Co
KG, Wiesbaden; 6Hämatologisch-Onkologische Gemeinschaftspraxis, München, Germany;
7Medizinische Abteilung für Hämatologie und Onkologie, Hanusch Krankenhaus, Wien,
Austria; 8Institute of Oncology Research, Bellinzona, Switzerland
Background: In chronic lymphocytic leukemia (CLL), the association of genetic markers
such as del(17p), TP53 and IGHV status with disease prognosis is well established.
In clinical trials, treatment with Venetoclax (Ven) monotherapy has shown promising
efficacy and good tolerability in all subgroups1,2. However, there is limited real-world
data on Ven usage in specific genetic subgroups.
Aims: The non-interventional observational study VeRVe aims to investigate safety
and effectiveness of Ven for treatment of CLL in a real world setting in Germany,
Austria, and Switzerland. Here, we present VeRVe data focusing on Ven monotherapy
in CLL patients with or without the genetic risk factors TP53 mut and/or del(17p)
as well as IGHV status.
Methods: CLL patients included in this study required therapy and were eligible for
Ven monotherapy treatment according to local label3. Study documentation is possible
at baseline, weekly during ramp-up, monthly until the end of month 6 and quarterly
afterwards up to a maximum of 2 years. Response assessment can be documented at end
of ramp-up, after 3, 12 and 24 months.
Results: On Nov 4th, 2021, 77 patients were enrolled in the VeRVe study and received
at least one dose of Ven monotherapy, 29 of which were female. 40 of them had TP53
aberrations (TP53 mut and/or del(17p)) and 27 were IGHV unmutated. For 14 patients
TP53 mut status, for 10 patients del(17p) and for 40 patients IGHV status was not
documented. At start of therapy, the median age of patients with TP53 aberrations
was 73 years and 71 years for patients with wildtype (wt) TP53. In the population
with TP53 aberrations, 40% had received 1, 28% 2 and 32% 3 or more prior therapies.
In the population with wild-type (wt) TP53, 12% had received 1, 40% 2 and 48% 3 or
more prior therapies. 80% of patients with TP53 aberration and 72% of patients with
wt TP53 had at least 1 comorbidity.
After 12 months of therapy, the best ORR (CR/CRi) was 74% (34%) in the total population.
Thereby, the best ORR (CR/CRi) was 76% (43%) in patients with TP53 aberrations and
69% (27%) in patients without TP53 aberrations. When subdivided according to IGHV
status, the best ORR (CR/CRi) was 83% (33%) in IGHV mutated and 85% (43%) in IGHV
unmutated patients (see figure).
Moreover, estimated PFS-rate after 1 year was at 80% in the total population. Thereby,
estimated PFS-rate was at 78% in patients with TP53 aberrations and at 83% in patients
with wt TP53. Estimated OS-rate after 1 year was 82% in the total population, 77%
in patients with TP53 aberration and 84% in patients with wt TP53.
With a median observation time of 294 days (range 0-1130), CTCAE grade AEs ≥3 were
reported in 56%(n=43) of the total population, 53% (n=21) in the TP53 aberrant and
64% (n=16) in the TP53 wt population. SAEs were reported in 37% (n=39) of the total
population, 45% (n=18) in the TP53 aberrant and 32% (n=8) in the TP53 wt population.
Overall, 5 patients had a TLS with 3 of them being clinical TLS. No new safety signals
were detected.
Image:
Summary/Conclusion: In summary, patients with TP53 aberration were slightly more pre-treated
and suffered from co-morbidities more frequently than patients with wt TP53. Nevertheless,
best ORR, estimated PFS-rates and OS-rates were comparably high in both populations.
IGHV status was often not determined in this real-world study. For the available data,
best ORR in IGHV mutated and IGHV unmutated patients were comparable. Surprisingly,
CR/CRi rates were highest in patients with TP53 aberration or IGHV unmutated patients.
Treatment was well tolerated in the overall population and all subgroups.
P648: T CELL PHENOTYPIC CHANGES IN PARTICIPANTS OF A MULTIPHASE OPTIMIZATION STRATEGY
(MOST) BEHAVIORAL INTERVENTION OF HEALTHY DIET AND EXERCISE IN PATIENTS (PTS) WITH
CHRONIC LYMPHOCYTIC LEUKEMIA (CLL)
M. Gordon1,*, J. Crane2, C. Y. Lee3, S. Fares3, K. Basen-Engquist3, M. Markofski4,
A. Ferrajoli5, E. LaVoy4
1Cancer Medicine, University of Texas MD Anderson Cancer Center; 2Biology, University
of Houston; 3Behavioral Science, University of Texas MD Anderson Cancer Center; 4Health
and Human Performance, University of Houston; 5Leukemia, University of Texas MD Anderson
Cancer Center, Houston, United States of America
Background: Clinical outcomes in pts with CLL are influenced by disease biology, host
immune response and comorbidities (Roessner et al, Leukemia, 2020 and Rotbain et al,
Leukemia, 2021). Intensive physical exercise can modulate T cell function and slow
leukemic progression (Sitlinger et al, Blood Adv., 2020). Here we report baseline
and 4 month follow up of T cell phenotype in CLL pts after participation in a MOST
exercise program.
Aims: Assess the association of comorbidities and T cell subsets in pts with CLL participating
in an organized exercise program.
Methods: Pts with CLL treated at MD Anderson were randomized to one of 16 combinations
of 5 evidence-based behavioral interventions. Pts with baseline and follow up data
were included. T cell phenotype was identified by multicolor flow cytometry assessing
CD3, CD4, CD8, HLADR and PD1. Difference in baseline and pre/post intervention T cell
subset was assessed by t-test and Mann-Whitney. A p-value <.1 was considered significant.
Results: Median age was 63 years in the 24 pts with CLL. TP53 aberrancy was present
in 8%, unmutated IGHV in 46% and 63% had moderate-severe comorbidities (CLL comorbidity
index [CI] score, 1-2). Median prior lines of therapy were 1, 85% were receiving a
BTK inhibitor (ibrutinib n=7, acalabrutinib n=2 and zanubrutinib n=2); 50% were untreated.
Baseline T cell phenotype showed that a CLL-CI score of 1-2, compared to 0 was associated
with lower HLADR+CD4+ T cells (p=.07). CLL-CI score of 1 vs 0 was associated with
high PD1+CD8+ T cells (p=.03) and PD1+CD4+ T cells (p=0.06; Fig 1). By individual
comorbidity category only upper gastrointestinal comorbidities (n=6) were associated
with T cell phenotype, lower HLADR+CD8+ T cells (p=.04). Pts ≥65 years had lower HLADR+CD4+
(p=.04) and HLADR+PD1+CD4+ (p=.09) T cell populations. Advanced Rai stage (2-4) was
associated with higher CD8+ T cells (p=.03). Treatment, IGHV and Beta 2 microglobulin
were not associated with T cell phenotype.
T cell phenotype at 4 months showed that a CLL-CI score of 1-2 vs 0 was associated
with lower HLADR+CD4+ T cells (p=.06). Both PD1+CD4+ and PD1+CD8+ T cells were higher
in pts with a CLL-CI score of 1 vs 0 (p=.09 for both). Pts with vascular comorbidities
had lower CD8+ (p=.09) and higher HLADR+PD1+CD4+ (p=.09) T cells. Endocrine comorbidities
were associated with lower HLADR+CD4+ (p=.08) and lower HLADR+PD1+CD4+ (p=.09) T cells.
Advanced Rai stage was associate with increased HLADR+CDC8+ T cells (p=.02). Treatment
and age did not significantly impact T cell phenotype.
Image:
Summary/Conclusion: Comorbidities are associated with shorter survival and more rapid
progression of CLL (Gordon et al, CCR, 2021 and Rotbain et al, Blood adv., 2022).
But the mechanism(s) underlying these observations are not well described. High HLADR+PD1+CD4+
T cells are associated with CLL progression (Elston et al, BJH, 2020). We hypothesized
that comorbidities, measured by the CLL-CI, could influence T cell function and thus
lead to CLL progression. CLL-CI score was associated T cell phenotype in this study.
Pts with comorbidities had higher PD1 expression at baseline. Interestingly, after
participation in the exercise intervention, pts with vascular disease had higher HLADR+PD1+CD4+
T cells while those with endocrine comorbidities had lower HLADR+PD1+CD4+ T cells.
This suggest that pts with obesity and diabetes may particularly benefit from participation
in behavioral interventions by lowering the % of HLADR+PD1+CD4+ T cells.
P649: A FIRST-IN-HUMAN PHASE 1 TRIAL OF NX-2127, A FIRST-IN-CLASS ORAL BTK DEGRADER
WITH IMID-LIKE ACTIVITY, IN PATIENTS WITH RELAPSED AND REFRACTORY B-CELL MALIGNANCIES
A. Mato1,*, A. Danilov2, M. R. Patel3, M. Tees4, I. Flinn5, W. Ai6, K. Patel7, M.
Wang8, S. M. O’Brien9, S. Nandakumar10, M. Tan10, E. Meredith10, M. Gessner10, S.
Y. Kim10, A. Wiestner11, W. G. Wierda8
1Memorial Sloan Kettering Cancer Center, New York; 2City of Hope National Medical
Center, Duarte; 3Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota;
4Colorado Blood Cancer Institute, Denver; 5Sarah Cannon Research Institute and Tennessee
Oncology, Nashville; 6University of California San Francisco Medical Center, San Francisco;
7Swedish Cancer Institute, Seattle; 8MD Anderson Cancer Center, Houston; 9Chao Family
Comprehensive Cancer Center, University of California, Irvine; 10Nurix Therapeutics,
Inc., San Francisco; 11National Heart, Lung, and Blood Institute, National Institutes
of Health, Bethesda, United States of America
Background: Bruton’s tyrosine kinase inhibitors (BTKi) have received regulatory approvals
and are standard of care for patients with chronic lymphocytic leukemia/small lymphocytic
lymphoma (CLL/SLL), mantle cell lymphoma (MCL), marginal zone lymphoma (MZL), and
WaldenstrÖm macroglobulinemia (WM). However, BTKi-resistant disease remains a clinical
challenge with limited options for subsequent therapy.
Immunomodulatory drugs (IMiDs, e.g., lenalidomide) are approved as monotherapy for
follicular lymphoma (FL), MZL, and MCL, in combination with other therapies for diffuse
large B-cell lymphoma (DLBCL) and have shown synergy with BTK-targeted therapy. Dual
activity of BTK protein degradation with IMiD-like activity offers a unique approach
to overcome known resistance to BTKi.
NX-2127 is an oral small molecule that induces BTK degradation via recruitment of
cereblon, an adaptor protein of the E3 ubiquitin ligase complex. NX-2127 has shown
preclinical activity similar to IMiDs by catalyzing the ubiquitination of Ikaros (IKZF1)
and Aiolos (IKZF3), ultimately leading to increased T-cell activation. NX-2127 was
shown to degrade both wild-type (WT) and C481-mutated (ibrutinib-resistant) BTK protein
in vitro. Robust BTK degradation was also shown in non-human primate studies. Further,
NX-2127 demonstrates potent tumor growth inhibition in BTK-dependent mouse xenograft
tumor models expressing either WT or ibrutinib-resistant C481S BTK-mutant protein.
This dual activity of BTK degradation and IMiD-like activity offers a promising treatment
for patients who have failed prior therapy.
Aims: The primary objectives are to evaluate safety and tolerability and to determine
the maximum tolerated dose (Phase 1a), and to evaluate the early clinical activity
of NX-2127 in expansion cohorts (Phase 1b).
Methods: NX-2127-001 is a first-in-human, dose escalation (Phase 1a) and cohort expansion
(Phase 1b) study designed to evaluate the safety, tolerability, and preliminary efficacy
of NX-2127 in adult patients with relapsed/refractory B-cell malignancies with once
daily oral dosing. Dose escalation will proceed using a modified Fibonacci design
with 1 patient per cohort, proceeding to a standard 3 + 3 design based on protocol
specified criteria. There will be up to 5 expansion cohorts in Phase 1b enrolling
patients with CLL/SLL, DLBCL, FL, MCL, MZL, and WM. Key eligibility criteria include
≥2 two prior lines of therapy (≥1 prior for WM); measurable disease; and an Eastern
Cooperative Oncology Group performance status of 0 or 1. Approximately 130 patients
(30 in Phase 1a, 100 in Phase 1b) will be enrolled and treated until disease progression
or unacceptable toxicity.
The primary endpoint are dose-limiting toxicities and maximum tolerated dose (Phase
1a), objective response (Phase 1b), and safety (Phase 1a and Phase 1b) of NX-1607.
Secondary endpoints (Phase 1a and Phase 1b, unless otherwise indicated) include pharmacokinetics,
pharmacodynamics, duration of response, progression-free survival, overall survival
(Phase 1b), safety (Phase 1b), and complete response rate/complete response rate with
incomplete marrow recovery.
Results: The Phase 1a part of this study is currently enrolling in the United States.
Summary/Conclusion: Accrual is ongoing. Clinical trial information: NCT04830137.
P650: A FIRST-IN-HUMAN PHASE 1 TRIAL OF NX-5948, AN ORAL BTK DEGRADER, IN PATIENTS
WITH RELAPSED AND REFRACTORY B-CELL MALIGNANCIES
K. Linton1,*, G. P. Collins2, D. El-Sharkawi3, R. Mous4, F. Forconi5, M. Tan6, S.
Nandakumar6, E. Meredith6, K. L. Jameson6, S. G. Injac6, J. Doorduijn7
1The University of Manchester, Manschester; 2Oxford University Hospitals NHS Trust,
Oxford; 3Royal Marsden NHS Foundation Trust, Sutton, United Kingdom; 4UMC Utrecht
Cancer Center, University Medical Center, Utrecht, Netherlands; 5University Hospital
Southampton NHS Trust, Southhampton, United Kingdom; 6Nurix Therapeutics, Inc., San
Francisco, United States of America; 7Erasmus MC Cancer Institute, Rotterdam, Netherlands
Background: Bruton’s tyrosine kinase (BTK) is a key component of the B-cell receptor
(BCR) signaling pathway and chronic activation of BTK-mediated BCR signaling is a
hallmark of many B-cell malignancies. BTK-targeted therapy has been shown to be safe
and effective in a variety of B-cell lymphomas, including chronic lymphocytic leukemia
(CLL), small lymphocytic lymphoma (SLL), mantle cell lymphoma (MCL), marginal zone
lymphoma (MZL), and Waldenström macroglobulinemia (WM). However, BTK inhibitor (BTKi)
resistant disease remains a clinical challenge with limited options for salvage therapy
and overall poor patient outcomes. BTK degradation may be effective in patients who
have developed resistance to BTKi or in B-cell indications where treatment with BTKi
has been less effective.
NX-5948 is an oral, small molecule that induces BTK degradation through the recruitment
of cereblon, an adaptor protein of the E3 ubiquitin ligase complex, and does not induce
degradation of other cereblon neosubstrates. NX-5948 degrades both wild type BTK and
BTKi resistant mutants (C481) and inhibits ibrutinib-resistant tumor cell line growth
at concentrations where ibrutinib and acalabrutinib are inactive. NX-5948 potently
inhibits tumor growth in xenograft models that contain either wild type BTK or BTKi-resistant
mutations. Further, NX-5948 crosses the blood-brain barrier, thus positioning it as
a therapeutic agent in patients with primary central nervous system lymphoma (PCNSL)
and other lymphomas that have CNS involvement.
Aims: The primary objectives are to evaluate safety and tolerability, determine the
maximum tolerated dose (MTD; Phase 1a only), and the early clinical activity in each
expansion cohort (Phase 1b only) of NX-5948 in patients with relapsed and refractory
B-cell malignancies.
Methods: NX-5948-301 is a first-in-human, dose escalation (Phase 1a) and cohort expansion
(Phase 1b) study designed to evaluate the safety, tolerability, and preliminary efficacy
of NX-5948 in adult patients with relapsed and refractory B-cell malignancies. NX-5948
will be given orally daily and dose escalation will proceed using a standard 3 + 3
design. There will be up to 5 expansion cohorts in Phase 1b composed of patients with
the following tumor types: (a) CLL/SLL with a C481 mutation; (b) CLL/SLL without a
C481 mutation; (c) aggressive subtypes diffuse large B-cell lymphoma (DLBCL) and MCL;
(d) indolent subtypes FL, MZL, and WM; and (e) primary/secondary CNS lymphoma. Other
key eligibility criteria include 2 or more prior lines of therapy (only 1 prior for
WM and PCNSL); measurable disease; and an Eastern Cooperative Oncology Group performance
status of 0 or 1. Up to 130 patients (30 in Phase 1a, 100 in Phase 1b) will be enrolled
and treated until disease progression or unacceptable toxicity.
Results: Enrollment in this study has begun in Europe.
Summary/Conclusion: Accrual is ongoing. Clinical trial information: NCT05131022.
P651: VENETOCLAX-BASED TREATMENT OF PATIENTS WITH RICHTER SYNDROME: OUTCOMES FROM
A MULTICENTER RETROSPECTIVE STUDY
P. Hampel1,*, S. Parikh1, W. Wierda2, A. Ferrajoli2, N. Jain2, J. Burger2, T. Call1,
Y. Wang1, S. Kenderian1, W. Ding1, P. Thompson2
1Hematology, Mayo Clinic, Rochester; 2Leukemia, The University of Texas M.D. Anderson
Cancer Center, Houston, United States of America
Background: Patients (pts) with chronic lymphocytic leukemia (CLL) who develop Richter
Syndrome (RS) have a poor prognosis with median overall survival (OS) often reported
as less than 12 months (Tsimberidou JCO 2006; Rogers BJH 2018), using chemoimmunotherapy
(CIT) regimens typically administered to de novo large cell lymphoma. Venetoclax (ven)
has demonstrated single-agent activity in RS with an overall response rate of 43%
(Davids JCO 2017). Ven combined with DA-EPOCH-R achieved a median OS of 19.6 months
and the highest complete response (CR) rate (50%) reported in pts with RS (Davids
Blood 2022). These results support a synergistic effect when compared to CR rates
of 20% and 0% with EPOCH-R and ven monotherapy, respectively.
Aims: To evaluate the outcomes of pts with RS treated with ven-based treatment, outside
clinical trials, including novel-novel combinations and CIT combinations.
Methods: We analyzed pts with RS treated with a ven-based regimen at MDACC (n=37)
or Mayo Clinic (n=18) between 12/2013 and 8/2021. Patient and disease characteristics
from the time of ven-based treatment start were ascertained. Retrospective response
assessment was as per Lugano 2014 guidelines. Toxicity was recorded per iwCLL 2018
guidelines (hematologic toxicity) or CTCAE v5.0 (non-hematologic toxicity). OS and
progression-free survival (PFS) were analyzed using the Kaplan-Meier method, with
and without censoring for allogeneic hematopoietic stem cell transplantation (alloSCT).
No formal statistical comparisons were made between different treatment groups.
Results: Fifty-five pts were identified with a median age of 66 years (range 43-83
years); 31% were female. High-risk CLL disease characteristics were frequently identified:
92% unmutated IGHV, 42% del(17p), 57% TP53 mutation, and 62% complex karyotype. The
median number of prior CLL-directed therapies was 2 (range 0-7), including prior chemotherapy
(42%), prior Bruton tyrosine kinase inhibitor (BTKi; 38%), and prior ven (22%); 62%
of pts were previously untreated for RS.
Median follow-up from the start of ven-based RS treatment was 9 months (mo). The most
common ven-based combination regimens achieved overall response and CR rates as follows:
50%/40% with intensive CIT + ven (n=20), 40%/30% with BTKi + ven +/- CD20 antibody
(n=20), 60%/50% with R-CHOP + ven (n=10). Among the remaining 5 patients, 2 received
ven monotherapy and 3 received varied ven-based combination regimens. Nine pts proceeded
to subsequent alloSCT.
The median PFS for the total cohort when censored at alloSCT was 4.0 mo; PFS uncensored
for alloSCT was 4.4 mo (Figure 1A). Median PFS by treatment group (Figure 1B) was
as follows: 3.7 mo for intensive CIT + ven, 3.9 mo with BTKi + ven +/- CD20 antibody,
not reached with R-CHOP + ven. The median OS for the total cohort was 9 mo. The estimated
median OS by treatment group (Figure 1C) was 8.4 mo with intensive CIT + ven, 10.6
mo with BTKi + ven +/- CD20 antibody and not reached with R-CHOP + ven.
Grade 3-4 neutropenia and thrombocytopenia were more common with intensive CIT + ven
(71%; 69%) and R-CHOP + ven (77%; 67%) compared to BTKi + ven +/- CD20 antibody (35%;
25%). Febrile neutropenia occurred in 42%, 33%, and 25% of pts in these three groups,
respectively, with 37%, 33%, and 30% experiencing a grade 3-4 infection.
Image:
Summary/Conclusion: In the notoriously difficult-to-treat RS patient population, ven-based
combination regimens achieve high response rates, including some with prolonged duration,
particularly with the use of R-CHOP + ven. A prospective clinical trial evaluating
this combination is ongoing (NCT03054896).
P652: MEASURING MINIMAL RESIDUAL DISEASE BEYOND 10-4 THROUGH IGHV LEADER-BASED NEXT
GENERATION SEQUENCING IMPROVES PROGNOSTIC STRATIFICATION IN CHRONIC LYMPHOCYTIC LEUKEMIA
P. Hengeveld1,2,*, M. van der Klift1, P. M. Kolijn1, F. Davi3, F. Kavelaars4, E. de
Jonge5, S. Robrecht6, J. Assmann1, L. van der Straten1,2, M. Ritgen7, P. Westerweel2,
K. Fischer6, V. Goede8, M. Hallek6, M.-D. Levin2, A. Langerak1
1Department of Immunology, Erasmus MC, Rotterdam; 2Department of Internal Medicine,
Albert Schweizer Ziekenhuis, Dordrecht, Netherlands; 3Department of Hematology, Pitié-Salpêtrière
Hospital, Paris, France; 4Department of Hematology; 5Department of Clinical Chemistry,
Erasmus MC, Rotterdam, Netherlands; 6Department I. of Internal Medicine, Center for
Intergrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne; 7Department of Medicine
II, University Hospital of Schleswig Holstein, Kiel; 8Division of Oncogeriatrics,
St Marien Hospital, Cologne, Germany
Background: The sensitivity of conventional techniques for reliable quantification
of minimal/measurable residual disease (MRD) in chronic lymphocytic leukemia (CLL)
is limited to MRD 10-4. Measuring MRD <10-4 could help to further distinguish between
CLL patients with durable remission and those at risk of early relapse.
Aims: To develop an academically sourced IGHV leader-based next-generation sequencing
(NGS) assay for the quantification of MRD in CLL.
Methods: The IGHV leader primer set developed by the EuroClonality-NGS working group
was used to amplify all IGH rearrangements present in an end of treatment (EOT) DNA
pool. The IGH clonal target was amplified in a single-step PCR and subsequent sequencing
was performed on the Illumina MiSeq platform. To calculate MRD depth, NGS output was
annotated through in the ARResT/Interrogate immunoprofiler. Technical validation of
the IGHV leader-based assay was performed on contrived MRD samples, created by serial
dilution of pretreatment CLL DNA pooled PBMC DNA from healthy donors. Clinical validation
of the IGHV leader-based assay was performed on EOT samples from the CLL11 trial (NCT01010061).
Results: In 176/183 (96%) measurements on contrived samples, the CLL-specific rearrangement
was detected. The limit of detection and limit of quantitation were estimated at 3.4
[95% CI 1.9-16.0] and 3.8 [95% CI 1.7-8.2] malignant cells per assay, respectivelly.
The limit of blank was found to be 0. Linearity was established in the MRD 10-2-10-5
range (r=0.94 [95%CI 0.91-0.96]). Of note, when provided with sufficient DNA input,
MRD could even be detected down to MRD 10-6. There was high inter-assay concordance
between measurements of the IGHV leader-based NGS assay and allele-specific oligonucleotide
quantitative PCR (r=0.92, [95%CI 0.86-0.96]) and droplet digital PCR (r=0.93, [95%CI
0.88-0.96]).
In a cohort of 67 patients from the CLL11 trial, using 5μg DNA input and MRD 10-5
as a cut-off, undetectable MRD (uMRD) was associated with superior progression-free
survival (PFS) and time to next treatment. Importantly, deeper MRD measurement allowed
for additional stratification of patients with MRD <10-4 but ≥10-5. Patients with
MRD in this range had a significantly shorter PFS, compared to patients with uMRD
<10-5, but significantly longer, compared to patients with MRD ≥10-4 (median PFS:
≥10-4, 10.4 months; <10-4 but ≥10-5, 27.5 months; uMRD <10-5, NR, 4-year PFS rate:
≥10-4, 66.7%; <10-4 but ≥10-5, 25.0%; uMRD <10-5, 7.2%, P<0.001) (Figure 1).
Image:
Summary/Conclusion: We here present an academically-developed, IGHV leader-based NGS
assay for the detection and quantification of MRD in CLL beyond MRD 10-4. The assay
has high sensitivity and is quantitative and linear up to MRD 10-5 when using 5μg
DNA input. Measurement to MRD 10-6 is feasible, conditional on sufficient DNA input.
The deeper MRD measurements enabled by the IGHV leader-based NGS assay resulted in
improved stratification of CLL patients following treatment.
P653: OUTCOMES FOLLOWING TREATMENT WITH A COVALENT BTK AND BCL2 INHIBITOR AMONG PATIENTS
WITH CHRONIC LYMPHOCYTIC LEUKEMIA (CLL)/SMALL LYMPHOCYTIC LYMPHOMA (SLL): A REAL-WORLD
STUDY OF A LARGE U.S. DATABASE
T. A. Eyre1,*, L. Hess2, T. Sugihara3, R. A. Walgren2, P. B. Abada2, H. Konig2, J.
Pagel4, L. E. Roeker5, A. Mato5
1Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom; 2Eli Lilly
and Company, Indianapolis; 3Syneos Health, Austin; 4Loxo Oncology, Stamford; 5Memorial
Sloan Kettering Cancer Center, New York, United States of America
Background: Covalent Bruton tyrosine kinase inhibitors (cBTKi) and B-cell lymphoma
2 inhibitors (BCL2i) have improved the outcomes for patients with CLL/SLL. Although
these agents are highly effective, they are not curative, and over time patients will
require additional therapy. Real-world data of patient outcomes post cBTKi and BCL2i
exposure in CLL/SLL are limited to small retrospective studies, and the optimal treatment
in this setting is unknown.
Aims: This study was designed to address this data gap by evaluating the characteristics
and clinical outcomes about patients with CLL/SLL after treatment with both cBTKi
and BCL2i.
Methods: Eligible patients were adult patients in the Flatiron Health Electronic Health
Record (EHR)-derived de-identified database diagnosed with CLL/SLL between December
2011 and October[LMH1] 2020. Follow-up data were available through October 2021 at
the time of analysis. Patients were required to have at least one record of receiving
both a cBTKi and a BCL2i. Time-to-event analyses using Kaplan-Meier method included
duration of therapy, time to next treatment discontinuation, transformation or death,
and overall survival (OS) from the time of cBTKi/BCL2i discontinuation.
Results: 339 patients (median age 66 years, interquartile range 59, 73; male 69.6%)
met eligibility criteria. Most patients received cBTKi and BCL2i as early lines of
therapy: 40.4% received cBTKi in first line and BCL2i in second line; an additional
21.8% received cBTKi in the second line and BCL2i in third line. Transformation was
recorded among 24 (7.1%) patients during the study period. Of all eligible patients,
215 had discontinued both cBTKi and BCL2i and could be evaluated for post-cBTKi/BCL2i
outcomes. A total of 47 patients (21.9% of the 215 who discontinued therapy) died
before receiving subsequent therapy, 116/215 (54%) received subsequent therapy, and
the remaining 52/215 (24.2%) were alive without additional treatment after cBTKi/BCL2i
exposure. The median time from discontinuation of cBTKi/BCL2i (whichever agent was
last) to the immediate next treatment discontinuation, transformation, or death was
4.6 months (95% confidence interaval [CI]: 3.0-6.9). Among those who received subsequent
therapy, the most common immediate next line of therapy included additional BCL2i-based
therapy (venetoclax re-treatment with or without other agents, n=71/116; 61.2%) or
anti-CD20 antibody-based therapy (with or without other agents, 73/116 (62.9%). Fewer
patients received PI3K inhibitor-based treatment (n=14/116; 12.1%) or additional cBTKi
therapy (n=19/116; 16.4%) at any time after discontinuation of the initial cBTKi/BCL2i.
As shown in the Figure, for those who received subsequent therapy (n=116), the median
duration of the immediate post-cBTKi/BCL2i therapy was 4.8 months (95% CI: 3.0-7.3).
Among the 215 who discontinued cBTKi/BCL2i therapy, median OS from discontinuation
was 14.1 months (95% CI: 11.7-21.0).
Image:
Summary/Conclusion: Patients with CLL/SLL who have been previously treated with both
a cBTKi and BCL2i experience poor outcomes as observed by time to next treatment discontinuation,
duration of subsequent therapy and overall survival. There remains a need for more
effective therapies for patients with CLL/SLL after progression on cBTKi/BCL2i.
P654: IBRUTINIB PLUS VENETOCLAX IN PATIENTS WITH COMPLEX KARYOTYPE AND CHRONIC LYMPHOCYTIC
LEUKEMIA
A. Petrenko1,2,*, M. Kislova1, E. Dmitrieva1, T. Novikova3, M. Kislitsyna3, T. Obukhova3,
E. Nikitin1,2, V. Ptushkin1,2
1Moscow City Center of Hematology, Botkin hospital; 2Russian Medical Academy of Continuous
Medical Education; 3Karyology laboratory, National Medical Research Center for Hematology,
Moscow, Russia
Background: In the largest study of Baliakas et al. (2019) the presence of at least
5 abnormalities, was associated with dismal clinical outcome, independently of the
somatic hypermutation status and TP53 status. The presence of 3 or 4 aberrations is
defined as clinically relevant in the absence of TP53. Studies by Kittai (2021) and
Al-Sawaf (2020) showed the impact of karyotypic complexity on survival in patients
with chronic lymphocytic leukemia (CLL) treated with ibrutinib or venetoclax. The
complex karyotype (CK) is a topic that is being intensively researched, both in the
aspect of increasing karyotypic complexity stratification and clonal evolution.
Optimal therapy for patients with CLL has not yet been developed. The combination
therapy of ibrutinib and venetoclax was superior to chlorambucil and obinutuzumab
in terms of undetectable minimal residual disease (MRD) responses according to data
from the GLOW trial (Tunir, 2021). The importance of achieving a complete response
with undetectable MRD as the goal of therapy in CLL was proposed (Montserrat, 2005).
Aims: The aim of our study is to evaluate the effectiveness of therapy with ibrutinib
and venetoclax in combination for the patients with CLL and CK.
Methods: This ambilinear observational study included patients with CLL with high
genetic complexity (high-CK), defined as >=5 aberrations or CK (>=3 aberrations) in
combination with a 17p deletion (CK+del17p). The first retrospective cohort included
patients treated with ibrutinib monotherapy (Imono) to progression or intolerable
toxicity since May 2015. The second prospective cohort included patients receiving
ibrutinib in combination with venetoclax (IVen) since July 2019. Venetoclax therapy
was started at the 3rd month of ibrutinib (from the escalation phase). Combination
therapy was continued until a complete response, defined as three consecutive PET-CT-negative
and MRD-negative results 3 months apart. If this criterion was not achieved at 24th
month of therapy, venetoclax was discontinued and ibrutinib continued indefinitely.
Results: Seventy-nine patients are included in the study. Twenty-nine patients in
the first cohort and 50 patients in the second cohort. The characteristic is presented
in Table. At the current follow-up periods, there were no significant differences
in PFS and OS regarding a follow-up period <= 24 months (with the exception of death
from COVID-19, since patients were not observed at parallel time intervals). In the
group of patients treated with Imono, the majority of patients achieved partial remission
or partial remission with lymphocytosis by 12 months. In 21 patients from Iven group,
with a median follow-up of 7.4 months, a complete remission was achieved (72.4%);
of these, 8 had unmeasurable MRD. Four patients did not complete the escalation period.
There was a significant difference in the median MRD response achieved between 3 (log10>10)
and 12 (log10<0,1) months in IVen group (p=0,03). In 2 patient from the IVen group
progression of the disease was noted.
Image:
Summary/Conclusion: Combination therapy with ibrutinib and venetoclax is an effective
oral regimen for high-risk patients with complex karyotype disorders. PFS in both
groups is currently not significantly different, which is obviously due to the short
follow-up period. Patients receiving the IVen regimen achieve a significantly better
response, which paves the way for allogeneic transplantation in these patients.
P655: RACIAL AND SOCIOECONOMIC DISPARITIES AMONG PATIENTS WITH CHRONIC LYMPHOCYTIC
LEUKEMIA IN THE US: ANALYSIS OF SURVEILLANCE, EPIDEMIOLOGY, AND END RESULTS PROGRAM
DATA
A. Kittai1,*, Y. Huang1, J. Fisher1, S. Bhat1, M. Grever1, E. Paskett1, K. Rogers1,
J. Woyach1
1The Ohio State University, Columbus, United States of America
Background: Therapy for chronic lymphocytic leukemia (CLL) has changed dramatically
over the past 20 years. With the cost of new therapies and rapid practice changes,
it is unclear if patients are benefiting equally from this progress. We assessed Surveillance,
Epidemiology, and End Results (SEER) program data to determine how race and socioeconomic
status (SES) affect survival for patients with CLL.
Aims: Aim 1 - Determine the prognostic significance of race on overall survival for
patients with CLL.
Aim 2 - Determine the prognostic significance of socioeconomic status on overall survival
for patients with CLL.
Methods: CLL cases reported to 18 SEER Program registries from 2006 – 2018 were included.
Patient characteristics such as age at diagnosis, sex, year of diagnosis, race, and
SES as determined by rural/urban census tract residence (RUCA), and neighborhood (as
represented by the Yost Index, a composite measure of 7 variables assessing different
aspects of the SES of a census tract) were collected and analyzed. Multivariable cox
regression (MVA) was used to determine adjusted odds of survival. Two separate databases
were utilized, one which included data to 2018, and another which contained SES data
but only had data available to 2016.
Results: 46,605 cases from 2009 – 2018 were identified without SES data. The median
age was 70 years, 60% were male, and there was an even distribution of patients diagnosed
with CLL annually from 2009 – 2018. Of the cases with race reported, 89.9% were white,
7.3% Black, 2.4% Asian/Pacific islander, and 0.3% American Indian/Alaska Native. After
a median follow up of 47 months, the median 3-, 5-, and 10-year overall survival (OS)
was 79.5%, 69.5%, and 48.8%, respectively. MVA showed Black race (HR 1.5, 95% CI 1.4
– 1.6) as the strongest independent prognostic variable for worse OS controlling for
year of diagnosis, suggesting race was a significant factor in OS in the era of modern
therapies.
Using the linked RUCA and Yost tertiles for SES, 47,867 cases of CLL from 2006 – 2016
were analyzed. Like the prior analysis, median age was 70 years, 60% were male, and
there was an even distribution of patients diagnosed with CLL annually from 2006 –
2016. Of the cases with race reported, 90.4% were white, 7.1% Black, 2.3% Asian/Pacific
islander, and 0.3% American Indian/Alaska Native. Per the Yost index, 24.8%, 33.8%,
and 41.4% of patients were in Groups 1, 2, and 3, respectively. Per RUCA categories,
8.9% and 91.1% of patients were residents of rural and urban areas, respectively.
MVA showed American Indian/Alaska Native, and Black race as independent prognostic
variables for worse OS, and Yost group 2 and 3, representing higher SES, were found
to be significant independent prognostic variables for improved OS (Table 1). In this
analysis, race remained an independent variable for worse OS after controlling for
SES.
Image:
Summary/Conclusion: Black race and low SES are prognostic of OS in CLL. Further research
is needed to determine whether this is due to access to therapy, quality of care,
social determinants of heath, or disease biology.
P656: INCIDENT VENOUS THROMBOEMBOLISM (VTE) IN PATIENTS WITH MONOCLONAL B-CELL LYMPHOCYTOSIS
(MBL) AND CHRONIC LYMPHOCYTIC LEUKEMIA (CLL) – A POPULATION-BASED STUDY
A. Koehler1,*, M. Moise1, T. Call1, K. Rabe2, S. Achenbach2, D. Crusan2, K. Bailey2,
T. Petterson2, W. Ding1, S. Kenderian1, J. Leis3, Y. Wang1, E. Muchtar1, P. Hampel1,
S. Schwager1, S. Slager2, N. Kay1, A. Ashrani1, S. Parikh1
1Hematology; 2Biostatistics, Mayo Clinic, Rochester; 3Hematology, Mayo Clinic, Scottsdale,
United States of America
Background: Data on risk of VTE after MBL/CLL diagnosis is limited;it is unclear if
MBL/CLL truly confers increased risk of VTE as seen in other malignancies.
Aims: To determine incidence of VTE after MBL/CLL diagnosis and compare it to age-
and sex-matched VTE incidence in the general population.
Methods: We identified all individuals with newly diagnosed, untreated MBL or CLL
between 1998-2021 within a 27-county region surrounding Rochester, MN. Patient demographics
and CLL-specific characteristics were extracted from the Mayo Clinic CLL Database
and the electronic health record (EHR). Incident VTE after diagnosis of MBL/CLL was
identified by querying a) the Mayo Clinic CLL Database; b) EHR for VTE-specific ICD-9
and ICD-10 diagnosis codes; and c) all radiographic studies performed during longitudinal
care of these patients. Risk of VTE was estimated using Cox proportional hazards model
with CLL/SLL status as a time-dependent variable. The unadjusted cumulative risk of
VTE was estimated using Kaplan-Meier methods in SAS 9.4. Rate of incident VTE among
age- and sex-matched population residing in Olmsted County, MN from 2001-2015 was
pulled from the Rochester Epidemiology Project. Age- and sex-specific VTE risk associated
with MBL/CLL (standardized incidence rate ratio; SIR) was estimated by dividing observed
post-MBL/CLL diagnosis VTE incidence rate by expected VTE incidence rate in the general
population. We performed an exact binomial test in the statistical package R (version
4.1.2) to evaluate whether the proportion of VTE cases after MBL/CLL diagnosis (versus
those occurring in the general population) was consistent with their respective proportion
of person-years at-risk for VTE.
Results: We identified 946 patients with newly diagnosed MBL/CLL, of whom 42 (4%)
reported a prior history of VTE and were excluded from this analysis. Of the 904 evaluable
subjects, 293 had MBL and 611 had CLL. The median age was 69 years (range, 28 - 96
years) and 587 (65%) were male.
After a median follow-up of 6 yrs (range: 1 day-23 yrs), 70 of 904 patients developed
VTE after MBL/CLL diagnosis; 43 (61%) with deep vein thrombosis (DVT), 24 (34%) with
pulmonary embolism (PE), and 3 (4%) with both DVT and PE as the first event. Risk
of VTE was similar in CLL compared to MBL [HR (95% CI) = 0.90 (0.49-1.65)]. The 5-year
and 10-year cumulative risk of VTE was 4.9% and 11.5%, respectively. Forty-seven (68%)
patients had an identifiable provoking factor apart from MBL/CLL including second
active malignancy (n=21), surgery (n=9), recent hospitalization (n=6), travel (n=4),
trauma (n=3), line-associated VTE (n=3), and immobility (n=1).
The age-adjusted VTE incidence rates for females and males with newly diagnosed MBL/CLL
were 1275 and 1228 per 100,000 person-years, respectively. In contrast, the age-adjusted
VTE incidence rates for females and males residing in Olmsted County, MN were 193
and 218 per 100,000 person-years, respectively. The overall VTE incidence rate in
females and males after MBL/CLL diagnosis was 6.0 (95% CI: 4.0-8.8) and 5.7 (95% CI:
4.1-7.7) higher, respectively, compared to the general population; the risk of VTE
was highest in 18–49-year-old MBL/CLL patients (SIR: 16.2, 95% CI: 3.3-47.6) (Figure
1).
Image:
Summary/Conclusion: In this large population-based cohort study, 1 in 12 patients
with MBL/CLL developed VTE after a median follow-up of 6 yrs. Patients with MBL/CLL
demonstrated a 6-fold increased risk of VTE compared to the age- and sex-matched general
population. Importantly, nearly 3 in 4 MBL/CLL patients had additional provoking factors
at time of VTE, most notably second active malignancy.
P657: REAL-WORLD TREATMENT PATTERNS OF PATIENTS DIAGNOSED WITH CHRONIC LYMPHOCYTIC
LEUKEMIA (CLL) IN THE UNITED STATES (US)
X. Yang1,*, E. Zanardo2, D. Lejeune3, E. De Nigris1, E. Sarpong1, N. Lema2, M. Farooqui1,
F. Laliberté3
1Merck & Co., Inc., Kenilworth; 2Analysis Group, Inc., Boston, United States of America;
3Groupe d’analyse, Ltée, Montréal, Canada
Background: CLL represents the most common type of leukemia among adults in the US,
but current data on the treatment patterns of patients in clinical practice is limited.
Standard of care frontline therapy consists of various types of targeted therapy or
chemoimmunotherapy (CIT), but it is reserved for patients with advanced-stage disease.
Aims: This study aims to describe real-world treatment patterns among patients diagnosed
with CLL in the US, with a focus on ibrutinib therapy.
Methods: A retrospective database analysis was conducted using the Optum Clinformatics
DataMartTM database (01/2007–07/2020). Patients with ≥2 medical encounters with a
diagnosis code for CLL or small lymphocytic lymphoma (SLL) on different dates were
selected (earliest date was defined as the index date). At least 12 months of continuous
enrollment pre-index date (baseline period) and ≥18 years of age as of the index date
were required. Patients with a diagnosis for mantle cell lymphoma or with evidence
of anticancer therapy (antineoplastic, radiation, or cell therapy) during baseline
were excluded. A subset of patients with ≥1 pharmacy claim for ibrutinib was identified.
An adapted algorithm developed from previously published studies was used to identify
lines of therapy (LOTs). Treatment patterns, including duration of therapy (DOT) and
regimens were reported. DOT spanned from LOT initiation up to discontinuation of all
agents in the LOT, a switch to another LOT, or the addition of a new agent. Median
time to the first line (1L) and from 1L to second line (2L) were evaluated using a
Kaplan-Meier analysis (KM) to account for censoring.
Results: Among 23,087 patients with CLL, the median age was 74 years and 43% were
female. Of these, 1,387 were treated with ibrutinib (median age: 75; female: 38%).
Patients had a mean Charlson comorbidity index score of 1.9. Analysis of treatment
patterns (Table 1) showed that 7,192 patients (31%) were treated with ≥1 LOT (mean
± standard deviation [SD] DOT of 1L: 1.6 ± 2.0 years), and 10% of patients were treated
with ≥2 LOT (mean ± SD DOT of 2L: 1.2 ± 1.7 years). Ibrutinib was used by 907 patients
in 1L (DOT in 1L: 1.0 ± 1.1 years) and 617 in subsequent LOTs (DOT in 2L+: 0.9 ± 1.1
years). Median time from index date to 1L initiation was 8.1 years. Of the patients
who used antineoplastics therapies in 1L, a majority used CLL-related ones (79%).
Most treated patients received targeted therapy (37%; rituximab: 19% and ibrutinib:
12%) and CIT (32%; bendamustine + rituximab: 13% and cyclophosphamide + fludarabine
+ rituximab: 5%). In 2L, 16% of patients used ibrutinib, 12% a bendamustine + rituximab
regimen, and 4% chlorambucil. Median time from 1L initiation to 2L initiation was
4.4 years. Proportion of patients treated with CIT tended to diminish with subsequent
LOT, whereas the proportion treated with ibrutinib tended to increase.
Image:
Summary/Conclusion: This real-world long-term data shows that patients receive their
1L many years after a first CLL diagnosis, with a median time to treatment of ~8 years.
Among US patients with CLL, rituximab, bendamustine + rituximab, and ibrutinib were
identified as the most used 1L regimens, and the latter two as the most used 2L regimens.
As novel therapies are increasingly used, and genetic testing becomes more available,
further research will be needed to evaluate the changes in the way CLL is treated
and its effects on clinical outcomes.
P658: COMPARISON OF CHARACTERISTICS AND OUTCOME OF COVID-19 INFECTION IN PATIENTS
WITH LYMPHOPROLIFERATIVE DISEASE AND IN PATIENTS FROM THE GENERAL POPULATION - EXPERIENCE
FROM THREE UNIVERSITY CENTERS
O. Markovic1,2,*, A. Divac2, Z. Cvetkovic1,3, D. Stanisavljevic4, K. Markovic5, I.
Bukurecki2, T. Bibic3, N. Stanisavljevic1,2, J. Bila1,6, M. Zdravkovic1,7
1University of Belgrade, Medical Faculty; 2Department of Hematology, CHC Bezanijska
kosa; 3Department of Hematology, CHC Zemun; 4Institut of Statistics, University of
Belgrade, Medical Faculty; 5Department of Hematology, CHC Zvezdara; 6Clinic of Hematology,
Clinical Center of Serbia; 7Department of Cardiology, CHC Bezanijska kosa, Belgrade,
Serbia
Background: Patients with lymphopproliferative diseases (LPD) and covid-19 have poor
outcome as consequence of inadequate humoral and cellular immunity due to the hematological
disease itself but also due to the administered chemotherapy which further increases
the risk of complications and mortality.
Aims: The aim of this study is to analyze demographic and clinical characteristics,
laboratory parameters, the presence of comorbidities, laboratory parameters, disease
status, as well as outcome of the patients with COVID-19 and lymphoproliferative disease
and compare them with characteristics of covid-19 infection in patients from general
population (GP).
Methods: This is a prospective multicenter observational study conducted in the following
3 University centers in period from 15 March 2020 to 31 October 2021. The study included
hospitalized patients diagnosed with COVID-19 infection: 161 patients with LPD and
162 patients from the GP. Statistical analysis included demographic statistics, the
χ2 test, the Mann-Whitney test, Kaplan-Meier method for analysis of survival and multivariate
logistic regression model for analysis of risk factors for mortality.
Results: In the LPD group, there were 54 patients (33.54%) with chronic lymphocytic
leukemia (CLL), 72 patients (44.72%) with Non-Hodgkin lymphoma/Hodgkin lymphoma (NHL/HL)
and 35 patients (21.74%) with multiple myeloma (MM). Ninety-six (59,63%) patients
were on active treatment and 14(8.7%) patients were newly diagnosed. The LPD and GP
group differed significantly in relation to age (66 vs. 54 years), gender (male: 60.2%
vs. 75.3%), presence of comorbidities (109, 67.7% vs. 81, 50%) patients, covid score
(mild 22.5% vs. 1.9%, moderate 80, 50.3% vs. 121, 74.7%), and severe/critical 44(27.1%)
vs. 38(23.4%) patients. Group of patients with LPD had also significantly lower level
of hemoglobin, lowest value of lymphocytes, platelets, higher level of CRP, ferritin,
D-dimer (on admission and maximal values) and LDH with respect to group of patients
from GP. Mortality rate was higher in LPD group of patients than in GP group (45,
28% vs. 26, 16%) patients. Among the LPD group, the highest mortality rate was observed
in patients with MM (16, 45.71%) patients, followed by CLL (15, 27.9%) patients and
NHL/HL group (14, 19.4%) patients. Independent factors related to survival are high
value of D dimer, anemia (hemoglobin <100g/l) and moderate/critical COVID score in
LPD group, while maximal value of CRP, anemia, leucocytosis and age (>60 years) in
GP group.
Summary/Conclusion: Our study showed significant difference in the characteristics
and outcome in covid-19 between patients with LPD and patients from GP. Patients with
LPD are older, they have significantly higher inflammatory parameters and more frequent
presence of comorbidities compared to patients from GP. Independent factors related
to survival in the LPD group are high values of D dimer, moderate/critical COVID score
and anemia, while maximal values of CRP, anemia and older age are identified in the
GP group.
P659: IBRUTINIB IN OVER-EIGHTIES PATIENTS WITH CHRONIC LYMPHOCYTIC LEUKEMIA: A MULTICENTER
ITALIAN COHORT
G. Reda1, V. Mattiello1,*, A. M. Frustaci2, A. Visentin3, F. R. Mauro4, I. Innocenti5,
M. Gentile6, D. Giannarelli7, A. Noto1, R. Cassin1, L. Laurenti5, A. Tedeschi2
1Hematology Unit, IRCCS Ca’ Granda Ospedale Maggiore Policlinico; 2Department of Hematology,
Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Milano; 3Hematology
and Clinical Immunology unit, Department of Medicine University of Padua, Padova;
4Hematology, Department of Translational and Precision Medicine, ‘Sapienza’ University,
Rome; 5Dipartimento Scienze Radiologiche ed Ematologiche, Divisione di Ematologia
Fondazione Policlinico universitario A Gemelli, Roma; 6Azienda Ospedaliera of Cosenza,
Cosenza; 7Biostatistic Unit, Scientific Directorate, Fondazione Policlinico Universitario
A. Gemelli, IRCCS, Roma, Italy
Background: Ibrutinib is a first-in-class covalent inhibitor of Bruton’s tyrosine
kinase that has changed the treatment paradigm of chronic lymphocytic leukemia (CLL)
in both treatment-naive (TN) and relapsed/refractory (R/R) setting. Clinical trials,
in selected populations showed that the BTK inhibitor is manageble even in the elderly.
Nevertheless, few studies are focused on the role of ibrutinib in unselected patients
aged ≥80 years.
Aims: We aimed to assess the activity and safety of ibrutinib in a cohort of patients
with CLL aged ≥80 years at therapy start. The primary endpoint was safety evaluation.
Key secondary endpoints were overall (ORR), complete (CR) and partial (PR) response
rates according to iwCLL 2018, median progression free survival (PFS) and overall
survival (OS).
Methods: Sixty consecutive patients (age ≥80 years) diagnosed with TN (20 patients)
or R/R CLL (40 patients) who started ibrutinib were enrolled in this multicenter study
from six Italian sites; data were retrospectively analyzed. Pre-existing cardiovascular
risk factors were present in 66.7% of patients; 23.3% had experienced a cardiovascular
event prior ibrutinib initiation. (Table 1)
Results: After a median follow up of 27 months, at least one adverse event (AE) occurred
in 68.3% patients, leading to treatment discontinuation in 23.3%. The most common
grade ≥3 events were infections (23%) and neutropenia (6%). Cardiovascular events
occurred in 31.6% of patients, with an incidence of atrial fibrillation (AF) and arterial
hypertension both increasing over time and reaching 16% at 24 months. Although no
significant increase in incidence of cardiovascular events was noted among patients
with concomitant cardiovascular risk factors or previous events, baseline higher left
atrial diameter at echocardiography was predictive of AF occurrence. OS did not differ
between patients experiencing AF or not. During treatment, two patients experienced
sudden cardiac death. Bleeding was the most frequent AE, occurring in 36.6% of patients,
with a median time to event of 24 months and predominantly in patients assuming concomitant
antiplatelet or anticoagulant drugs. A total of 23 infective events was registered,
mostly in the first 12 months, leading to drug permanent discontinuation in 5 patients.
Of note, the infectious rate was higher in del(17p) CLL (p=0.05). The obtained ORR
was 88.3%, with 21.7% of patients achieving CR and 66.7% PR; median PFS was 51.8 months
(95% CI: 47.4-56.2). No significant difference in PFS was observed comparing TN to
R/R patients (p=0.83), or IGHV mutated and unmutated patients (p=0.45). Furthermore,
no difference emerged comparing PFS data of patients with TP53 dysfunction (del17
and/or TP53 mutation) to patients without TP53 dysfunction (p=0.13). Patients achieving
a response during ibrutinib experienced a prolonged PFS compared to patients achieving
SD (p<0.0001). Drug withholding for more than 7 days due to ibrutinib-related toxicities
affected patients’ outcome, translating in a shorter PFS with a trend to statistical
significance (p=0.07). Median overall survival was 53.2 months (95% CI: 43.3-63.0).
Image:
Summary/Conclusion: A high proportion of patients had an overall response to ibrutinib
and the risk-benefit profile was favourable, providing further evidence for use of
ibrutinib in this subset of elderly and unselected patients. Safety profile remains
consistent with literature data with no emergent adverse events, thus making ibrutinib
an attractive therapeutic possibility even in patients with advanced age and multiple
comorbidities.
P660: SEROLOGIC RESPONSE TO THE SECOND AND THIRD DOSE OF THE SARS-COV-2 VACCINE IN
PATIENTS WITH CHRONIC LYMPHOCYTIC LEUKEMIA: RESULTS OF A PROSPECTIVE, CENTRALIZED,
MULTICENTER STUDY.
F. R. Mauro1,*, D. Giannarelli2, C. Galluzzo3, A. Visentin4, A. M. Frustaci5, P. Sportoletti6,
C. Vitale7, G. Reda8, M. Gentile9, L. Levato10, R. Murru11, D. Armiento12, C. Ielo13,
R. Maglione13, E. Crisanti13, A. Cipiciani14, V. Mattiello15, V. Gianfelici10, L.
Barabino16, R. Amici17, M. Coscia18, A. Tedeschi19, L. Trentin20, S. Baroncelli17
1Department of Translational and Precision Medicine-Sapienza University of Rome, Italy,
Sapienza University; 2Design and Analysis of Clinical Trials Unit, Scientific Directorate,
Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy; 3National Center
for Global Health, Istituto Superiore di Sanità, Rome; 4Hematology and Clinical Immunology
Unit, Department of Medicine University of Padua, Padova, University of Padua, Padova;
5Hematology, ASST Grande Ospedale Metropolitano Niguarda, Milan; 6Institute of Hematology-Centro
di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia; 7Department of
Molecular Biotechnology and Health Sciences, University of Torino and Division of
Hematology, A.O.U. Città della Salute e della Scienza di Torino, Rome; 8Hematology
Unit, Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore, University of
Milan, Milan, Italy., Milan; 9Hematology and Oncology Department, Biotechnology Research
Unit, Cosenza, Italy., Cosenza; 10Department Hematology-Oncology, Azienda Ospedaliera
Pugliese-Ciaccio, Catanzaro; 11Hematology and Stem Cell Transplantation Unit, Ospedale
Oncologico A. Businco, ARNAS “G. Brotzu”, Cagliari; 12Hematology, Stem Cell Transplantation
Unit, University Campus Bio-Medico; 13Hematology, Department of Translational and
Precision Medicine, Sapienza University, Rome; 14Institute of Hematology-Centro di
Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia; 15Hematology Unit,
Fondazione IRCCS Ca’ Granda Ospedale Maggiore, University of Milan, Milan; 16Hematology
and Stem Cell Transplantation Unit, Ospedale Oncologico A. Businco, ARNAS “G. Brotzu”,
Cagliari; 17National Center for Global Health, Istituto Superiore di Sanità, Rome;
18Department of Molecular Biotechnology and Health Sciences, University of Torino
and Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino;
19ASST Grande Ospedale Metropolitano Niguarda, Milan; 20livio.trentin@unipd.it, Hematology
and Clinical Immunology Unit, Department of Medicine, University of Padua, Padova,
Italy
Background: Patients with chronic lymphocytic leukemia (CLL) show high infection-related
morbidity and mortality due to variable degree of humoral and cellular immune deficiency.
High Covid-related mortality and reduced response to the SARS-Cov-2 vaccine have been
reported in this patient population.
Aims: We carried out a prospective multicenter study to define the rate of CLL patients
with an appropriate immune response after the mRNA SARS-CoV2 vaccine (Pfizer-BioNTech;
Moderna).
Methods: Two-hundred patients with CLL received the first dose of the SARS-CoV-2 vaccine
between February and August 2021. Centralized assessment of the anti-SARS-Cov-2 IgG
levels (Sero Index, Kantaro Quantitative SARS-CoV-2 IgG Antibody, RUO-R&D System)
was performed at the Istituto Superiore di Sanità of Rome, Italy. The median follow-up
of this study is 10.7 months (range 1-12.9).
Results: The median age of patients was 70 years, the median IgG level was 635 mg/dl,
61% of patients were IGHV unmutated, and 34% showed TP53 disruption. The majority
of patients, 83.5%, were previously treated. Prior treatment included chemoimmunotherapy
in 20 (10%) patients, ibrutinib–based therapy in 72 (36%; front-line, 21%; advanced
line, 15%), venetoclax-based therapy in 75 (37.5%; front-line, 13.5%; advanced line,
24%). Overall, 135 (77.5%) patients had been previously treated with rituximab, 33
(16.5%) of them within 12 months before vaccination. We assessed the serologic response
after the second dose of the SARS-CoV2 vaccine in 195 patients while five were excluded
from the analysis (positive test before vaccination, 3 patients; lost to the follow-up,
1; Richter syndrome, 1). Adequate levels of anti-SARS-Cov-2 IgG were detected in 76/195
(39%) patients. Age (<70 vs.≥ 70 years; p <0.0001), CIRS value (<6 vs. ≥6; p=0.005),
beta-2 microglobulin (<3.5 vs. ≥ 3.5mg/dl; p=0.04), IgG levels (<550 vs. ≤ 550 mg/dl;
p <0.0001), prior treatment (p=0.0001), number of prior treatments (0 + 1 vs. ≥ 2;
p=0.002) and the time between prior rituximab and vaccination (>12 vs. ≤12 month;
p=0.001) showed a significant impact on the humoral response. In multivariate analysis
only age (OR: 0.92 [95% CI: 0.92-0.97] p=0.0001), IgG levels (OR: 0.28 [95% CI: 0.13-0.58]
p<0.001), and the time between prior rituximab and vaccination (OR: 0.10 [95% CI:
0.03-0.37] p=0.001), revealed a significant and independent impact on response. When
the analysis was restricted to patients who received targeted therapy, in addition
to the younger age (OR: 0.96 [95% CI: 0.92-0.99] p=0.04), higher IgG levels at baseline
(OR: 0.31 [95% CI: 0.12-0.79] p=0.014), longer time between the start of ibrutinib
or venetoclax-based therapy and vaccination (<18 vs.≥18 months; OR: 0.17 [95% CI:
0.06-0.44], p <0.0001) showed a favorable and independent impact on response. Ninety-three%
(182/195) of patients received a third dose of the vaccine. A significant increase
in the rate of serologic responses, 51.5% (85/165 evaluated patients, p=0.019), was
observed after the booster dose. Moreover, a response was detected in 25% (26/103
evaluated patients) of previously seronegative patients.
Summary/Conclusion: In this prospective, multicenter, centralized study, we recorded
an effective immune response to the SARS-CoV-2 vaccine in about a third of patients
with CLL. Younger age, higher IgG levels, no prior treatment, or stable disease after
targeted therapy that suggest preserved immunocompetence were associated with a greater
likelihood of achieving an effective immune response. A booster dose of the SARS-CoV-2
vaccine proved beneficial also in previously seronegative patients.
P661: POPULATION-WIDE PATTERNS OF CARE IN CHRONIC LYMPHOCYTIC LEUKEMIA IN AUSTRALIA:
AN ANALYSIS OF THE PHARMACEUTICAL BENEFITS SCHEME DATASET
C. Tam1,*, F.-L. Zhao2, R. Gauba2, K. Yang3, S. Azmi3, S. C. Li4
1Peter MacCallum Cancer Centre, St. Vincent’s Hospital, and University of Melbourne,
Melbourne; 2BeiGene AUS PTY Ltd., Sydney, Australia; 3BeiGene USA, Inc., San Mateo,
United States of America; 4University of Newcastle, Callaghan, Australia
Background: The treatment landscape in patients with chronic lymphocytic leukemia
(CLL) is changing with the approvals of Bruton’s tyrosine kinase inhibitors (BTKis)
in Australia.
Aims: We sought to understand the practice impact of the introduction of publicly
funded novel agents for the treatment of CLL. The objective of this study was to describe
the evolving treatment patterns of Australian patients with CLL over the last 10 years
using population-wide prescription records.
Methods: Patients who initiated a treatment for CLL between 01/01/2011 and 07/31/2021
were extracted from the Services Australia 10% Pharmaceuticals Benefits Scheme (PBS)
dataset. This dataset includes the dispensing records for 10% of the Australian population
and captures all publicly funded treatments in Australia. The index date was defined
as the commencement of any drug for the treatment of CLL. First-line (1L) therapy
was defined as the first treatments prescribed for CLL. A patient was defined as relapsed/refractory
(R/R) if they had commenced a drug which was in a different therapeutic category,
or if they re-started the same regimen after a gap of more than 180 days. Descriptive
analyses were conducted to examine the use of treatment regimens for the overall 10-year
population by line of therapy. Analyses by calendar year were also performed to assess
changes in treatment patterns.
Results: Overall, 803 patients with CLL were identified. The majority of patients
were male (65%) and age > 60 years (77%), with most being aged 70-79 years (33% of
total). Many patients were receiving comedications at baseline, including antihypertensives
(47%), antipsychotics or antidepressants (17%), and/or anticoagulants (13%). In the
overall population (2011-2021), the majority of patients had received 1L treatment
with fludarabine-cyclophosphamide-rituximab (FCR, 49%), chlorambucil ± CD20 (27%),
or CD20 monotherapy (17%). The most commonly used regimens in R/R patients at any
subsequent episode of treatment included CD20 monotherapy (56%), BTKi (41%) or FCR
(33%). A trend in adoption of novel agents was observed throughout the years following
their PBS listing. Analysis by calendar year showed that from 2011 to 2020 use of
FCR in 1L decreased from 78% to 10%; and use of BTKis in R/R increased from 0% to
62%.
Summary/Conclusion: CLL treatment patterns have significantly changed in Australia
since the introduction of the BTKis (e.g., ibrutinib, acalabrutinib). The use of FCR
in 1L CLL has decreased and use of BTKis in R/R patients has increased.
P662: PATIENT-REPORTED OUTCOMES FROM A PHASE 3 RANDOMIZED STUDY OF ZANUBRUTINIB VERSUS
BENDAMUSTINE PLUS RITUXIMAB (BR) IN PATIENTS WITH TREATMENT-NAÏVE (TN) CLL/SLL
P. Ghia1,*, G. Barnes2, K. Yang2, C. Tam3,4,5,6, P. Hillmen7, T. Robak8, J. Brown9,
B. Kahl10, T. Tian2, A. Szeto2, J. Paik2, M. Shadman11,12
1Università Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele, Milano, Italy;
2BeiGene USA, Inc., San Mateo, United States of America; 3Peter MacCallum Cancer Centre,
Melbourne; 4University of Melbourne, Parkville; 5St Vincent’s Hospital Melbourne,
Fitzroy; 6Royal Melbourne Hospital, Parkville, Australia; 7St James’s University Hospital,
Leeds, United Kingdom; 8Medical University of Lodz, Lodz, Poland; 9Dana-Farber Cancer
Institute, Boston; 10Washington University School of Medicine, St. Louis; 11Fred Hutchinson
Cancer Research Center; 12Medicine, University of Washington, Seattle, United States
of America
Background: Zanubrutinib is an oral, highly selective, next-generation Bruton tyrosine
kinase (BTK) inhibitor. In cohort 1 of the phase 3 SEQUOIA trial (BGB-3111-304; NCT03336333),
efficacy and safety of zanubrutinib and BR were compared in adult patients (pts) with
TN chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) without del(17p).
Aims: Here, we report the health-related quality of life (HRQoL) outcomes from an
interim analysis of the SEQUOIA trial.
Methods: Patient-reported outcomes (PROs) were secondary endpoints and assessed using
the EORTC QLQ-C30 and EQ-5D-5L VAS. Patients completed these questionnaires at baseline,
every 12 weeks for 96 weeks, and then every 24 weeks until disease progression, death,
or withdrawal from study. The PRO endpoints included global health status (GHS), physical
and role functions, and symptoms of fatigue, pain, diarrhea, and nausea/vomiting,
measured by QLQ-C30, with critical clinical cycles of Weeks 12 and 24. Descriptive
analyses were performed on all questionnaire responses, and a mixed model for repeated
measures was performed on the PRO endpoints at Weeks 12 and 24.
Results: Baseline demographics and disease characteristics between the zanubrutinib
(n=241) and BR (n=238) arms were similar. Across all pts, adjusted completion rates
for PROs (# of pts who completed the questionnaire at each cycle divided by # of pts
expected to complete the questionnaires) were high (~80%) at Weeks 12 and 24. Compared
with pts who received BR, pts treated with zanubrutinib experienced greater improvements
in HRQoL at Weeks 12 and 24 as reported on the QLQ-C30 (Table). By Week 24, significant
improvements were observed with zanubrutinib vs BR in GHS, physical functioning, role
functioning as well as greater reductions in diarrhea, fatigue, and nausea/vomiting.
Per EQ-5D-5L VAS, comparable improvements from baseline between zanubrutinib and BR
in the health status were observed at Weeks 12 (4.3 vs 3.5) and 24 (4.5 vs 4.9), respectively.
Image:
Summary/Conclusion: In the SEQUOIA trial, zanubrutinib was associated with significant
improvements in HRQoL in pts with TN CLL/SLL without del(17p), as indicated by the
PRO endpoints of the GHS, physical and role functions, and greater reductions in symptoms
of fatigue, diarrhea and nausea/vomiting compared with BR.
P663: HEALTH-RELATED QUALITY OF LIFE OUTCOMES ASSOCIATED WITH ZANUBRUTINIB VS IBRUTINIB
MONOTHERAPY IN PATIENTS WITH RELAPSED/REFRACTORY (RR) CLL/SLL: RESULTS FROM THE RANDOMIZED
PHASE 3 ALPINE TRIAL
P. Hillmen1,*, J. Brown2, N. Lamanna3, S. O’Brien4, C. Tam5,6,7,8, L. Qiu9, K. Yang10,
G. Barnes10, K. Wu10, T. Salmi11, B. Eichhorst12
1St James’s University Hospital, Leeds, United Kingdom; 2Medical Oncology, Dana-Farber
Cancer Institute, Boston; 3Herbert Irving Comprehensive Cancer Center, Columbia University,
New York; 4Chao Family Comprehensive Cancer Center, University of California, Irvine,
United States of America; 5Peter MacCallum Cancer Centre, Melbourne; 6University of
Melbourne, Parkville; 7St Vincent’s Hospital Melbourne, Fitzroy; 8Royal Melbourne
Hospital, Parkville, Australia; 9Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin, China; 10BeiGene USA, Inc., San Mateo, United States of
America; 11BeiGene Switzerland GmbH, Basel, Switzerland; 12Department of Internal
Medicine, University of Cologne, Center for Integrated Oncology Aachen, Cologne, Germany
Background: In the phase 3 ALPINE trial (BGB-3111-305; NCT03734016), efficacy and
safety of zanubrutinib, a highly selective, next-generation Bruton tyrosine kinase
inhibitor (BTKi), were compared with the first-generation BTKi, ibrutinib, in adult
patients with RR chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL).
Aims: This abstract, based on the interim analysis of the ALPINE trial, describes
the effects of zanubrutinib monotherapy and ibrutinib monotherapy on the health-related
quality of life (HRQoL).
Methods: Health-related quality of life was examined by patient-reported outcomes
(PROs) measures assessed by EORTC QLQ-C30 and EQ-5D-5L at baseline, Cycle 1, and then
every 3rd cycle until end of treatment. Key PRO endpoints included global health status
(GHS), physical and role functions, and fatigue, pain, diarrhea, and nausea/vomiting.
Descriptive analysis on all the scales was conducted as was a mixed model repeated-measure
(MMRM) analysis of the longitudinal QLQ-C30 data. Data presented are from key cycles
(7 and 13), corresponding to 6 and 12 months of treatment, respectively.
Results: In the intent-to-treat population (N=652; zanubrutinib, n=327; ibrutinib,
n=325), adjusted completion rates were high (>85%) in both arms at Cycles 7 and 13.
On the QLQ-C30, estimated mean treatment differences and 95% CI in key PRO endpoints
demonstrated treatment differences, in favor of zanubrutinib, in GHS, physical functioning,
and fatigue in Cycle 7, and diarrhea in Cycle 13 (Table). Mean change from baseline
(SD) in EQ-5D-5L VAS showed consistently more improvement with zanubrutinib compared
with ibrutinib at both Cycle 7 (8.4 [18.2] vs 4.0 [16.6]) and Cycle 13 (6.8 [18.8]
vs 5.2 [17.5]).
Image:
Summary/Conclusion: In the ALPINE trial, patients with RR CLL/SLL who received zanubrutinib
monotherapy reported improvements in key HRQoL endpoints compared with patients who
received ibrutinib monotherapy.
P664: GRADING QUALITY OF EVIDENCE AND STRENGTH OF RECOMMENDATIONS FOR THE TREATMENT
OF RELAPSED/REFRACTORY CLL.
S. Molica1,*, C. Patti2, P. Sportoletti3, A. Chiarenza4, A. M. Giordano5, F. Chiurazzi6,
N. Di Renzo7, P. Musto8, F. Pane9, F. Di Raimondo4, L. Trentin10, F. R. Mauro11, D.
Giannarelli12
1Department Hematology, Hull University Teaching Hospitals NHS Trust, Hull, UK, Hall,
United Kingdom; 2hematology, Ospedale Cervello, Palermo; 3Hematology Unit, University
Perugia, Perugia; 4Hematology Unit, Ferrarotto Hospital, Catania; 55Department of
Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of
Bari, Bari; 6Department of Clinical Medicine and Surgery, Hematology Unit, Federco
II Medical School Napoli, Napoli; 7Hematology Unit, Presidio Ospedaliero Vito Fazi,
Lecce; 8Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section,
University of Bari, Bari; 9Department of Clinical Medicine and Surgery, Hematology
Unit, Federico II Medical School Napoli, Napoli; 10Hematology and Clinical Immunology
Unit, Department of Medicine, University of Padua, Padua; 11Hematology, Department
of Translational and Precision Medicine, Sapienza University Rome; 12Biostatistic
Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Rome, Italy
Background: Changes in the treatment landscape of CLL have complicated the decision-making
processes, especially in the context of relapsed/refractory (R/R) disease. In this
setting current guidelines do not provide clear information on the sequencing of targeted
agents (TAs).
Aims: With this in mind a series of evidence-based recommendations were produced to
support pts, clinicians, and other health care professionals in their decisions about
the management of R/R CLL.
Methods: A multidisciplinary panel that included specialists in methodology, CLL management,
and hematology, performed a systematic review of the literature using the databases
MEDLINE, EMBASE, and the Cochrane Library, then participated in a virtual consensus
conference held at the end of December 2021. The Grading of Recommendations Assessment,
Development, and Evaluation (GRADE) approach was used to formulate recommendations.
Specifically, the strength of recommendations was determined by the balance between
desirable and undesirable consequences of alternative management strategies, quality
of evidence, variability in values, preferences, and resource used.
Results: Firstly, the panelists prioritized 10 scientific questions in the context
of 4 specific scenarios of R/R CLL: (1) relapse after frontline chemo-immunotherapy
(CIT), (2) progression of disease (PD) while receiving ibrutinib (IBR), (3) progression
after treatment with venetoclax (VEN), and (4) impact of adherence to IBR or VEN on
the clinical outcome.
The following recommendations were produced:
1- A TA should be offered to all pts relapsing after first-line CIT (strong recommendation
based on high-quality evidence). In absence of head-to-head trials comparing a BTKi
vs an anti-BCL2 inhibitor, the panel suggests a patient-oriented choice consisting
of either continuous BTKi (i.e., IBR or acalabrutinib) or time-limited (TL) VEN-based
therapy in both standard or high genomic risk (conditional recommendation based on
moderate-quality evidence).
Remarks: TL VEN-based regimen is more cost-effective than continuous IBR.
2- The panel recommends switching to VEN for those pts who experience PD whilst on
a BTKi (in absence of a valid alternative the recommendation becomes strong even if
the quality evidence is moderate). There is no evidence to recommend the combination
of VEN plus rituximab or an alternative covalent BTKi over VEN single agent (conditional
recommendation based on low certainty in the evidence of effects). Noncovalent BTKi
pirtobrutinib needs validation in phase 3 studies.
3- The panel recommends utilizing a BTKi monotherapy instead of re-challenging VEN
for pts who have progressed after completing VEN-based therapy (conditional recommendation
based on low certainty in the evidence of effects).
4- Finally, panelists agreed that the impact of adherence to IBR or VEN on PFS in
R/R pts is supported by a low quality of evidence (conditional recommendation based
on low certainty in the evidence of effects).
Summary/Conclusion: These evidence-based consensus recommendations strongly support
the use of a TA in R/R CLL patients. The choice between TL and continuous TA is patient-oriented
with the remark that a TL approach is cost-effective within current pricing structures.
A cross-over approach to an alternative TA is suggested in pts progressing on BTKi
or BCL2i.
However, these recommendations are guided by the fundamental principle that optimal
patient care involves ongoing discussions between clinicians and pts, continuously
addressing goals of care and the relative risk-benefit balance of treatment.
P665: CARDIAC ADVERSE EVENTS AND SECOND PRIMARY MALIGNANCIES WITH ACALABRUTINIB IN
CHRONIC LYMPHOCYTIC LEUKEMIA -A SYSTEMATIC REVIEW AND META-ANALYSIS OF RANDOMIZED
CONTROLLED TRIALS
K. Parmar1,*, D. Pawar1, K. Bharathidasan1, G. DelRioPertuz1, L. Tijani1
1Texas Tech University Health Sciences Center, Lubbock, United States of America
Background: Cardiac adverse events were the most common cause of treatment discontinuation
in patients with chronic lymphocytic leukemia (CLL) on ibrutinib. Acalabrutinib is
a new, selective, irreversible Bruton’s tyrosine kinase inhibitor (BTKi) that is approved
for the treatment of CLL.
Patients with CLL are inherently immunodeficient and are at increased risk of second
primary malignancies (SPM). Additionally, BTKi have been studied in association with
risk for secondary malignancies. The risk of cardiac adverse events and SPM with acalabrutinib
in patients with CLL.
Aims: We conducted this systematic review and meta-analysis to estimate risk of cardiac
adverse events and SPM with acalabrutinib in patients with CLL.
Methods: A comprehensive literature search was performed in PubMed, EMBASE, Clinical
trials.gov and meeting abstracts till January 31,2021. All randomized controlled trials
comparing acalabrutinib-based regimens with a control group in patients with CLL,
with cardiac adverse events and SPM reported as adverse effects were included. Pooled
risk ratios (RR) with 95% confidence intervals (CI) were calculated using the Mantel-Haenszel
method of the fixed-effect model. Heterogeneity of effect size was quantified using
the I2 statistic. Statistical analysis was performed using the Review Manager, version
5.3.
Results: Three phase III RCT with total of 1362 patients were included for our analysis
(Figure). Byrd et al., compared acalabrutinib with ibrutinib. The ASCEND trial compared
acalabrutinib with investigator’s choice chemotherapy, either idelalisib plus rituximab
or bendamustine plus rituximab. In the ELEVATE -TN study, the comparator arm included
the combination of chlorambucil and Obinutuzumab. Acalabrutinib was used at a dose
of 100 mg twice daily in all 3 studies. All but ELEVATE -TN study included patients
previously treated with CLL. Atrial fibrillation was the most common cardiac adverse
event across all trials. Non melanoma skin cancer was the most common SPM across all
trials.
There was no increase in any grade cardiac adverse events with acalabrutinib when
compared to control arm (pooled RR 1.06; 95%CI 0.84-1.34). There was no increased
risk of atrial fibrillation (pooled RR 0.85; 95%CI 0.57-1.26), ventricular arrythmias
and cardiac arrest (Figure). There was significant increase in hypertension (HTN)
in the patients on the acalabrutinib containing regimens (pooled RR 1.78; 95%CI 1.24-2.56;
P= 0.002); however, there was no increase in grade 3+ HTN.
There was a decrease in the SPM with acalabrutinib as compared to the control group
(pooled RR 0.67; 95%CI 0.46-0.96; P= 0.03) (Figure). There was also significant decrease
in non-melanoma skin cancer in the acalabrutinib as compared to the control group
(pooled RR 0.59; 95%CI 0.37-0.93; P 0.02). There was no significant difference in
the genitourinary cancer, lung cancer and hematologic cancer.
Image:
Summary/Conclusion: There is no increase in risk of atrial fibrillation with the acalabrutinib
based regimens as compared to the non-acalabrutinib – based therapies; however, there
was a significantly increased risk of HTN. There is lower risk of SPM and non-melanoma
SPM in the acalabrutinib based regimens than the control arm.
P666: ACALABRUTINIB ± OBINUTUZUMAB VS OBINUTUZUMAB + CHLORAMBUCIL IN TREATMENT-NAIVE
CHRONIC LYMPHOCYTIC LEUKEMIA: 5-YEAR FOLLOW-UP OF ELEVATE-TN
J. P. Sharman1,*, M. Egyed2, W. Jurczak3, A. Skarbnik4, K. Patel5, I. W. Flinn6, M.
Kamdar7, T. Munir8, R. Walewska9, L. M. Fogliatto10, Y. Herishanu11, V. Banerji12,
G. Follows13, P. Walker14, K. Karlsson15, P. Ghia16, A. Janssens17, F. Cymbalista18,
E. Ferrant19, W. G. Wierda20, V. Munugalavadla21, T. Yu21, M. H. Wang21, J. A. Woyach22
1Willamette Valley Cancer Institute and Research Center, Eugene, OR, United States
of America; 2Somogy County Mór Kaposi General Hospital, Kaposvár, Hungary; 3Maria
Skłodowska-Curie National Research Institute of Oncology, Krakow, Poland; 4Novant
Health Cancer Institute, Charlotte, NC; 5Swedish Cancer Institute, Center for Blood
Disorders and Stem Cell Transplantation, Seattle, WA; 6Sarah Cannon Research Institute
and Tennessee Oncology, Nashville, Tennessee; 7University of Colorado Cancer Center,
Aurora, CO, United States of America; 8Haematology, Haematological Malignancy Diagnostic
Service (HMDS), St. James’s Institute of Oncology, Leeds; 9Cancer Care, University
Hospitals Dorset, Bournemouth, United Kingdom; 10Hospital de Clinicas de Porto Alegre,
Porto Alegre, Brazil; 11Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; 12Departments
of Internal Medicine, Biochemistry & Medical Genetics, Max Rady College of Medicine,
Rady Faculty of Health Sciences, University of Manitoba and CancerCare Manitoba, Winnipeg,
Canada; 13Department of Haematology, Addenbrooke’s Hospital NHS Trust, Cambridge,
United Kingdom; 14Peninsula Health and Peninsula Private Hospital, Frankston, Melbourne,
Australia; 15Skåne University Hospital, Lund, Sweden; 16Università Vita-Salute San
Raffaele and IRCCS Ospedale San Raffaele, Milano, Italy; 17University Hospitals Leuven,
Leuven, Belgium; 18Bobigny: Hématologie biologique, Hopital Avicenne, Université Sorbonne
Paris Nord, Bobigny; 19Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Service
d’Hématologie Clinique, Pierre-Bénite, France; 20Department of Leukemia, Division
of Cancer Medicine, MD Anderson Cancer Center, Houston, Texas; 21AstraZeneca, South
San Francisco, CA; 22The Ohio State University Comprehensive Cancer Center, Columbus,
OH, United States of America
Background: For ELEVATE-TN (NCT02475681), we previously reported superior efficacy
of acalabrutinib (A) ± obinutuzumab (O) vs O + chlorambucil (Clb) in patients with
treatment-naive (TN) chronic lymphocytic leukemia (CLL) at 28.3 and 46.9 months median
follow-up.
Aims: To report the updated efficacy and safety results from the ELEVATE-TN study
after a median follow-up of 5 years.
Methods: Patients were randomized to A+O, A, or O+Clb after informed consent was obtained.
Patients who progressed on O+Clb could cross over to A monotherapy. Investigator-assessed
(INV) progression-free survival (PFS), INV overall response rate (ORR), overall survival
(OS), and safety were evaluated.
Results: A total of 535 patients (A+O, n=179; A, n=179; O+Clb, n=177) had a median
age of 70 years; 63% had unmutated IGHV and 9% had del(17p). At a median follow-up
of 58.2 months (range, 0.0–72.0; data cutoff Oct 1, 2021), median PFS was not reached
(NR) (hazard ratio [HR]: 0.11) for A+O and A (HR: 0.21) vs 27.8 months for O+Clb (both
P<0.0001). The estimated 60-month PFS rates were 84% (A+O), 72% (A), and 21% (O+Clb).
In patients with unmutated IGHV, the median PFS was NR for A+O and A vs 22.2 months
among patients in the O+Clb arm (HR: 0.06 [A+O] and 0.12 [A]; both P<0.0001); estimated
60-month PFS rates were 82% (A+O), 72% (A), and 6% (O+Clb). In patients with del(17p),
the median PFS was NR and 64.1 months for A+O and A vs 17.7 months for O+Clb (HR:
0.21, P=0.0031 [A+O]; HR: 0.29, P=0.0130 [A]); estimated 60-month PFS rates were 75%
(A+O), 71% (A), and 27% (O+Clb). Median OS was NR in any treatment arm, and significantly
longer in the A+O vs O+Clb arms (HR: 0.55; P=0.0474); estimated 60-month OS rates
were 90% (A+O), 84% (A), and 82% (O+Clb). ORR was significantly higher with A+O (96%;
95% CI 92–98) and A (90%; 85–94) vs O+Clb (83%; 77–88; P<0.0001 [A+O], P=0.0499 [A]).
Complete response (CR)/CR with incomplete hematologic recovery (CRi) rates were higher
with A+O (29%/3%) vs O+Clb (13%/1%); 13%/1% had CR/CRi with A; CR increased since
the interim analysis (previously 21% [A+O] and 7% [A]). Adverse events (AEs) and treatment
exposure are shown in the Table. Treatment is ongoing in 65% (A+O) and 60% (A) of
patients; the most common reasons for treatment discontinuation were AEs (17% [A+O],
16% [A], 14% [O+Clb]) and progressive disease (6%, 10%, 2%, respectively). Crossover
from O+Clb to A occurred in 72 (41%) patients; 25% of these patients discontinued
A (10% due to AEs and 11% due to progressive disease).
Image:
Summary/Conclusion: After 5 years of follow-up, efficacy and safety of A+O and A monotherapy
were maintained, with significantly longer OS in the A+O arm compared with O+Clb.
P667: LONG-TERM EFFICACY OF ACALABRUTINIB-BASED REGIMENS IN PATIENTS WITH CHRONIC
LYMPHOCYTIC LEUKEMIA AND HIGHER-RISK GENOMIC FEATURES: POOLED ANALYSIS OF CLINICAL
TRIAL DATA
M. Davids1,*, J. Sharman2, P. Ghia3, J. Woyach4, W. Jurczak5, T. Siddiqi6, P. Miranda7,
M. Shahkarami8, T. Yu8, U. Emeribe7, J. Byrd9
1Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts;
2Willamette Valley Cancer Institute and Research Center, Eugene, Oregon, United States
of America; 3Università Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele,
Milano, Italy; 4The Ohio State University Comprehensive Cancer Center, Columbus, Ohio,
United States of America; 5Maria Skłodowska-Curie National Research Institute of Oncology,
Krakow, Poland; 6City of Hope Comprehensive Cancer Center, Duarte, California; 7AstraZeneca,
Gaithersburg, Maryland; 8AstraZeneca, South San Francisco, California; 9Department
of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio,
United States of America
Background: Chronic lymphocytic leukemia (CLL) has a highly variable disease course;
patients (pts) with higher-risk genomic features typically have poor response to chemoimmunotherapy,
and pts with del(17p) also have short, suboptimal response to venetoclax + obinutuzumab.
Thus, novel agents such as Bruton tyrosine kinase inhibitors (BTKi), including the
highly selective covalent BTKi acalabrutinib (A), are preferred treatments in this
higher-risk population based on demonstrated effectiveness in higher-risk subgroups
in individual studies.
Aims: To assess long-term efficacy of A-based regimens in pts with treatment-naive
(TN) or relapsed/refractory (R/R) CLL and higher-risk genomic features.
Methods: Data were pooled from CLL pts with higher-risk genomic features treated with
A ± obinutuzumab (O) in 3 clinical studies in TN CLL (ELEVATE-TN, CL-001 [TN cohort],
CL-003) and 3 in R/R CLL (ASCEND, ELEVATE-RR, CL-001 [R/R cohort]). Higher risk was
defined as del(17p) and/or TP53 mutation [del(17p)/TP53m], unmutated IGHV (uIGHV),
or complex karyotype (CK, ≥3 chromosomal abnormalities). Efficacy analyses (progression-free
survival [PFS], overall survival [OS], response rates) focus on del(17p)/TP53m and
uIGHV; safety analyses also include pts with CK.
Results: 801 pts (TN 313; R/R 488, median 2 prior therapy lines [range 1–10]) were
included; 64 (20%) TN and 219 (45%) R/R pts had del(17p)/TP53m, among whom 44 (69%)
and 170 (78%) also had uIGHV. Overall, 288 (92%) TN and 425 (87%) R/R pts had uIGHV
and 47 (15%) and 160 (33%) had CK. Median pt age was 68 (TN) and 66 (R/R) y. For A-based
regimens combined (A and A+O), overall response rate (ORR) was 91% (n=58/64; complete
response [CR]: 20% [n=13/64]) in TN pts with del(17p)/TP53m and 96% (n=276/288; CR
16% [n=47/288) in TN pts with uIGHV. At 47.3 mo median follow-up (range 1.0–82.0),
median PFS was not reached (NR) in TN pts with del(17p)/TP53m with A-based regimens;
PFS rates at 48 mo suggest similar efficacy with A and A+O in TN pts with del(17p)/TP53m
(76% and 77%, respectively) (Fig 1A). Median PFS was NR among TN pts with uIGHV with
both A and A+O; PFS rates at 48 mo with A and A+O were 84% and 86%, respectively (Fig
1B). Median OS was NR with A and A+O in TN pts with del(17p)/TP53m or uIGHV; OS rates
at 48 mo were similar for A and A+O in pts with del(17p)/TP53m (89% for both) and
in pts with uIGHV (92% and 95%).
In the R/R cohort (A monotherapy only), ORR was 86% (n=184/214; CR 5% [n=11/214])
in pts with del(17p)/TP53m and 87% (n=356/408; CR 7% [n=29/408]) in pts with uIGHV.
At 44.0 mo median follow-up (range 0–88.0), median PFS was 38.6 mo and 46.9 mo and
PFS rates at 36 mo were 54% and 65% in pts with del(17p)/TP53m and uIGHV, respectively
(Fig 1C). Median OS was 60.6 mo and NR in pts with del(17p)/TP53m and uIGHV, respectively;
OS rates at 36 mo were 73% and 82%.
Among all higher-risk pts, incidences of grade ≥3 treatment-related AEs were 30% (TN)
and 35% (R/R). Discontinuations due to treatment-related AEs occurred in 4% (TN) and
6% (R/R) of pts; 60% and 27%, respectively, remained on treatment at data cutoff.
Image:
Summary/Conclusion: In this pooled analysis of clinical trial data in 801 CLL pts
with higher-risk genomic features, efficacy of A-based regimens led to high PFS and
OS rates at a median follow-up of nearly 4 y. The safety profile in this analysis
was similar to the overall safety profile of acalabrutinib. Our results demonstrate
the long-term benefit of A-based regimens in CLL pts with higher-risk genomic features,
regardless of line of therapy.
P668: ACALABRUTINIB VS RITUXIMAB PLUS IDELALISIB OR BENDAMUSTINE IN RELAPSED/REFRACTORY
CHRONIC LYMPHOCYTIC LEUKEMIA: ASCEND RESULTS AT ~4 YEARS OF FOLLOW-UP
P. Ghia1,*, A. Pluta2, M. Wach3, D. Lysak4, M. Simkovic5, I. Kriachok6, A. Illes7,
J. de la Serna8, S. Dolan9, P. Campbell10, G. Musuraca11, A. Jacob12, E. Avery13,
J. H. Lee14, G. Usenko15, M. H. Wang16, T. Yu17, W. Jurczak18
1Università Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele, Milano, Italy;
2Department of Hematological Oncology, Oncology Specialist Hospital, Brzozow; 3Department
of Hemato-Oncology and Bone Marrow Transplantation, Medical University of Lublin,
Lublin, Poland; 4Fakultní Nemocnice Plzen, Pilsen; 5University Hospital Hradec Kralove,
Charles University, Hradec Kralove, Czechia; 6National Cancer Institute, Kiev, Ukraine;
7University of Debrecen, Faculty of Medicine, Department of Hematology, Debrecen,
Hungary; 8Hospital Universitario 12 de Octubre, Madrid, Spain; 9Saint John Regional
Hospital, University of New Brunswick, New Brunswick, Canada; 10Barwon Health, University
Hospital Geelong, Geelong, Victoria, Australia; 11Istituto Scientifico Romagnolo per
lo Studio e la Cura dei Tumori, Meldola, Italy; 12The Royal Wolverhampton NHS Trust,
Wolverhampton, United Kingdom; 13Nebraska Hematology Oncology, Lincoln, Nebraska,
United States of America; 14Gachon University Gil Medical Center, Incheon, South Korea;
15City Clinical Hospital No. 4 DCC, Dnipro, Ukraine; 16AstraZeneca, South San Francisco,
California; 17AstraZeneca, South San Francisco, California, United States of America;
18Maria Sklodowska-Curie National Institute of Oncology, Krakow, Poland
Background: Acalabrutinib (acala) is a next-generation, highly selective, covalent
Bruton tyrosine kinase (BTK) inhibitor approved for patients (pts) with chronic lymphocytic
leukemia (CLL). In the primary analysis of ASCEND (median follow-up 16.1 mo), acala
showed superior efficacy with an acceptable tolerability profile vs idelalisib (Id)
plus rituximab (R) (IdR) or bendamustine (B) plus R (BR) in pts with relapsed/refractory
(R/R) CLL (Ghia et al. J Clin Oncol. 2020;38:2849-2861).
Aims: We report the results of the ASCEND study at ~4 years of follow-up.
Methods: In this multicenter, randomized, open-label, phase 3 study (NCT02970318),
pts with R/R CLL received oral (PO) acala 100 mg BID until progression or unacceptable
toxicity or investigator’s (INV) choice of IdR (Id: 150 mg PO BID until progression
or unacceptable toxicity; R: 375 mg/m2 x1 then 500 mg/m2 IV [8 total infusions]) or
BR (B: 70 mg/m2 IV; R: 375 mg/m2 x1 then 500 mg/m2 IV [6 cycles]). Progression-free
survival (PFS), overall survival (OS), overall response rate (ORR), and safety were
assessed.
Results: A total of 310 pts (acala, n=155; IdR, n=119; BR, n=36) were randomized (median
age 67 y; del(17p) 15%, unmutated IGHV 74%, Rai stage 3/4 42%). At median follow-up
of 46.5 mo (acala) and 45.3 mo (IdR/BR), acala significantly prolonged INV-assessed
PFS vs IdR/BR (median not reached [NR] vs 16.8 mo; P<0.0001); 42-mo PFS rates were
62% for acala vs 19% for IdR/BR. In pts with del(17p), median PFS was NR (acala) vs
13.8 mo (IdR/BR; P<0.0001). In pts with unmutated IGHV, median PFS was NR (acala)
vs 16.2 mo (IdR/BR; P<0.0001). Median OS was NR in both arms; 42-mo OS rates were
78% (acala) vs 65% (IdR/BR). ORR was 83% (acala) vs 84% (IdR/BR) (ORR + partial response
with lymphocytosis: 92% [acala] vs 88% [IdR/BR]). AEs led to drug discontinuation
in 23% of acala, 67% of IdR, and 17% of BR pts. Events of clinical interest (acala
vs IdR/BR) included all-grade atrial fibrillation/flutter (8% vs 3%), all-grade hypertension
(8% vs 5%), all-grade major hemorrhage (3% vs 3%), and grade ≥3 infections (29% vs
29%).
Image:
Summary/Conclusion: At ~4 years of follow-up, acala maintained efficacy compared with
standard-of-care regimens and a consistent tolerability profile in R/R CLL.
P669: FIXED-DURATION (FD) IBRUTINIB + VENETOCLAX FOR FIRST-LINE TREATMENT OF CHRONIC
LYMPHOCYTIC LEUKEMIA (CLL)/SMALL LYMPHOCYTIC LYMPHOMA (SLL): 3-YEAR FOLLOW-UP FROM
THE PHASE 2 CAPTIVATE STUDY FD COHORT
C. Moreno1,2,*, W. G. Wierda3, P. M. Barr4, T. Siddiqi5, J. N. Allan6, T. J. Kipps7,
L. Trentin8, R. Jacobs9, S. Jackson10, A. Tedeschi11, S. Opat12, R. Bannerji13, B.
J. Kuss14, L. J. Croner15,16, E. Szafer-Glusman15,16, C. Zhou16, A. Szoke16, J. P.
Dean16, P. Ghia17, C. S. Tam18
1Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona; 2Josep
Carreras Leukemia Research Institute, Barcelona, Spain; 3Department of Leukemia, University
of Texas MD Anderson Cancer Center, Houston, TX; 4Wilmot Cancer Institute, University
of Rochester Medical Center, Rochester, NY; 5City of Hope National Medical Center,
Duarte, CA; 6Weill Cornell Medicine, New York, NY; 7UCSD Moores Cancer Center, La
Jolla, CA, United States of America; 8University of Padova, Padova, Italy; 9Levine
Cancer Institute, Charlotte, NC, United States of America; 10Middlemore Hospital,
Auckland, New Zealand; 11ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy;
12Monash University, Clayton, VIC, Australia; 13Rutgers Cancer Institute of New Jersey,
New Brunswick, NJ, United States of America; 14Flinders University and Medical Center,
Bedford Park, SA, Australia; 15AbbVie, North Chicago, IL; 16Pharmacyclics LLC, an
AbbVie Company, South San Francisco, CA, United States of America; 17Università Vita-Salute
San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy; 18Peter MacCallum Cancer
Center & St. Vincent’s Hospital and the University of Melbourne, Melbourne, VIC, Australia
Background: CAPTIVATE (PCYC-1142) is a multicenter phase 2 study of first-line ibrutinib
+ venetoclax in CLL. The primary analysis evaluating FD treatment with ibrutinib +
venetoclax was previously presented (Ghia et al., ASCO 2021).
Aims: To present 3-year follow-up results from the FD cohort of CAPTIVATE.
Methods: Patients aged ≤70 years with previously untreated CLL/SLL received 3 cycles
of ibrutinib then 12 cycles of ibrutinib + venetoclax (ibrutinib 420 mg/day orally;
venetoclax ramp-up to 400 mg/day orally). Responses were investigator assessed per
iwCLL 2008 criteria. Undetectable minimal residual disease (uMRD; <10-4) was measured
by 8-color flow cytometry. Serious AEs (SAEs) deemed related to ibrutinib reported
>30 days after last dose of study drug were collected.
Results: 159 patients were enrolled (median age 60 years), including patients with
high-risk features of del(17p)/TP53 mutation (17%), unmutated IGHV (uIGHV; 56%), and
complex karyotype (19%). 147 (92%) and 149 (94%) patients completed treatment with
ibrutinib and venetoclax, respectively. With 1 year of additional follow-up since
primary analysis, median time on study was 39 months (range 1-41). ORR was 96% and
was consistent (96%-97%) in patients with high-risk features (Table). The primary
endpoint of complete response (CR) including CR with incomplete bone marrow recovery
(CRi) rate in patients without del(17p) (n=136) increased nominally from 56% (95%
CI, 48-64) to 58% (95% CI 50-66); in all patients, CR rate increased from 55% (95%
CI 48-63) to 57% (95% CI 50-65). In patients achieving CR, 93% had durable responses
lasting ≥12 months post-treatment. Of patients with uMRD in peripheral blood at 3
months post-treatment, 66/85 (78%) evaluable patients maintained uMRD through 12 months
post-treatment. At 36 months, PFS was 88% (95% CI 82‒92) and OS was 98% (95% CI 94-99);
similar rates were seen in patients with high-risk features (Table). All patients
are off treatment; no new SAEs of any kind have occurred since the primary analysis.
Available data on relevant mutations in BTK, PLCɣ2, or BCL-2 at time of PD will be
presented. As of January 2022, 12 patients were retreated with single-agent ibrutinib
after progressive disease (treatment duration range 3-29 months); of evaluated patients,
7/9 had partial responses and 2/9 had stable disease.
Image:
Summary/Conclusion: Fixed duration ibrutinib + venetoclax continues to provide deep,
durable responses and clinically meaningful PFS, including in patients with high-risk
disease features, representing an all-oral, once-daily, chemotherapy-free FD regimen
for previously untreated patients with CLL/SLL. With an additional 1 year of follow-up,
no OS events or SAEs occurred. Manageable safety profile is unchanged as previously
reported. To date, successful single-agent ibrutinib retreatment responses are observed.
P670: ABSENCE OF BTK, BCL2, AND PLCG2 MUTATIONS IN RELAPSING CHRONIC LYMPHOCYTIC LEUKEMIA
(CLL) AFTER FIRST-LINE TREATMENT WITH FIXED-DURATION IBRUTINIB (I) PLUS VENETOCLAX
(V)
L. J. Croner1,2,*, J. N. Allan3, N. Jain4, J. F. Seymour5, K. Eckert2, L. W. K. Cheung1,2,
E. Szafer-Glusman1,2
1AbbVie, North Chicago, IL; 2Pharmacyclics LLC, an AbbVie Company, South San Francisco,
CA; 3Weill Cornell Medicine, New York, NY; 4MD Anderson Cancer Center, Houston, TX,
United States of America; 5Peter MacCallum Cancer Center & Royal Melbourne Hospital,
Melbourne, VIC, Australia
Background: CAPTIVATE (PCYC-1142; NCT02910583) is a multicenter phase 2 study of first-line
I+V in patients (pts) with CLL in 2 cohorts: Minimal Residual Disease (MRD) and Fixed
Duration (FD). We previously reported that fixed-duration I+V provides deep, durable
responses (Ghia et al, ASCO 2021; Wierda et al, J Clin Oncol 2021). Clinically-relevant
mutations in BTK, PLCG2, and BCL2 have previously been reported in pts with progressive
disease (PD) following single agent BTK or BCL2 inhibitor therapies. Such resistance-associated
mutations may occur with increasing frequency after prolonged drug exposure. Fixed
duration I+V is hypothesized to decrease the likelihood of developing resistance mutations
and facilitate extended clinical benefit.
Aims: We sought to ascertain presence or absence of resistance mutations at the time
of PD in samples from pts treated with fixed-duration I+V in CAPTIVATE. Baseline genomic
risk features and potentially prognostic mutations in CLL-related genes of interest
were also evaluated in pts with and without subsequent PD.
Methods: The analysis was conducted in 191 pts aged ≤70 y with previously untreated
CLL who completed fixed-duration I+V, defined as 3 x 28d cycles of I then 12 cycles
of I+V (I 420 mg/d orally; V ramp-up to 400 mg/d orally); this included pts from the
FD cohort, and pts from the MRD cohort with confirmed undetectable MRD who received
an additional cycle of I+V and were randomized to placebo. Samples from pts at the
time of PD were evaluated for resistance mutations by targeted next-generation sequencing
(NGS) using a custom-designed high-read-depth 100-gene panel (limit of detection 1%
VAF, negative predictive value ≥95% assuming population prevalences <20%, for BTK,
PLCG2, or BCL2 variants) in CD19-enriched peripheral blood (PB) or bone marrow. Mutations
in additional genes of interest (Table) were evaluated by NGS using the Personalis
ACE panel in baseline PB samples. Genomic risk features (Table) were also assessed
at baseline. Mutations and genomic risk features evident at baseline were compared
across PD and non-PD pts.
Results: Baseline mutations in genes of interest and genomic risk features in 190
pts who completed fixed-duration I+V and with available baseline data are shown in
the Table. Rates of mutations were similar between pts with (n=30) and without (n=160)
subsequent PD. Of 30 pts with PD events (occurring between 16 and 53 mos after start
of treatment), 22 (73%) had baseline mutations in genes of interest and 27 (90%) had
≥1 baseline genomic risk feature. Pts with PD generally had more baseline genomic
risk features than pts without PD (median 3 vs 2; Wilcoxon P=0.02; analysis restricted
to FD cohort pts to avoid bias). To date, 26 of 30 PD patient samples have been evaluated
for mutations; no resistance-associated variants in BTK, PLCG2, or BCL2 were detected.
Image:
Summary/Conclusion: Earlier work has demonstrated a promising safety profile and efficacy
of fixed-duration I+V in first-line CLL. Importantly, the current results show that
no resistance-associated mutations in BTK, PLCG2, or BCL2 were identified in the pts
with PD evaluated to date after first-line, fixed-duration I+V. The absence of such
mutations at PD suggests that fixed-duration I+V treatment may offer the possibility
of effective subsequent retreatment options with I and/or V and extend clinical benefit.
P671: TREATMENT SEQUENCES AND OUTCOMES OF PATIENTS WITH CLL TREATED WITH TARGETED
AGENTS IN REAL-WORLD SETTINGS
A. R. Mato1,*, B. S. Manzoor2, C. C. Coombs3, N. Lamanna4, H. H. Tuncer5, J. R. Brown6,
L. E. Roeker1, M. Shadman7, J. N. Allan8, C. Ujjani7, B. Eichhorst9, L. Leslie10,
I. Fleury11, H. Alhasani2, J. Rhodes12, B. T. Hill13, P. M. Barr14, A. Skarbnik15,
M. S. Davids6, R. Bannerji16, M. Fuldeore2, F. Lansigan17, A. Schuh18, L. Pearson19,
C. P. Fox20, I. Pivneva21, L. Popadic22, A. Guerin21, T. A. Eyre18
1Memorial Sloan Kettering Cancer Center, New York; 2AbbVie, Inc., North Chicago; 3Lineberger
Comprehensive Cancer Center, Chapel Hill; 4Columbia University, New York; 5The Cancer
Center at Lowell General Hospital, Lowell; 6Dana-Farber Cancer Institute, Boston;
7Fred Hutchinson Cancer Research Center, Seattle; 8Weill Cornell Medicine University
Medical Center, New York, United States of America; 9University of Cologne, Cologne,
Germany; 10John Theurer Cancer Center, Hackensack, United States of America; 11Hôpital
Maisonneuve-Rosemont, Montreal, Canada; 12Northwell Health Cancer Institute, Lake
Success; 13Cleveland Clinic, Cleveland; 14Wilmot Cancer Institute, Rochester; 15Novant
Health, Charlotte; 16Rutgers Cancer Institute of New Jersey, New Brunswick; 17Dartmouth-Hitchcock
Medical Center, Lebanon, United States of America; 18Churchill Hospital, Oxford, United
Kingdom; 19Tufts Medical Center, Boston, United States of America; 20Nottingham University
Hospitals NHS Trust, Nottingham, United Kingdom; 21Analysis Group, Inc., Montreal,
Canada; 22Analysis Group, Inc., New York, United States of America
Background: Treatment (tx) options for patients (pts) with chronic/small lymphocytic
leukemia (CLL/SLL) have expanded following regulatory approval of targeted agents.
However, real-world evidence regarding pt outcomes and tx sequences is evolving.
Aims: We assessed tx trends, sequences and outcomes following the introduction of
targeted agents.
Methods: The CLL Collaborative Study of Real-World Evidence (CORE), a retrospective,
international, observational study (23 centers) for CLL/SLL pts provided data for
this analysis. Pts were diagnosed with CLL/SLL, treated in the relapsed/refractory
(R/R) setting after 2/12/2014. Tx sequences following targeted agents (ie, BTKi: acalabrutinib,
ibrutinib; BCL2i: venetoclax), clinical response (physician-assessed in medical charts
and reported as overall response rate [ORR]), and progression-free survival (PFS)
were assessed.
Results: Of the 1,624 study pts, in 1L 48.8% initiated CT/CIT, 40.6% targeted agents
(BTKi: 32.6%; BCL2i: 4.5%), and 10.6% other therapies. We observed 1L CT/CIT use decreased
starting 2014 (pre-2014: 58.1%; 2014–2016: 46.0%; 2017–2019: 24.6%; 2020–2021: 0%),
while 1L targeted agent use increased over time (pre-2014: 30.5%; 2014–2016: 43.1%;
2017–2019: 67.0%; 2020–2021: 94.7%). Post-targeted agent introduction in 2014, the
most frequent tx sequence has changed from 1L CT/CIT→BCRi in 2014–2016 to 1L BCRi→BCL2i
in 2017–2021 (Figure 1).
Across lines and years of tx, among 650 R/R CLL/SLL pts who initiated therapy >2/12/2014,
536 pts (82.5%) received BTKi-based regimens and 214 pts (32.9%) received BCL2i-based
regimens. Of the 536 pts receiving BTKi-based regimens (ibrutinib alone: 454 [85.5%];
acalabrutinib alone: 21 [4.0%]), majority was in 2L and 3L+ (291 pts (54.3%); 167
pts (31.2%), respectively). Of the 457 pts with response assessed, 343 pts (75.1%)
achieved ORR in the line of therapy (LOT) with BTKi-based regimen. The median PFS
for pts receiving BTKi-based regimen was 36.4 months (95% confidence interval [CI]:
33.1, 41.9). 236 pts (44.0%) receiving a BTKi-based regimen had a subsequent LOT;
of the 177 pts with response assessed, 120 pts (67.8%) achieved ORR. The most common
subsequent tx were BCL2i-based (N=113; ORR: 76.2%) or another BTKi-based (N=41; ORR:
54.8%) regimens.
Of the 214 pts receiving BCL2i (venetoclax alone: 129 [60.3%]), majority was in 2L
and 3L+ (77 pts [36.0%] and 132 pts [61.7%], respectively). Of the 164 pts with response
assessed, 127 pts (77.4%) achieved ORR in the LOT with BCL2i-based regimen. The median
PFS for pts receiving BCL2i was 26.2 months (95% CI: 20.0, 38.7). 47 pts (22.0%) receiving
BCL2i had a subsequent LOT; of the 37 pts with response assessed, 27 pts (73.0%) achieved
ORR. The most common subsequent tx were BTKi-based (ibrutinib-based regimen: 14 [29.8%];
acalabrutinib-based regimen: 5 [10.6%]; ORR: 76.9%) or another BCL2i-based (venetoclax-based
re-tx: 8 [17.8%]; ORR: 80.0%) regimens.
Image:
Summary/Conclusion: 1L CT/CIT use is declining since 2014 and 1L targeted agent use
is now the standard of care in real-world practice. Of currently available targeted
therapies, ibrutinib was the most commonly used. These results demonstrate sequences
of BTKi use following venetoclax (venetoclax→BTKi) are effective, as well rechallenging
with venetoclax (venetoclax→venetoclax), providing support for either strategy in
the R/R CLL/SLL management. These results provide clinicians with valuable insights
regarding pt outcomes associated with different tx sequences currently considered
in modern clinical practice. Updated pt level data and survival analyses are underway.
P672: REAL-LIFE EFFICACY AND SAFETY OF VENETOCLAX MONOTHERAPY IN RELAPSED/ REFRACTORY
CHRONIC LYMPHOCYTIC LEUKEMIA - INTERIM ANALYSIS OF MULTICENTRIC STUDY VERONE
L. Ysebaert1,*, X. Troussard2, V. Levy3, R. Le Calloch4, R. Guieze5, S. Leprêtre6,
A.-S. Michallet7, V. Leblond8, P. Feugier9, J. Ramier10, A. Delmer11
1Hematology Unit, IUCT Oncopole, Toulouse; 2Hematology Unit, CHU Caen, Caen; 3Hematology
Unit, Hopital Avicenne, Bobigny; 4Hematology Unit, CH Cornouaille, Quimper; 5Hematology
Unit, CHU Clermont-Ferrand, Clermont-Ferrand; 6Hematology Unit, Henri Becquerel Cancer
Research Center, Rouen; 7Hematology Unit, Leon Berard Cancer Reseach Center, Lyon;
8Hematology Unit, Hôpital Pitié-Salpêtrière, Paris; 9Hematology Unit, CHRU Nancy,
Nancy; 10AbbVie France, Rungis; 11Hematology Unit, CHU Reims, Reims, France
Background: The efficacy and safety of venetoclax (VEN), the first oral inhibitor
of BCL-2, has been demonstrated in clinical trials in patients with chronic lymphocytic
leukemia (CLL). However, few real-life data in France is available today on this treatment.
Aims: VERONE is an observational ongoing study, it aims to describe the efficacy and
tolerance of VEN in a real-life situation, as well as its use in current medical practice.
Methods: This longitudinal non-interventional study is conducted in adults starting
treatment with VEN for CLL, with a follow-up over 48 months. This second interim analysis
was performed in patients treated with monotherapy until progression, 43 months after
the first inclusion. The overall response rate (ORR, assessed by investigator) and
progression-free survival (PFS) were estimated 1 and 2 years after the first intake
of VEN in patients with at least one follow-up visit in the study (evaluable population).
Results: From March 2018 to September 2021, 196 patients included in 64 centers started
VEN as monotherapy for CLL (tolerance population), 195 met the selection criteria
(analysis population) and 113 could be evaluated (evaluable population). In the analysis
population (n = 195), patients and CLL characteristics at baseline were: median age
74.0 years (33.0-91.0), male 68.2%, ECOG ≥2 18.5%, ≥1 comorbidities 87.7%, median
time since diagnosis 8.3 years (0.0-29.4), 37.3% of 169 genetic examinations was TP53
and/ or del (17)p, median number of previous treatments 2 (1-9) (chemoimmunotherapy
87.2%, BCR inhibitors (ibrutinib, idelalisib) 70.8%). Before the first intake of VEN,
preventive measures against tumor lysis syndrome (TLS) were taken in the majority
of cases (hospitalization 85.2%, hydration 85.2% and anti-hyperuricemia prophylaxis
66.3%). The median duration of the titration period was 5.0 weeks, and 77.0% of patients
reached the recommended dose of 400mg at the time of analysis.
In the 113 evaluable patients, the overall response rate was 91.2% [95%CI 84.5%-95.1%]
at 1 year and 92.9% [95%CI 86.7%-96.4%] at 2 years. After a median follow-up of 23.2
months, the PFS rate was 81.8% [95%CI 72.5%-88.2%] at 1 year and 71.1% [95%CI 58.0%
80.7%] at 2 years. The median duration of treatment was 15.5 months.
In terms of safety, 85.2% of the 196 patients presented adverse events [AEs, neutropenia
(46.4%) and diarrhea (24.5%)], including 64.3% of AEs of grade ≥3 (including 32.7%
neutropenia). Permanent discontinuation of treatment due to AEs related to VEN was
observed in 20.4% of patients. 4 AEs grade 5 related to VEN was observed in 3 patients
(deterioration of general condition, ascites, hepatic failure, death).
At inclusion, the risk level of developing TLS, assessed in 149 patients, was low
(28.2%) moderate (69.8%) or high (2.0%). During the dose escalation phase, 19 patients
(9.7%) developed TLS and 4 (2.0%) stopped VEN treatment.
Summary/Conclusion: This real-life study confirms the efficacy of VEN in heavily treated
patients with a safety profile little different from that observed in selected populations
in clinical trials.
P673: DEPLETION AND RECOVERY OF NORMAL B-CELLS DURING AND AFTER TREATMENT WITH CHEMOIMMUNOTHERAPY,
IBRUTINIB OR VENETOCLAX.
A. Rawstron1,*, N. Webster2, A. Pitchford3, S. Dalal2, A. Bloor4, R. de Tute1, A.
Hockaday3, S. Jackson3, D. Cairns3, N. Greatorex3, D. Allsup5, T. Munir6, P. Hillmen2
1HMDS, Leeds Cancer Centre; 2Experimental Haematology, University of Leeds; 3Leeds
Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research,
Leeds; 4Haematology, The Christie NHS Foundation Trust, Manchester; 5Haematology,
Hull York Medical School, Hull; 6Haematology, Leeds Cancer Centre, Leeds, United Kingdom
Background: Infectious complications are a major cause of morbidity and mortality
in Chronic Lymphocytic Leukaemia (CLL). Therapeutic approaches that deplete CLL cells
also affect normal B-cells. Optimal treatment would result in eradication of CLL cells
and recovery of normal immune function. FLAIR (ISRCTN01844152) is a phase III trial
for previously untreated CLL comparing ibrutinib plus rituximab (IR) with fludarabine,
cyclophosphamide and rituximab (FCR) and subsequently amended to also compare ibrutinib
plus venetoclax (I+V) and ibrutinib alone (I) with FCR. Measurable residual disease
(MRD) and normal B-cell levels were assessed at multiple timepoints.
Aims: To assess the depletion of normal B-cells during treatment and recovery after
end of treatment.
Methods: Participants aged under 75 years with <20% TP53-deleted cells were initially
randomised to FCR or IR and subsequently to FCR, IR, I+V or I with the IR arm closed
after randomisation of 771 participants to FCR/IR. FCR was given for 6 cycles, while
treatment in the IR, I and I+V arms continued for up to 6 years except in participants
attaining <0.01% MRD who continued treatment for the time taken to achieved MRD <0.01%
and then stopped if MRD remained <0.01%. Month (M) 24 was earliest permitted stopping
point. MRD flow cytometry was performed according to ERIC guidelines (panel: CD19/5/20/43/79/81+ROR1,
acquisition of 0.5–2.2 million cells, BD Biosciences Lyric). Additional analysis of
normal B-cell subsets was performed in a cohort of >500 patients (panel: CD19 to identify
B-cells, CD20/5/79b+ROR1 and CD3 to exclude CLL & contaminating cells, with CD27/
38/IgD/IgM to characterise normal B-cell subsets using a Coulter Cytoflex LX).
Results: Normal B-cells were undetectable during FCR treatment and only rarely detectable
until 12 months after last FCR cycle. Circulating normal B-cells were reduced in number
or undetectable in participants receiving ibrutinib-containing regimens with greater
depletion in the I+V and IR arms relative to I monotherapy. B-progenitors persist
through FCR treatment but were depleted during I, I+R or I+V treatment. Normal B-cell
levels at 24 and 36 months after randomisation, with time off-treatment if applicable,
are shown in Figure 1.
In the ibrutinib-containing arms (IR, I, and I+V), there was a trend towards fewer
COVID-associated SAE at any time point for participants with detectable B-cells at
24M (4/181, 2.2%) compared to those with no detectable B-cells (14/344, 4.1%) and
COVID-associated SAEs were not observed in FCR-treated participants who had recovered
any level of normal B-cells by 24M (0/215). However, the data on COVID infections
are limited and there was no apparent association between normal B-cell levels at
24M with the proportion of participants experiencing an infectious SAE overall. Assessment
of normal B-cell subsets during ibrutinib-based treatment demonstrated a mix of naïve
and memory B-cells. Serological response to COVID infection/vaccination in this cohort
is currently being performed.
Participants stopping I+V treatment at 24-30 months post-randomisation due to MRD
eradication showed rapid recovery of normal naive B-cells within 6-12 months after
end of treatment in the vast majority (>95%) of evaluable cases.
Image:
Summary/Conclusion: Normal circulating B-cells are depleted during treatment with
rituximab but can persist at a low level during I, IR or I+V treatment. Most patients
in remission after treatment with FCR or I+V show recovery of normal B-cells at 12
months of stopping treatment.
P674: UPDATED RESULTS OF A PHASE 1B STUDY OF IBRUTINIB (IBR) PLUS OBINUTUZUMAB (OBIN)
IN PATIENTS WITH RELAPSED OR REFRACTORY (R/R) CHRONIC LYMPHOCYTIC LEUKEMIA (CLL)
C. Ryan1,*, D. Brander2, P. Barr3, S. Tyekucheva4, Y. Ren4, L. Hackett1, M. McDonough1,
K. Pena Del Aguila1, M. McEwan1, M. Collins1, J. Abramson1, E. Jacobsen1, A. LaCasce1,
D. Fisher1, J. Brown1, M. Davids1
1Medical Oncology, Dana-Farber Cancer Institute, Boston; 2Duke University Cancer Center,
Durham; 3University of Rochester Wilmot Cancer Institute, Rochester; 4Data Science,
Dana-Farber Cancer Institute, Boston, United States of America
Background: Combination ibr + obin was approved in frontline CLL based on results
of iLLUMINATE; however, less data are available on this regimen in R/R CLL. Though
active in R/R CLL, ibr monotherapy in RESONATE led to 4-year (yr) PFS/OS of ~45%/65%,
leaving room for improvement. We previously reported initial safety and efficacy results
of this phase 1b study investigating ibr + obin in R/R CLL (Davids et al., EHA 2020).
Here, we report updated results, with median follow-up now of 41.5 months (mos).
Aims: To assess longer-term safety and efficacy of ibr + obin in patients (pts) with
R/R CLL.
Methods: We conducted a phase 1b, investigator-initiated study of ibr + obin in R/R
CLL (NCT02537613). The primary objective was to assess safety of 3 different drug
sequencing regimens in cycle 1; the main secondary objective was to assess efficacy.
Pts were randomized 1:1:1 to either receive obin 1 mo before ibr, ibr 1 mo before
obin, or start concomitantly. Both drugs were given at the approved doses and schedule,
with 6 total cycles of combination followed by ibr monotherapy until progression or
unacceptable toxicity. Notable eligibility criteria: requiring treatment (tx) for
R/R CLL/SLL by 2008 iwCLL criteria, ECOG PS ≤2, no prior ibr or obin. Assessments:
toxicities by CTCAE v4, efficacy by 2008 iwCLL criteria, MRD by 4-8 color flow (10-4
sensitivity). Serum concentrations of 21 cytokines were assessed pre-tx and after
1 week of combination tx in a representative subset of 27 pts (n=9 per arm) by bead-based
multiplex immunoassay (Eve Technologies, Calgary).
Results: 52 pts were accrued, and all received ≥1 cycle of combination tx. Median
age: 67 yrs (range 33-84); 77% male; median # prior tx: 1 (range 1-6); 25% del(17p)
or TP53 mut; 27% del(11q); 50% unmutated IGHV; 27% bulky LAD (nodes ≥5 cm).
As of 14 Feb 2022: median follow-up 41.5 mos (range 2.3-73.3). 27 pts (52%) remain
on tx. All-grade heme toxicities: thrombocytopenia (88%; 23% Gr3/4), neutropenia (71%;
37% Gr3/4), febrile neutropenia (8%, all Gr3/4), anemia (73%; 8% Gr3). Notable non-heme
toxicities: bruising (58%, all Gr1/2), arthralgia (38%, all Gr1/2), diarrhea (37%,
all Gr1/2), transaminitis (35%; 4% Gr3), ≥Gr3 infection (17%, all Gr3), and cardiotoxicities:
hypertension (46%; 10% Gr3), atrial fibrillation (21%; 10% Gr3). 1 pt had sudden cardiac
death after 11 mos on study. 7 pts required ibr dose-reduction due to toxicity.
The best ORR is 96%, including 50% CR and 46% PR (Fig. 1A). All 3 pts who previously
progressed on venetoclax responded (1 CR, 2 PR). By ITT, 10/52 (19%) achieved a best
response of undetectable MRD in bone marrow (BM, 44 pts tested), and 14/52 (27%) in
peripheral blood (PB, 34 pts tested). 4-yr PFS and OS are 74% and 93% (Fig. 1B-C),
with 3 deaths (n=1 each: sudden death, MDS, Richter’s syndrome). Pts who achieved
BM and PB undetectable MRD had a significantly larger decrease in circulating CCL4
(p=0.02) and CXCL13 levels (p=0.01), respectively, comparing baseline to 1 week of
combination tx (Fig. 1D).
Image:
Summary/Conclusion: Ibr + obin is a highly active combination in R/R CLL, achieving
50% CR and 4-yr PFS/OS of 74%/93%, suggesting a potential positive impact of obin
(though possibly also reflecting an earlier relapse population than RESONATE). Responses
were observed in all 3 pts who had progressed after venetoclax, including 1 CR. With
~3.5 yrs median follow-up, and some pts on tx over 6 yrs, cardiotoxicity was consistent
with prior ibr studies, and no new safety concerns have emerged. Our data support
continued exploration of combination BTK inhibitor plus obin in R/R CLL.
P675: TREATMENT OF HYPERTENSION IN PATIENTS RECEIVING BTK INHIBITORS: A MULTICENTER
RETROSPECTIVE STUDY
M. Shadman1,2,*, J. Voutsinas3, B. Fakhri4, S. Khajavian3, S. Spurgeon5, D. Stephens6,
A. Skarnbik7, A. Mato8, C. Broome9, A. Gopal1,2, S. Smith1, R. Lynch1, M. Rainey10,
S. Kim5, O. Barrett-Campbell11, E. Hemond11, M. Tsang12, D. Ermann6, N. Malakhov13,
D. Rao8, M. Shakib Azar3, B. Morrigan3, A. Chauhan14, T. Plate15, T. Gooley1, K. Ryan16,
F. Lansigan11, B. Hill10, G. Pongas15, S. Parikh17, L. Roeker8, J. Allan18, R. Cheng2,
C. Ujjani1
1Fred Hutchinson Cancer Research Center / University of Washington; 2University of
Washington; 3Fred Hutchinson Cancer Research Center, Seattle; 4University of California
San Francisco (UCSF), San Francisco; 5Oregon Health and Science University (OHSU),
Portland; 6University of Utah, Salt Lake City; 7Novant Health, Charlotte; 8Memorial
Sloan Kettering Cancer Center, New York; 9Georgetown University, Washington DC; 10Cleveland
Clinic, Cleveland; 11Dartmouth-Hitchcock Medical Center, Lebanon; 12UCSF, San Francisco;
13New York-Presbyterian Hospital, Weill Cornell Medicine, New York; 14Georgia Cancer
Center at Augusta University, Agusta; 15Sylvester Comprehensive Cancer Center, University
of Miami, Miami; 16Astra Zeneca, Gaithersburg; 17Mayo Clinic, Rochester; 18Weill Cornell
Medicine, New York, United States of America
Background: Hypertension (HTN) is an adverse event (AE) associated with the use of
ibrutinib and less commonly with second generation BTK inhibitors (BTKis) as shown
by a significant lower rate of HTN with acalabrutinib compared to ibrutinib in the
ELEVATE-RR trial. Nevertheless, HTN is considered an AE of BTKis as a class.
Aims: We aimed to identify a class or combination of antihypertensive drugs (anti-HTNs)
that may be most effective in pts with HTN while taking a BTKi.
Methods: In this multicenter retrospective real-world study, we included consecutive
pts with lymphoid malignancies taking BTKis and anti-HTNs with at least 3 months of
follow-up. Eligible pts were stratified into 2 groups: 1) Pts with pre-existing HTN
before starting BTKi (pre-HTN) and 2) Pts who developed incident HTN after starting
BTKis (de novo HTN). The anti-HTN drugs were categorized as follows: 1) Angiotensin-converting-enzyme
inhibitors and Angiotensin receptor blockers (ACEi/ARB); 2) Beta-blockers (BB); 3)
Calcium channel blockers (CCB); 4) Hydrochlorothiazide (HCTZ) and 5) non-HCTZ diuretics
(other-DU). Timepoints for mean arterial pressure (MAP) measurements and start/end
time of each anti-HTN were documented in relation to the index date (first date of
concurrent use of BTKi and anti-HTN drug). Generalized estimating equations were used
to assess the association between time varying MAPs and the individual drug categories,
as well as the number of medication categories.
Results: We included 196 pts (118 pre-HTN and 78 de novo HTN) from 12 centers. The
majority of pts had CLL (94%) on ibrutinib (90%) for first (49%), second (25.5%) or
later (25.5%) lines of therapy. The median age was 67 (IQR 37-88), most pts were male
(72%), and caucasian (94.4%). The median duration of follow-up was 38.9 months (IQR
3.5-110). Pre-BTKi comorbidities included diabetes (17.3%), coronary disease (11.7%),
cerebrovascular accidents (2.6%) and chronic kidney disease (12.8%). BTKi dose was
reduced in 53 pts (27%) with 9 (4.6%) having dose reduction because of HTN. Twelve
pts (6.2%) switched to other BTKis and 5 pts (2.6%) did so for HTN. In the pre-HTN
cohort, a trend for statistically significant BP reduction was only achieved when
4+ anti-HTN drugs were used [Est -4.81; 95% CI (-9.93, 0.307); p = 0.07]. In this
cohort, concurrent use of HCTZ and BB showed a significant trend for a lower BP [Est
-5.05; 95% (-10.0, -0.06); p= 0.04]. In de novo HTN cohort, treatment with 3+ anti-HTN
drugs was associated with a statistically significant reduction in the BP [Est -5.70;
95% CI (-9.94, -1.46); p =0.008]. In de novo cohort combination of HCTZ and ACEi/ARB
was associated with a significant MAP reduction [Est -5.47; 95% CI (-10.9, -0.001);
p= 0.05]. When the 2 cohorts were combined, treatment with 4+ drugs [Est -6.27; 95%
CI (-11.8, -0.69); p = 0.02] and treatment with HCTZ and ACEi/ARB combination [Est
-5.05; 95% CI (-9.61, -0.48); p = 0.03] or HCTZ and BB [Est -5.05; 95% CI(-9.61,-0.48);
p= 0.03] were associated with MAP reduction. Gender or race did not modify these associations.
Image:
Summary/Conclusion: Treatment of HTN in pts taking Ibrutinib is challenging and requires
utilization of multiple anti-hypertensive drugs both in pre-HTN and de novo HTN pts.
Among different anti-HTN classes, we observed a statistically significant MAP reduction
in pts with pre-HTN on HCTZ and BB combination and in pts with de novo HTN on HCTZ
and ACEi/ARB combination. These findings need to be further assessed in pts taking
newer generation BTKi and confirmed in large prospective studies.
P676: LOW-DOSE FCR COMPARED TO BR IN PREVIOUSLY UNTREATED PATIENTS WITH CHRONIC LYMPHOCYTIC
LEUKEMIA WITHOUT TP53 ABERRATIONS: A REAL – WORLD RETROSPECTIVE ANALYSIS BY THE CZECH
CLL STUDY GROUP.
L. Smolej1,*, Y. Brychtová2, E. Cmunt3, M. Oršulová2, P. Vodárek1, M. Dostál1, P.
Turcsányi4, R. Urbanová4, A. Panovská2, J. Zuchnická5, J. Mihályová5, D. Lysák6, M.
Brejcha7, H. Móciková8, K. Klásková8, M. Šimkovič9, M. Špaček3, M. Doubek10
14th Department of Medicine - Hematology, University Hospital and Faculty of Medicine,
Hradec Králové; 2Department of Internal Medicine – Hematology and Oncology, University
Hospital, Brno; 3First Department of Internal Medicine - Hematology, General University
Hospital, Prague; 4Department of Hemato - oncology, University Hospital, Olomouc;
5Department of Hematooncology, University Hospital, Ostrava; 6Department of Hematology
- Oncology, University Hospital, Pilsen; 7Department of Hematology, Hospital Agel,
Nový Jičín; 8Department of Internal Medicine - Hematology, University Hospital Královské
Vinohrady, Prague; 94th Department of Medicine - Hematology, University Hospital,
Hradec Králové; 10Department of Internal Medicine - Hematology, University Hospital,
Brno, Czechia
Background: Low – dose fludarabine, cyclophosphamide, and rituximab (LDFCR) regimen
showed promising activity in treatment-naïve elderly / comorbid patients (pts) with
chronic lymphocytic leukemia (CLL) (Smolej et al., Br J Haematol. 2021). However,
no data are available regarding comparison to bendamustine and rituximab (BR), which
has been one of the predominant first - line options for CLL patients ineligible for
full – dose FCR.
Aims: To perform a historical comparison of the efficacy and safety of LDFCR and BR
regimens used as the first - line therapy of CLL within routine practice. Patients
with 17p deletion and/or TP53 mutation were not included in this analysis because
chemoimmunotherapy is no longer indicated in this extremely unfavourable subgroup
due to very low activity.
Methods: The analysis included 237 pts treated with LDFCR and 320 pts treated with
BR (median age, 69 vs 70 years; males, 70 vs 61%; median CIRS score, 6 vs 7; Rai stage
III/IV, 57 vs 59%; bulky lymphadenopathy > 5cm, 35 vs 35%; unmutated IGHV, 74 vs 70%,
deletion 11q, 28 vs 26%) treated between March 2009 and December 2019 at 17 centers
cooperating within the Czech CLL Study Group. LDFCR consisted of fludarabine 20 mg/m2
orally or 12 mg/m2 on days 1-3, cyclophosphamide 150mg/m2 orally or iv on days 1-3,
and rituximab 500 mg/m2 D1 (375 mg/m2 in Cycle 1). BR included bendamustine 90 mg/m2
iv on days 1-2 and rituximab 500 mg/m2 D1 (375 mg/m2 in Cycle 1). Median follow –
up was 98 months for LDFCR and 57 months for BR.
Results: Distribution of demographic data and prognostic factors was comparable between
LDFCR and BR groups. The overall response rate / complete remissions were similar
for LDFCR vs BR (84/46% vs. 88/51%, p=n.s.). The median progression – free survival
(PFS) was 30 vs 34 months (hazard radio [HR] 0.90; p=n.s.); PFS was markedly longer
in pts with mutated IGHV and absence of del 11q (LDFCR vs BR, median 58 vs 59 months,
HR 0.84; p=n.s.; Fig. 1a). Median overall survival (OS) was 73 vs 77 months for LDFCR
vs BR (HR 1.09; p=n.s.) but markedly longer (median 116 months vs not reached) in
patients with mutated IGHV without del 11q (HR 0.90; p=n.s.; Fig. 1b). Grade ≥ 3 neutropenia
occurred in 55% (LDFCR) vs 51% (BR) pts but serious infections developed in 15% vs
20% pts only.
Image:
Summary/Conclusion: Our data indicate that both low-dose FCR and BR are well – tolerated
regimens with similar efficacy and safety and represent a suitable first - line treatment
alternative for elderly / comorbid CLL patients with favourable biological prognosis.
Detailed results including multivariate analysis will be presented.
P677: CLINICAL EFFICACY AND TOLERABILITY OF VENETOCLAX PLUS RITUXIMAB IN RELAPSED
OR REFRACTORY CHRONIC LYMPHOCYTIC LEUKEMIA – REAL WORLD ANALYSIS OF POLISH ADULT LEUKEMIA
STUDY GROUP
A. Soboń1,*, J. Drozd-Sokołowska2, E. Paszkiewicz-Kozik3, L. Popławska3, M. Morawska4,5,
J. Tryc-Szponder6, L. Bołkun7, J. Rybka8, K. Pruszczyk1, A. Juda9, A. Majeranowski10,
E. Iskierka-Jażdżewska11, P. Steckiewicz12, K. Wdowiak13, B. Budziszewska1, K. Jamroziak2,
I. Hus1, E. Lech-Marańda1, B. Puła1
1Department of Hematology, Institute of Hematology and Transfusion Medicine; 2Department
of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw;
3Department of Lymphoid Malignancies, Maria Sklodowska-Curie National Research Institute
of Oncology, Warszawa; 4Experimental Hematooncology Department, Medical University
of Lublin & Hematology Department; 5St. John’s Cancer Center, Lublin; 6Department
of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences,
Poznań; 7Department of Hematology, Medical University of Białystok, Białystok; 8Department
of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University,
Wrocław; 9Department of Hematology and Bone Marrow Transplantation, Medical University
of Lublin, Lublin; 10Department of Hematology and Transplantology, Medical University
of Gdańsk, Gdańsk; 11Department of Hematology, Medical University of Łódź, Copernicus
Memorial Hospital, Łódź; 12Department of Hematology, Holy Cross Cancer Center, Kielce;
13Department of Internal Diseases and Oncological Chemotherapy Faculty of Medical
Sciences, Medical University of Silesia, Katowice, Poland
Background: The introduction of venetoclax into clinical practice has improved the
outcome of patients with relapsed/refractory chronic lymphocytic leukemia (RR-CLL).
The results of the MURANO trial published in March 2018 showed significantly longer
progression-free survival (PFS) and overall survival (OS) in RR-CLL patients treated
with venetoclax and rituximab (VEN-R) comparing to bendamustine and rituximab (BR)
and resulted in the approval of VEN-R in the therapy of RR-CLL in the European Union
and the United States. It should be noted that the results of registration studies
often do not correspond with the data from real-life observations.
Aims: To study the clinical efficacy and safety profile of VEN-R treatment in RR-CLL
patients outside clinical trials.
Methods: We performed retrospective analysis of RR-CLL patients treated with VEN-R
in hematology centers of the Polish Adult Leukemia Study Group (PALG) from 2019 to
2021.
Results: Clinical data of 117 RR-CLL patients treated with VEN-R were collected. Median
patient age upon initiation of VEN-R therapy was 67 years (range 33 – 84 years). Seventy-two
patients (61.5%) were men. Median Cumulative Illness Rating Scale (CIRS) was 6 (range
2 - 16). Patients were treated with a median of 2 (range 1–9) previous lines of therapy,
whereas 32 patients (27.4%) had relapsed following the first line of treatment. Overall,
25 patients (21.4%) had 17p deletion, whereas TP53 mutation was identified in 13 patients
(11.1%). The median follow-up was 9.96 months (range 0.27 - 29.13). The overall response
rate (ORR) was 95.2%. Seventeen patients (14.5%) achieved complete remission (CR),
83 (70.9%) partial remission (PR), while in 5 patients (4.3%) disease progression
was noted. In the patients with 17p deletion (n=22) or TP53 mutation (n=11), CR and
PR were observed in 4 (12.1%) and 29 (87.9%) patients, respectively. The median PFS
in the whole cohort was 20.8 (95% CI 18.43 - not reached) months and the median OS
was not reached. In our study none of the analyzed clinico-pathological factors had
significant impact on ORR, PFS and OS. During the follow-up time four (3.4%) cases
of Richter transformation were diagnosed. There were 18 deaths recorded during the
course of observation; 3 (16.7%) due to disease progression and 7 (38.9%) due to COVID-19
infection. The others were due to infections other than SARS-CoV-2 (n=3, 16.7%) and
the cause of death could not be specified in five cases (27.8%). Eighty-three patients
(70.9%) remain on treatment, while treatment was discontinued in thirty-four cases
(29.1%). Reasons for therapy discontinuation included patient’s death (52.9%), treatment-related
cytopenias (17.6%), disease progression (14.7%), Richter’s transformation (11.8%),
autoimmune hemolytic anemia (5.9%), diarrhea (2.9%) and infections (8.8%). In one
case treatment discontinuation was due to consent withdrawal and one patient was lost
to follow-up. The following adverse events of VEN-R treatment were reported during
the study: all grade neutropenia (71.8% with grade 3/4 in 55.6%), anemia (51.3%),
thrombocytopenia (47%), pneumonia (9.4%), neutropenic fever (6.8%), autoimmune hemolytic
anemia (4.3%), immune thrombocytopenic purpura (1.7%), diarrhea (4.3%) and in one
case exacerbation of heart failure was observed.
Summary/Conclusion: In this retrospective analysis the outcomes of treatment with
the VEN-R regimen in real-life setting were worse than those reported in the MURANO
trial.
P680: SARCOPENIA EVALUATED BY CT SCAN IS ASSOCIATED WITH SHORTER SURVIVAL IN PATIENTS
WITH CHRONIC LYMPHOCYTIC LEUKEMIA TREATED WITH TARGETED THERAPIES. A PROSPECTIVE STUDY.
A. Visentin1,*, F. Crimì2, A. Corso2, R. Angelone2, C. A. Cavarretta1, A. Cellini1,
F. Angotzi1, V. Ruocco1, S. Pravato1, E. Quaia2, L. Trentin1
1Hematology and Clinical Immunology unit, Department of Medicine, University of Padova;
2Radiology Institute, Department of Medicine, University of Padova, Padova, Italy
Background: Chronic lymphocytic leukemia (CLL) usually affects elderly patients with
comorbidities. Although drugs targeting BTK, BCL2 and PI3K have been proved to be
better tolerated than chemoimmunotherapy and to improve the survival of CLL patients,
comorbidities, assessed by Cumulative Illness Rating Scale (CIRS) score, are still
associated with a worse outcome. Indexes of sarcopenia, subcutaneous or visceral adipose
tissues assessed by CT scan, are emerging as survival markers in cancer patients.
However, CT scan is not recommended by iwCLL guideline nor we known the clinical impact
of body composition in CLL.
Aims: The aim of this study was to assess the impact of muscle and adipose tissue
indexes in patients with CLL treated with targeted agents.
Methods: We performed a single center prospective study of CLL patients treated with
BTK, BCL2 and PI3K inhibitors out of clinical trials. Inclusion criteria were diagnosis
of CLL and need of treatment according to iwCLL guideline, creatinine clearance >30ml/min
and CT scan performed at baseline. For each patient, with NIH ImageJ software, we
extracted the areas of subcutaneous fat, visceral fat and the skeletal muscle from
an unenhanced axial CT image at vertebra L3. The values obtained (cm2) were divided
by the square of patient’s height (m2), obtaining the subcutaneous adipose tissue
index (SATI), the visceral adipose tissue index (VATI) and the skeletal muscle index
(SMI). Overall survival (OS) was calculated as months from starting targeted therapies
to death (event) or last available follow-up (censored). Survival curves with Log-rank
test.
Results: We included 118 patients, 70 (59%) were males, mean age was 71.6±8.6 years,
74 (63%) were at Rai stage III-IV, 50 (42%) had a CIRS≥6, 96 (81%) unmutated IGHV,
41 (35%) 17p13 deletion, 39 (33%) TP53 mutation. The median number of therapies was
3 (range 1-9). Sixty-six patients (56%) received ibrutinib, 27 (23%) idelalisib+rituximab
and 25 (21%) venetoclax±rituximab, including 17 (14%) front line therapies (13 ibrutinib,
4 venetoclax).
We observed that SMI values correlated with SATI (p=0.035) and VATI (p<0.0001). SMI,
SATI and VATI were higher in males vs females, median 48.4 vs 42.2 (p=0.007), 61.0
vs 67.9 (p=0.0087) and 70.0 vs 50.3 (p=0.0113)(Fig 1A). Using these thresholds, low-SMI,
suggestive of sarcopenia, and low SATI correlated with a more advance number of previous
therapies (p=0.027 and p=0,037). Low-SMI was more common in patients with CIRS≥6 (p=0.039).
SMI, SATI and VATI did not correlated with age, stage and biological markers.
After a median follow-up of 32 months the median OS for the whole cohort was 46.7
months. We confirmed that CIRS≥6 correlated with a worse OS in our study (p<0.0001,
Fig. 1B). The 3-year OS was 53% and 66% for patients with low-SMI vs high-SMI, and
the median OS was 40.3 months for low-SMI but not reached for high-SMI patients (p=0.0199,
Fig. 1C). SATI and VATI did not correlate with OS.
Combining data of CIRS≥6 and low-SMI, 40 (34%) patients, 47 (40%) and 31 (26%) displayed
0, 1 and 2 markers (Fig. 1D). The 3-year OS was 80%, 56% and 42% for patients at score
0, 1 and 2. The median OS was not reached for score 0 patients, but decreased from
42 to 23 months for patients at score 1 and 2 (p=0.0006, Fig. 1E).
Image:
Summary/Conclusion: In this prospective study we found that that basal CT scan identifies
sarcopenic CLL patients with low SMI featured by an adverse prognosis even if treated
with targeted oral drugs, in particular when combined with the CIRS score. The prognostic
and predictive impact of basal CT scan in CLL deserves further investigation.
P681: EUROFLOW STANDARDIZATION TECHNIQUE AND NORMALIZATION PROCEDURES IN LONGITUDINAL
FLOW CYTOMETRIC EXPRESSION ANALYSIS OF CD20 IN CLL PATIENTS RECEIVING ANTI-CD20 DIRECTED
THERAPY
P. J. Walter1,*, A. Schilhabel1, P. Cramer2, J. von Tresckow3, S. Kohlscheen1, K.
Fischer2, B. Eichhorst2, S. Böttcher4, M. Brüggemann1, M. Kneba1, M. Hallek2, M. Ritgen1
1Laboratory for Specialized Hematological Diagnostics, Medical Department II, University
Hospital Schleswig-Holstein, Kiel; 2Department I of Internal Medicine, Center for
Integrated Oncology Aachen Bonn Cologne Duesseldorf, German CLL Study Group, University
Hospital Cologne, Cologne; 3Department of Hematology and Stem Cell Transplantation,
West German Cancer Center, University Hospital Essen, University of Duisburg-Essen,
Essen; 4Medical Department III Hematology, Oncology, Palliative Care, Center for Inner
Medicine, University Hospital Rostock, Rostock, Germany
Background: CD20 expression is still a controversial issue regarding its prognostic
value for therapy outcome. While up to 50% of patients relapse within four of years
after anti-CD20 directed treatment and constitute a need for a salvage or relapse/refractory
treatment, only few publications address the longterm influence on CD20 expression
assessed by flow cytometry. The technical complexity of the method and factors affecting
the variability of measurements of samples at different time points and from different
sources/patients are well recognized by the field and addressed by different approaches
as standardization of instruments, automated gating and alignment of population or
normalization.
Aims: In order to evaluate long term influence of anti-CD20 directed therapy on CD20
expression in patients with chronic lymphocytic leukemia who showed only weak MRD
responsiveness to therapy in different clinical trials, we aimed to establish a normalization
approach based on the instrument standardization to reduce the proportion of technical
variance within data.
Methods: Mean fluorescence intensities (MFI) of peak seven of standardized fluorescence
beads from daily quality control and instrument standardization of flow cytometers
according to the EuroFlow protocol were used to establish a normalization approach.
Proof of prinicple was performed using the residual peaks covering the MFI range provided
by the beads. To assess different sources of variance ANOVA of a 3x3x3 experiment
evaluating marker expressions on different cell populations in the blood of healthy
donors using Quantibrite-PE beads was performed addressing technical variation (day,
instrument) and biological variation. CD20 and CD19 MFI on CLL cells were retrospectively
assessed from MRD measurements of peripheral blood samples of patients enrolled in
the phase II GCLLSG trials CLL2-BIG (n=57), -BAG (n=58), -BIO (n=58), -BCG (n=22),
combining facultative Bendamustin (B) debulking with either Obinutuzumab (G), Ofatumumab
(O), Venetoclax (A), Ibrutinib (I) or Idelalisib (C).
Results: Long term shifts of fluorescence intensities, and coefficients of variation
were reduced 2 to 10 times across the MFI range covered by the fluorescence beads
without distorting the longitudinal course of instrument performance. Most of the
variance in assessments of molecules equivalent soluble fluorochrome from MFIs of
NK cell and B cell markers in peripheral blood of healthy donors were related to inter-donor
variations (p<0.002). Normalized MFIs for CD19 and CD20 on CLL cells do not significantly
differ from the measured values in the Mann-Whitney-U test (p>0.85). Higher CD20 expression
at therapy start seems to be correlated to strong MRD response solely in the CLL2-BIO
trial (p=0.036, Fig. 1). Prognostic factors TP53-, IGHV-, and NOTCH1- mutation did
not seem to correlate with the observed CD20 expression differences. Therapy strata
showed differences (statistically not significant) in the proportion of firstline
and r/r treated patients.
Figure 1 Pre-treatment CD20 MFI on CLL cells of patients categorized according to
treatment and variable (strong, medium, weak, and no) MRD response.
Image:
Summary/Conclusion: Following standardized staining and instrument monitoring, it
should be possible to robustly assess longitudinal biological variations of marker
expression based on MFI values. In a cross trial comparison Obinutzumab showed highest
proportion of patients with strong MRD response independent from initial CD20 expression,
whereas strong response to Ofatumumab seems to correlate with higher CD20 expression
levels.
P682: NEMTABRUTINIB (MK-1026), A NON-COVALENT INHIBITOR OF WILD-TYPE AND C481S MUTATED
BRUTON TYROSINE KINASE FOR B-CELL MALIGNANCIES: EFFICACY AND SAFETY OF THE PHASE 2
DOSE-EXPANSION BELLWAVE-001 STUDY
J. Woyach1,*, I. W. Flinn2, F. T. Awan3, H. Eradat4, D. Brander5, M. Tees6, S. A.
Parikh7, T. Phillips8, W. Wang9, N. M. Reddy10, M. Z. Farooqui10, J. C. Byrd11, D.
M. Stephens12
1Division of Hematology, The Ohio State University, Columbus, Columbus; 2Sarah Cannon
Center Research Institute, Nashville, Nashville; 3Department of Internal Medicine,
University of Texas Southwestern Medical Center, Dallas; 4Department of Hematology-Oncology,
David Geffen School of Medicine at UCLA, Los Angeles; 5Duke Cancer Institute, Duke
University Medical Center, Durham; 6Colorado Blood Cancer Institute, Denver; 7Division
of Hematology, Mayo Clinic, Rochester; 8Division of Hematology and Oncology, University
of Michigan Rogel Cancer Center, Ann Arbor; 9Veristat, LLC, Southborough; 10Merck
& Co., Inc., Kenilworth; 11Department of Internal Medicine, University of Cincinnati
College of Medicine, Cincinnati; 12Division of Hematology and Hematologic Malignancies,
University of Utah Huntsman Cancer Institute, Salt Lake City, United States of America
Background: For patients (pts) with chronic lymphocytic leukemia/small lymphocytic
lymphoma (CLL/SLL) and certain B-cell neoplasms, resistance to Bruton tyrosine kinase
inhibitors (BTKi) develops primarily through mutations at the cysteine binding site
(C481) or PLCγ2 mutations. Nemtabrutinib (MK-1026, formerly ARQ-531) is a non-covalent,
potent inhibitor of both wild type and C481-mutated BTK.
Aims: In the phase 1/2 dose-escalation and dose-expansion BELLWAVE-001 study (NCT03162536),
the preliminary recommended phase 2 dose (RP2D) of nemtabrutinib was determined to
be 65 mg once daily. The efficacy and safety of nemtabrutinib in patients (pts) with
CLL/SLL and B-cell non-Hodgkin lymphoma (NHL) were also evaluated at a higher dose
during the dose-expansion phase.
Methods: In this open-label, single-arm phase 2 study, 9 expansion cohorts were initiated
following determination of preliminary nemtabrutinib RP2D. Approximately 10-25 eligible
pts were enrolled into Cohort A (relapsed/refractory (r/r) CLL/SLL, with ≥2 prior
therapies including covalent BTKi, with documented C481 mutation), Cohort B (r/r CLL/SLL
progressed on/intolerant to a BTKi, with ≥2 prior therapies without C481 mutation),
Cohort C (Richter transformation with ≥1 prior therapy), Cohort D-H (follicular lymphoma,
mantle cell lymphoma, marginal zone lymphoma, high-grade B cell lymphoma with known
MYC, BCL-2 or BCL-6 translocations, and Waldenström macroglobulinemia [WM], respectively,
who received ≥2 prior therapies), Cohort I (food-effect cohort including pts with
B-cell NHL, CLL/SLL, and WM). Primary end point was ORR (per iwCLL criteria, by investigator)
for participants with CLL/SLL. Secondary end points included DOR and safety and tolerability.
Results: Among 118 pts enrolled, 44 had B-cell NHL, 68 CLL/SLL, and 4 WM. Of these,
94 (79.6%) were treated at the preliminary RP2D, including 51 (54.3%) participants
with CLL/SLL. Pts with CLL/SLL had a median (range) number of prior therapies of 4
(1-18), 43 (84%) had prior BTKi therapy, 12 (23%) had del17p, 30 (59%) had IGHV unmutated
status, and 32 (63%) had C481S BTK mutation. At data cut-off (April 7, 2021), median
(range) follow-up was 4.6 mo (0.1-26.7) for all treated pts. ORR was 57.9% (22/38;
1 CR, 21 PR/PRL) per iwCLL criteria in the efficacy-evaluable population of pts with
CLL/SLL treated at preliminary RP2D. Median duration of response was not estimable
[NE] (range, 8.3 mo-NE). Among all treated participants, 114 (97%) had a treatment-emergent
adverse event (TEAE), with 78 (66%) having a drug-related TEAE, and 9 (8%) having
a drug-related TEAE that led to discontinuation. Common TEAEs (≥ 20%) included fatigue
(33%), constipation (31%), dysgeusia (28%), cough (25%), nausea (25%), pyrexia (25%),
dizziness (23%), hypertension (23%), peripheral edema (22%), diarrhea (21%), and arthralgia
(20%). Grade ≥3 TEAEs occurred in 80 (68%) participants. Grade 5 TEAEs occurred in
7 (6%) participants and included death following disease progression 3 (3%), sepsis
1 (1%), dyspnea 1 (1%), and respiratory failure 2 (2%). Common drug-related TEAEs
(≥10%) included dysgeusia (15%), nausea (11%), fatigue (11%), and decreased neutrophil
count (10%). Grade 3-4 drug-related TEAEs occurred in 31 (26%) participants. No drug-related
TEAEs led to death.
Summary/Conclusion: Nemtabrutinib has promising antitumor activity with a manageable
safety profile in pts with CLL/SLL exposed to multiple lines of therapy, including
in those who had progression of disease on prior covalent BTKi. Further evaluation
of nemtabrutinib in B-cell malignancies is ongoing.
P683: A FIRST-IN-HUMAN PHASE 1 STUDY OF ORAL LOXO-338, A SELECTIVE BCL2 INHIBITOR,
IN PATIENTS WITH ADVANCED HEMATOLOGIC MALIGNANCIES (TRIAL IN PROGRESS)
W. Jurczak1,*, A. J. Alencar2, G. S. Guru Murthy3, M. Hoffmann4, J. M. Pagel5, V.
Patel5, J. M. Pauff5, P. L. Zinzani6, S. Le Gouill7, A. R. Mato8, L. E. Roeker8
1Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, Poland; 2Sylvester
Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami,
FL; 3Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI;
4Hematologic Malignancies and Cellular Therapeutics, University of Kansas Cancer Center,
Overland Park, KS; 5Loxo Oncology at Lilly, Stamford, CT, United States of America;
6Institute of Hematology Seràgnoli, University of Bologna, Bologna, Italy; 7Institut
Curie Hospital, Paris, France; 8Memorial Sloan Kettering Cancer Center, New York,
NY, United States of America
Background: B-cell lymphoma 2 (BCL2) is a key regulator of apoptosis and provides
protection from cell death in many hematological malignancies. LOXO-338 is a novel,
orally bioavailable small molecule inhibitor of BCL2, designed to achieve selectivity
over BCL-xL and thus avoid dose-limiting thrombocytopenia associated with BCL-xL inhibition.
In preclinical studies, LOXO-338 showed a favorable pharmacological profile, selectively
inhibited BCL2, and was well-tolerated in vivo. LOXO-338 also demonstrated dose-dependent
tumor growth inhibition in various murine xenograft models, and showed improved efficacy
in combination with pirtobrutinib, a highly selective, non-covalent (reversible) BTK
inhibitor (Brandhuber et al. Cancer Res 2021;81,13 Suppl:1258).
Aims: To determine the recommended phase 2 dose of LOXO-338 and evaluate the safety
profile of LOXO-338 as monotherapy and in combination.
Methods: LOXO-BCL-20001 is a global, open-label, multi-center, first-in-human phase
1 study of oral LOXO-338 in patients with advanced hematologic malignancies who have
received standard therapy (NCT05024045). The study will be conducted in 2 parts. Part
1 will evaluate LOXO-338 as monotherapy, and will explore different dosing strategies.
Part 2 will evaluate LOXO-338 in combination with pirtobrutinib. Part 1 dose escalation
portion of the study will follow an i3 + 3 design. Each cycle will be 28 days (4 weeks).
Eligible patients include those with CLL/SLL, mantle cell lymphoma, and Waldenstrӧm
macroglobulinemia (WM) who received standard therapy. Patients with other B-cell non-Hodgkin
lymphomas (NHL) who failed standard therapy or, in the opinion of the investigator,
have no known available options to provide benefit for the patient’s condition, are
also eligible. Patients must have recovered from prior treatment-related adverse events.
Patients with active or suspected Richter transformation, transformed low grade lymphoma,
Burkitt or Burkitt-like lymphoma, and multiple myeloma are also eligible. Patients
with AL amyloidosis are eligible in monotherapy dose expansion; all other patients
are eligible in both monotherapy and combination dose expansion. Patients must not
have progressed while receiving prior BCL2 inhibitor; those with WM or AL amyloidosis
must not have received a prior BCL2 inhibitor. Key exclusion criteria include history
of CNS involvement, stem cell transplant or CAR-T therapy <60 days, concurrent anticancer
therapy, and clinically significant cardiovascular disease.
The primary objective of part 1 is to determine the recommended phase 2 dose of oral
LOXO-338 in patients who were previously treated for CLL/SLL and other B-cell NHL,
administered alone or in combination with pirtobrutinib. Determining antitumor activity
in patients with WM and AL amyloidosis is an additional primary objective of part
1 monotherapy. Secondary objectives include determining the safety profile and tolerability,
PK properties and antitumor activity of LOXO-338 in combination with pirtobrutinib.
Antitumor activity will be evaluated based on overall response rate (ORR), progression-free
survival (PFS), time to progression (TTP) and duration of response (DOR) based on
disease-specific response criteria per investigator assessment.
Results: N/A
Summary/Conclusion: N/A
P684: PHASE 1 STUDY OF LP-108, A SELECTIVE BCL-2 INHIBITOR, IN PATIENTS WITH RELAPSED
OR REFRACTORY B-CELL NON-HODGKIN LYMPHOMA
W. Xu1,*, R. Feng2, L. Wang1, Y. Song3, H. Zhu1, L. Fan1, X. Guo2, X. Wei2, N. Lin3,
C. Xu4, Z. Wang5, X. Xiao5, Y. Shen5, Y. Chen6, Y. Chen6, F. Tan5, S. P. Anthony6,
J. Li1
1Department of Hematology, The First Affiliated Hospital of Nanjing Medical University,
Jiangsu Province Hospital, Nanjing; 2Department of Hematology, Nanfang Hospital of
Southern Medical University, Guangzhou; 3Department of Lymphoma, Beijing Cancer Hospital,
Beijing; 4Phase I Unit, Nanfang Hospital of Southern Medical University; 5Department
of Clinical Development, Guangzhou Lupeng Pharmaceutical Co., Ltd., Guangzhou, China;
6Department of Clinical Development, Newave Pharmaceutical Inc., Pleasanton, United
States of America
Background: Targeting BCL-2 family proteins is a well-recognized therapeutic approach.
Globally approved BCL-2 inhibitor venetoclax has shown efficacies in certain hematologic
malignancies (HMs) but requires weekly ramp-up to the target dose to mitigate the
risk of tumor lysis syndrome (TLS). LP-108 is a novel, highly potent, orally bioavailable,
and selective BCL-2 inhibitor that has shown promising preclinical antitumor activity
in various HMs.
Aims: To present preliminary results from an ongoing phase 1 study of LP-108 under
daily dose ramp-up in patients with relapsed or refractory (r/r) B-cell non-Hodgkin
lymphoma (NHL) (NCT04356846).
Methods: This is an open-label, multicenter, phase I study to assess the safety, pharmacokinetics
(PK) and preliminary efficacy of LP-108 in Chinese patients with r/r B-cell NHL, consisting
of a dose-escalation phase and a dose-expansion phase. Adult r/r B-cell NHL patients
requiring therapy are eligible if they had received at least one prior line of treatment
(except BCL-2i). LP-108 is orally administered once daily in 28-day cycles until disease
progression, unacceptable toxicity, or per investigator discretion. All patients provided
and signed informed consent.
Results: As of February 18, 2022, 19 patients have been enrolled and were evaluable
for safety, with disease types of MCL (n = 7), CLL/SLL (n = 7), DLBCL (n = 2), FL,
WM, and MALT (n = 1 each), and median age of 61 years (range, 39 to 83). Patients
had received a median of 3 prior lines of therapy (range, 1~6), of which 13 (68%)
patients were previously BTKi exposed. No dose-limiting toxicity (DLT) has been observed
in patients who were treated with LP-108 up to 600 mg, thus the maximum tolerated
dose (MTD) was not identified. With median duration of treatment of 55 days (range,
4~472), treatment-related adverse events (TRAEs) were reported in 17 patients (89.4%),
most of which were grade 1 to 2 in severity. TRAEs of any grade occurred ≥20% of patients
were neutropenia (36.8%), leukopenia (31.5%), ALT elevation (31.5%), AST elevation
(26.3%), hyperphosphatemia (21%), hyperuricemia (21%), and diarrhea (21%). Grade 3
or 4 TRAEs were reported in 7 patients (36.8%), including neutropenia, thrombocytopenia,
lymphopenia, laboratory TLS, and rash. Serious TRAEs included rash, thrombocytopenia,
and laboratory TLS, each occurring in 1 patient. No patient discontinued treatment
due to toxicity, and no clinical TLS was observed. No deaths occurred during treatment
or within 28 days of the end of treatment. At doses ≥200 mg, 3 of 10 efficacy evaluable
patients achieved an objective response (1 CR and 2 PR), 4 patients had stable disease
(2 with substantially reduced lymphomegaly), and 3 patients had progressive disease.
Rapid reduction in ALC was observed in CLL patients (pending imaging assessment) during
ramp-up and the first cycle of treatment at doses as low as 20mg. At data cutoff,
11 patients remain on treatment and 8 patients discontinued treatment due to disease
progression or patient decision. The PK profile showed that plasma exposure proportionally
increased with doses ranging from 20 to 600 mg (mean AUC24h range, 1.26~46.5 h*μg/ml,
mean Cmax range, 0.087~2.67 μg/ml). Plasma concentrations peaked approximately 4 to
10 hours after dosing, and the average terminal half-life of LP-108 was approximately
11.8 hours (range, 6.9~16.6).
Image:
Summary/Conclusion: LP-108 showed favorable safety profile up to 600 mg with daily
dose ramp-up schedule, and antitumor activity in patients with r/r B-cell NHL. Dose
escalation beyond 600 mg and further dose expansion are ongoing.
P685: REAL WORLD EVIDENCE OF IMPACT OF ATRIAL FIBRILLATION ON CLINICAL AND ECONOMIC
OUTCOMES IN PATIENTS WITH CHRONIC LYMPHOCYTIC LEUKEMIA
K. Yang1, T. Liu2, B. Tang1, A. Chanan-Khan3,*
1Beigene USA, San Mateo; 2Beigene USA, Emeryville; 3Mayo Clinic, Jacksonville, United
States of America
Background: While the incidence of atrial fibrillation (AF) in chronic lymphocytic
leukemia (CLL) has been increasing, the implications of AF in real-world CLL patients
remain understudied.
Aims: This study aimed to assess the impact of AF on clinical and economic outcomes
in CLL patients.
Methods: This retrospective observational study used the US IBM MarketScan® commercial
claims/Medicare supplement database (2017-2020) to identify newly diagnosed CLL patients
(≥18 years) and incidence of AF after index date, defined as the first CLL diagnosis
date. Patients were followed for ≥3-months pre-index, and from index to last follow-up
or death. In addition to patient characteristics, clinical outcomes (incidence of
heart failure [HF], bleeding and stroke) and economic outcomes (costs and healthcare
resource utilization [HRU]) were compared between CLL patients with AF (CLL+AF) and
CLL patients without AF (CLL-AF). HRU was evaluated by inpatient, outpatient, ER,
and pharmacy visits. Multivariable regression analyses were conducted to examine the
association between AF and clinical outcomes.
Results: Among a total of 16,800 newly diagnosed CLL patients included in the study,
20% developed AF. CLL+AF were significantly older than CLL-AF (median: 77 vs 62 years;
P < .001). Compared to CLL-AF, CLL+AF also had significantly more comorbidities at
baseline, as shown by higher Charlson comorbidity index (CCI) (median: 1.0 vs 3.0;
P < .001) and more patients with previous AF history (0.3% vs 8.4%; P < .001). Further
assessing clinical outcomes in CLL+AF vs CLL-AF, significantly higher incidence of
HF (26.3% vs 3.0%; P < .001), bleeding (12.2% vs 4.8%; P < .001) and stroke (6.6%
vs 1.2%; P < .001) were observed. For HRU, CLL+AF were reported to have significantly
higher rates of ER visits (29.4% vs 12.9%; P < .001) and hospitalizations (42.2% vs
14.5%; P < .001) than CLL-AF. In CLL+AF, the average total AF-related costs were $13,520.21
within 30 days after AF diagnosis, and $22,304.82 within 60 days after AF diagnosis.
Controlling for demographics and comorbidities, multivariable regressions reported
statistically significant associations between AF and HF, as well as AF and stroke
(Table).
Image:
Summary/Conclusion: This real-world study reported significantly higher incidence
of HF, bleeding and stroke incurred by CLL patients who developed AF compared with
those who did not. The presence of HF, bleeding and stroke further increased HRU and
costs. These findings highlight the importance of better disease management and treatment
selection to prevent AF in CLL patients.
P686: A PHASE 1 FIRST IN-HUMAN STUDY OF BGB-16673, A BRUTON TYROSINE KINASE PROTEIN
DEGRADER, IN PATIENTS (PTS) WITH B-CELL MALIGNANCIES (TRIAL IN PROGRESS)
C. S. Tam1,2,3,*, C. Cheah4,5,6, D. A. Stevens7, K. By8, X. Chen8, B. Tariq8, G. S.
Vosganian8, J. Huang8, M. Alwan9
1Peter MacCallum Cancer Centre, Melbourne, Victoria; 2University of Melbourne; 3Royal
Melbourne Hospital, Parkville, Victoria; 4Department of Haematology, Sir Charles Gairdner
Hospital and Pathwest Laboratory Medicine, Nedlands, Western Australia; 5Medical School,
University of Western Australia, Crawley, Western Australia; 6Linear Clinical Research,
Nedlands, Western Australia, Australia; 7Norton Cancer Institute, Louiseville, KY;
8BeiGene (Beijing) Co., Ltd., Beijing, China and BeiGene USA, Inc., San Mateo, CA,
United States of America; 9Perth Blood Institute, West Perth, Western Australia, Australia
Background: Bruton tyrosine kinase (BTK) functions downstream of the B-cell antigen
receptor (BCR) and plays a critical role within the BCR signaling pathway and the
pathogenesis of several B-cell malignancies. BTK inhibitors (BTKi) have revolutionized
management of B-cell malignancies. However, resistance caused by BTK mutations, which
can abrogate BTKi binding capacity or BTK scaffold function, or cause kinase hyper
activation, may limit therapeutic options in subsequent lines of therapy. Re-challenging
patients with agents that can overcome the resistance due to BTK mutations may provide
a novel treatment option. BGB-16673 is an investigational, orally available agent
with preclinically demonstrated BTK degradation activity against both wild type and
mutant forms commonly identified in pts who have progressed on BTKi.
Aims: In the dose-escalation part of the study, we aim to assess the safety and tolerability
of BGB-16673 in selected relapsed or refractory (R/R) B-cell malignancies, to characterize
its pharmacokinetic (PK) and pharmacodynamic (PD) profiles, to determine a recommended
phase 2 dose (RP2D) and to evaluate anti-tumor activity. In the dose-expansion part
of the study, we aim to evaluate the safety, tolerability, PK, PD, and anti-tumor
activity under the RP2D in pts with chronic lymphocytic leukemia (CLL)/small lymphocytic
lymphoma (SLL) and mantle cell lymphoma (MCL).
Methods: BGB-16673-101 (NCT05006716) is a phase 1 open-label, dose-escalation and
-expansion study evaluating BGB-16673 in adult pts with R/R B-cell malignancies. Key
inclusion criteria include confirmed diagnosis of R/R B-cell malignancy (including
marginal zone lymphoma, follicular lymphoma, MCL, CLL/SLL, and Waldenström macroglobulinemia
[WM]), Eastern Cooperative Oncology Group performance status score of 0 to 2, and
adequate organ function. Key exclusion criteria include current or history of central
nervous system involvement, autologous stem cell transplant (ASCT) within 3 months
or chimeric antigen receptor T cell therapy or allogeneic SCT within 6 months of the
first dose of BGB-16673 or requiring treatment with strong inhibitors or inducers
of CYP3A. All pts will be followed for safety and tolerability, including treatment-emergent
adverse events that occur during treatment and up to 30 days after treatment discontinuation,
or until the initiation of another anti-cancer therapy, whichever occurs first. The
totality of the available safety, efficacy, PK, and PD data from the dose-escalation
part will be used by the Safety Monitoring Committee to determine the RP2D. The dose-expansion
part will commence subsequent to RP2D determination. Responses will be evaluated per
the 2014 Lugano Classification, the 2018 International Workshop on CLL guidelines
response assessment with modification for treatment-related lymphocytosis, or the
6th International Workshop on WM consensus criteria. Additional efficacy analyses
will include progression-free survival and overall survival. All pts will give informed
consent.
Results: This is a trial in progress; safety and tolerability results of BGB-16673
are expected.
Summary/Conclusion: BGB-16673-101 is the first in-human study of the BTK degrader
BGB-16673.
P687: A PHASE 1 STUDY WITH THE NOVEL B-CELL LYMPHOMA 2 (BCL2) INHIBITOR BGB-11417
AS MONOTHERAPY OR IN COMBINATION WITH ZANUBRUTINIB (ZANU) IN PATIENTS (PTS) WITH B-CELL
MALIGNANCIES: PRELIMINARY DATA
S. Opat1,2,*, C. Y. Cheah3,4,5, M. Lasica6, E. Verner7,8, P. J. Browett9, H. Chan10,
J. D. Soumerai11, E. González Barca12, J. Hilger13, Y. Fang13, J. Huang13, D. Simpson13,
C. S. Tam14,15,16
1Monash Health, Clayton; 2Monash University, Clayton, Victoria; 3Sir Charles Gairdner
Hospital and Pathwest Laboratory Medicine, Nedlands; 4Medical School, University of
Western Australia, Crawley; 5Linear Clinical Research, Nedlands, Western Australia;
6St. Vincent’s Hospital Melbourne, Fitzroy, Victoria; 7Concord Repatriation General
Hospital, Concord; 8University of Sydney, Sydney, New South Wales, Australia; 9Auckland
City Hospital; 10North Shore Hospital, Auckland, New Zealand; 11Massachusetts General
Hospital Cancer Center, Harvard Medical School, Boston, MA, United States of America;
12Institut Català d’Oncologia-Hospitalet, IDIBELL, Universitat de-Barcelona, Barcelona,
Spain; 13BeiGene (Beijing) Co., Ltd., Beijing, China and BeiGene USA, Inc., San Mateo,
CA, United States of America; 14Peter MacCallum Cancer Centre, Melbourne; 15University
of Melbourne, Parkville; 16Royal Melbourne Hospital, Parkville, Victoria, Australia
Background: BCL2 is aberrantly expressed in many hematologic malignancies and promotes
tumorigenesis by enabling cells to evade apoptosis. BCL2 inhibitors have an established
role in the treatment of chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL)
and acute myeloid leukemia. The currently approved BCL2 inhibitor, venetoclax, is
associated with mild gastrointestinal toxicities, neutropenia, and the development
of BCL2 mutations leading to resistance. BGB-11417 is a highly selective inhibitor
of BCL2 with superior potency to venetoclax in human acute lymphoblastic leukemia,
mantle cell lymphoma (MCL) cell lines, and xenograft model of diffuse large B-cell
lymphoma (DLBCL). BGB-11417 has favorable pharmacokinetics with excellent bioavailability
and selectivity for BCL2. Toxicology studies have shown a broad therapeutic index
and an improved safety profile.
Aims: BGB-11417-101 is an ongoing first-in-human phase 1/1b dose-escalation and expansion
study (NCT04277637) evaluating the safety, tolerability, maximum tolerated dose (MTD),
and recommended phase 2 dose of oral BGB-11417 as monotherapy or in combination with
the BTK inhibitor ZANU, in pts with B-cell malignancies.
Methods: In separate monotherapy and combination therapy dose-escalation cohorts,
pts with relapsed/refractory (R/R) B-cell malignancies received escalating doses of
BGB-11417 (40, 80, 160, 320, or 640 mg once daily [QD]) with a weekly or daily ramp-up
to intended target dose. Pts in the combination therapy cohorts received ZANU (320 mg
QD or 160 mg twice daily) 8-12 weeks before BGB-11417. Dose-limiting toxicity for
each dose cohort was evaluated by a Bayesian logistic regression model during dose
ramp-up through day 21 at intended dose. Adverse events (AEs) were reported per Common
Terminology Criteria for AEs v5.0.
Results: As of 17 Dec 2021, 58 pts received BGB-11417 (32 monotherapy; 26 combination).
Of the pts receiving monotherapy, 26 with R/R non-Hodgkin lymphoma (NHL; 17 DLBCL,
6 follicular lymphoma, and 3 marginal zone lymphoma) received BGB-11417 ≤640 mg and
6 with R/R CLL/SLL received ≤160 mg. Of the pts receiving combination treatment, 19
with R/R CLL/SLL received BGB-11417 ≤160 mg and 7 with R/R MCL received ≤80 mg. MTD
has not yet been reached. Median follow-up was 3.9 months (range, 0.1-20.4). AEs across
all dose levels are listed in the table. Only 2 grade ≥3 AEs (1 neutropenia, 1 autoimmune
hemolytic anemia) were reported in combination cohorts. Twenty pts discontinued treatment
(17 disease progression; 1 AE; 2 other reasons). One high-risk pt with CLL on monotherapy
had laboratory tumor lysis syndrome (TLS) that resolved with no intervention (laboratory
TLS <2%). Efficacy data are early: most pts had reduction in sum of product of perpendicular
diameters; 2 pts with NHL (monotherapy) achieved responses (1 complete response).
Pts with CLL/SLL had notable reductions in absolute lymphocyte count at doses as low
as 1 mg; 2 responses (partial response or better) were seen with monotherapy and 12
responses with combination (partial response with lymphocytosis or better).
Image:
Summary/Conclusion: These preliminary data show promising efficacy for BGB-11417 and
an improved safety profile, particularly in combination cohorts. Grade ≥3 neutropenia
was uncommon. BGB-11417 is tolerable up to doses of 640 mg as monotherapy and up to
160 mg in combination with ZANU. Dose escalation continues as an MTD has not yet been
reached in any dose-escalation cohort. Enrollment continues, with data for Waldenström
macroglobulinemia and treatment-naïve CLL/SLL cohorts forthcoming.
P688: VIRTUAL SCREENING PROTOCOL TO IDENTIFY TYROSINE KINASE INHIBITORS WITH A POTENTIAL
TO INTERACT WITH P-GLYCOPROTEIN AND TO ACT AS MDR CHEMOSENSITIZERS: A VALIDATION STUDY
E. Beleva1,2,3,*, I. Tsakovska1, P. Alov1, T. Pencheva1, I. Lessigiarska1, I. Pajeva1
1QSAR and Molecular Modeling, Institute of Biophysics and Biomedical Engineering,
Bulgarian Academy of Sciences, Sofia; 2Clinical Oncology, Medical University - Plovdiv,
Plovdiv; 3Faculty of Mathematics and Informatics, Sofia University “St. Kliment Ohridski”,
Sofia, Bulgaria
Background: Resistance to tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia
(CML) has been attributed to both BCR-ABL-dependent pathways - acquisition of point
mutations and BCR-ABL-independent ones – drug efflux mediated by ATP-binding cassette
proteins. Primary multidrug resistance (MDR) has been mostly attributed to aberrant
expression of P-glycoprotein (Pgp). Therefore research efforts have been directed
to improving anticancer drug efficacy by co-administration of TKIs as Pgp modulators.
Recently novel SRC family kinase (SFK) inhibitors based on the pyrazolol[3,4-d]-pyramidine
scaffold were shown to inhibit Pgp in two MDR cancer cell lines with the proforms
outlining a stronger potential to modulate Pgp [1].
Aims: In this study we aimed: (i) to validate the in-house developed virtual screening
(VS) protocol for TKIs with a potential to interact with Pgp; (ii) to compare the
Pgp binding potential of the novel SFK inhibitors (SI306, SI221, pro-SI306, pro-SI221)
with that of widely explored TKIs (core group), including those used for treatment
of CML - imatinib, nilotinib and dasatinib.
Methods: Molecular modelling was performed in MOE software [2]. Two VS protocols were
explored in the study: (A) docking without pharmacophore filter and with triangle
matching placement; (B) an in-house developed VS protocol based on pharmacophore filter
and docking with pharmacophore placement. The following docking settings were applied:
(i) rigid receptor / flexible ligand mode; (ii) London dG scoring function applied
at both the placement stage and the rescoring stage. VS protocol B is based on an
in-house library of 43 TKIs collected from the scientific literature and DrugBank
database. The library includes structural identificators of the compounds and experimental
data proving their P-gp binding. It is freely available at http://biomed.bas.bg/qsarmm/Tyrosine_kinase_inhibitors.The
VS protocols were applied to predict the Pgp binding affinity of the SFK inhibitors
SI306, SI221, pro-SI306, and pro-SI221 as well as the core group drugs.
Results: The results are presented in Table 1. The pro-forms showed higher interaction
energies in the docking simulations compared to their parent ones in agreement with
the experimental results pointing to the pro-forms having a stronger potential to
modulate Pgp (Table 1) [1]. Further the comparison with the reference compound Dasatinib
shows that except for SI221, the novel SFK inhibitors have a similar or even higher
potential to interact with P-gp. The comparison with the core group of drugs outlines
the following order of predicted P-gp affinity: (i) for VS protocol A: pro-SI306>pro-SI221>Nilotinib;
(ii) for VS protocol B: pro-SI221>Lapatinib>pro-SI306>Tepotinib>Nilotinib. In both
cases pro-SI306 and pro-SI221 are among the top-ranked P-gp ligands.
Image:
Summary/Conclusion: In summary, the previously developed VS screening protocol based
on pharmacophore filtering and docking was validated on novel SFK inhibitors that
were predicted as Pgp ligands in agreement with the experimental results. Further,
the VS results outlined their similar or even higher affinity to Pgp compared to Dasatinib,
which was used as reference compound due to its dual BCR-ABL and SRC inhibitory activity.
Therefore they are suitable lead structures in the design of Pgp modulators able to
restore sensitivity to TKIs in resistant tumour cell lines. The presented protocols
as well as the investigated compounds might be useful for virtual screening/drug design
of TKIs that act as MDR chemosensitizers in a combined anti-cancer therapy.
P690: GENE MUTATIONAL PROFILE ASSOCIATED WITH TREATMENT FAILURE OR PROGRESSION IN
CHRONIC MYELOID LEUKEMIA
N. Estrada1,2,*, B. Xicoy3, M. Cabezón3, S. Marcé3, A. Senin4, A. Angona5, E. Alonso4,
M. Ratia4, M. E. Plensa6, J. Buch7, X. Ortín8, L. Zamora3
1Myeloid Neoplasms, Josep Carreras Leukaemia Research Institute; 2Germans Trias i
Pujol Health Science Research Institute (IGTP); 3Institut Català d’Oncologia (ICO)-Hospital
Germans Trias i Pujol, Josep Carreras Leukaemia Research Institute, Badalona, Barcelona;
4Institut Català d’Oncologia - Hospital Duran i Reynals, Hospitalet de Llobregat;
5Institut Català d’Oncologia - Hospital Universitari Doctor Josep Trueta, Girona;
6Hospital de Mataró, Mataró; 7Hospital de Calella and Institut Català d’Oncologia-Girona,
Girona; 8Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
Background: Approximately one third of chronic myeloid leukemia (CML) patients treated
with tyrosine kinase inhibitors (TKI) fail to achieve optimal response due to resistance.
It is known that BCR-ABL1 kinase domain (KD) mutations are associated with TKI resistance.
However, only half on non-responsive patients carry BCR-ABL1 KD mutations. Other resistance
mechanisms independent of BCR-ABL1 KD mutations, such as mutations in myeloid-related
genes, may participate in the lack of response. Moreover, resistance to TKI therapy
confers a much higher risk of progression to advanced disease stages of CML, such
as accelerated phase (AP) and blast crisis (BC), and consequently, a worse prognosis.
Aims: The main objective of this study was to explore the impact of mutations in myeloid-related
genes in TKI resistance and progression to AP/BC by targeted deep-sequencing (TDS).
Methods: The mutation profile study was done by TDS using a 32 myeloid-related gene
panel (Myeloid Solution Capture Kit, Sophia Genetics, Switzerland). The platform Sophia
DDM (Sophia Genetics) was used for data analysis. Regarding variant filtering, sequencing
and mapping errors were eliminated and intronic and synonymous variants were filtered
out. Variants present with a minor allele frequency (MAF) >1%, according to population
databases (ExAc, 1000 genomes), were considered polymorphic changes without clinic
relevance. COSMIC, VARSOME and Franklin databases as well as in silico functional
predictors (SIFT, PolyPhen2 and MutationTaster) were used for variant interpretation.
Variants were also filtered according to the variant allele frequency (VAF): all variants
with VAF ≥5% were reported, as well as hot-spot variants with VAF 2-5% and at least
25 reads. Only variants described as pathogenic or potentially pathogenic were reported.
Finally, BCR-ABL1 mutations were analyzed by Sanger in resistant patients.
Results: TDS was performed in a total of 78 samples from 67 patients. Across patients
with available sample at diagnosis, 25% (16/64) of patients harbored at least one
mutation. Overall, 11 (17.2%) patients had 1 mutation, 3 (4.6%) had 2 mutations, 1
(1.6%) patient had 3, and 1 (1.6%) patient had 4 mutations (Figure 1A). Most frequently
mutated genes at diagnosis were ASXL1 (14.1%), DNMT3A (6.3%), JAK2 (3.1%) and TET2
(3.1%), followed by ETV6, EZH2, IDH2, SETBP1, SRSF2, TP53 and WT1, all of them at
a frequency of 1.6% (Figure 1B). Of the 67 CML patients included, 15 (22.4%) had resistance
to first-line TKI and 5 of them (33%) progressed to AP/BC. From the 15 resistant patients,
TDS was performed in 12 cases at diagnosis, in 5 at time of resistance, and at progression
in 4/5 patients. At diagnosis, 6 (50%) resistant patients harbored 1 mutation in ASXL1
(3/12), DNMT3A (2/12) or TET2 (1/12) and 1 patient harbored 2 mutations (ASXL1 and
WT1). Only 9 of 52 (17.3%) non-resistant patients harbored ≥1 mutation at diagnosis;
most frequently mutated gene was ASXL1 (5/9). At time of resistance, 5 patients had
BCR-ABL1 mutations by Sanger technique and 3 patients harbored cancer-related gene
mutations by TDS (ASXL1 in 2 patients). At progression, all patients harbored at least
1 mutation by TDS, except one patient in which only 2 mutations in BCR-ABL1 KD were
detected.
Image:
Summary/Conclusion:
ASXL1 mutation was the most frequent mutation in CP at diagnosis and ASXL1 and ABL1
were the most common variants at resistance and progression. Acquisition of mutations
was detected during CML progression.
Acknowledgments: this work was supported by a grant from Incyte (EU-ES-H-18021)
P691: MODELLING OF IMMUNE RESPONSE IN CHRONIC MYELOID LEUKEMIA PATIENTS SUGGESTS POTENTIAL
FOR TREATMENT REDUCTION PRIOR TO CESSATION
E. Karg1, C. Baldow1, T. Zerjatke1, R. E. Clark2, I. Roeder1,3, A. C. Fassoni4, I.
Glauche1,*
1Institute for Medical Informatics and Biometry (IMB), TU Dresden, Dresden, Germany;
2Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool,
United Kingdom; 3National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden,
Germany; 4Instituto de Matemática e Computação, Universidade Federal de Itajubá, Itajubá,
Brazil
Background: Chronic myeloid leukemia (CML) is a malignancy driven by the characteristic
BCR-ABL1-fusion oncogene, while the BCR-ABL1 transcript is used as a marker for diagnosis
and monitoring. The introduction of tyrosine kinase inhibitors (TKI) marked a substantial
progress in CML therapy. Current efforts focus on the discontinuation of TKI therapy
for well-responding patients as the long-term drug administration is associated with
side effects and high economical costs. Many studies reported that about 50% of patients
can achieve sustained treatment free remission (TFR) while 50% present with molecular
disease recurrence, typically within two years of stopping. It has been speculated
that the immune system (IS) plays a major role in the control of residual disease
levels and influences the individual recurrence behaviour. The recently published
DESTINY trial (Clark et al., Lancet Haematol, 2019; NCT 01804985) showed that TKI
dose reduction prior to cessation can proactively increase the fraction of patients
to remain in sustained TFR. However, there has been no systematic investigation to
evaluate how dose reduction regimens can further improve the success of TKI stop trials.
We have previously shown that mathematical models of CML can correctly describe patient
time courses after TKI stop (Hahnel et al., Cancer Res, 2020). We classified patients
into different groups according to their predicted immune system configuration and
showed that one class of patients required complete CML eradication to achieve TFR,
while other patients were able to control residual leukemia levels after treatment
cessation. While a prospective stratification of the patients is difficult to achievable,
we aimed to investigate whether a further optimized dose reduction scheme prior to
stopping could further improve TFR.
Aims: We aim to use a mathematical description of patient time courses to investigate
different TKI dose reductions schemes prior to therapy cessation and evaluate them
with respect to the total amount of drug used and the expected TFR success.
Methods: Our analysis is based on a cohort of 72 patients from the DESTINY trial (Clark
et al., Lancet Hematol, 2019; NCT 01804985). We applied an established mathematical
model (Hahnel et al., Cancer Res, 2020) and obtain individual fits for all patients
using a genetic algorithm. Applying a re-sampling approach we derive estimates of
recurrence times and fractions for different dose reduction schemes in which the timing
and the amount of dose reduction are varied in a systematic manner.
Results: We use computer simulations to show that TKI dose reduction prior to treatment
cessation appears as an equally efficient approach to achieve high levels of CML patients
remaining in TFR compared to full dose treatment for the same duration. Our model
simulations confirm clinical findings that the overall time of TKI treatment is a
major determinant of TFR success, while at the same time indicating that lower dose
TKI treatment is sufficient for many patients. Our model results indicate that a stepwise
dose reduction prior to TKI cessation (e.g. 12 month post MR4 at 100% TKI, 12 more
months at 50% and 12 more months at 25%) does not limit the overall success rate of
TFR, while it substantially reduces the amount of total administered TKI and thereby
decreases side effects as well as overall treatment costs.
Summary/Conclusion: Our findings illustrate the potential of dose reduction schemes
prior to treatment cessation and suggest corresponding and clinically verifiable strategies
that are applicable to many CML patients.
P692: IMMUNE FACTORS MAINTAINING TREATMENT-FREE REMISSION IN PATIENTS WITH CHRONIC
MYELOID LEUKEMIA AFTER DISCONTINUATION OF IMATINIB
P. Kwaśnik1,*, J. Zaleska1, D. Link-Lenczowska2, M. Zawada2, B. Ochrem2, G. Bober3,
E. Wasilewska4, E. Mędraś5, I. Hus6,7, M. Szarejko8, W. Prejzner8, O. Grzybowska-Izydorczyk9,
A. Klonowska-Szymczyk9, T. Sacha2, K. Giannopoulos1,10
1Department of Experimental Hematooncology, Medical University of Lublin, Lublin;
2Department of Haematology, Jagiellonian University Hospital in Krakow, Krakow; 3Department
of Hematooncology and Bone Marrow Transplantation, Medical University of Silesia,
School of Medicine in Katowice, Katowice; 4Department of Hematology, Medical University
of Bialystok, Bialystok; 5Department of Hematology, Blood Neoplasms and Bone Marrow
Transplantation, Medical University of Wroclaw, Wroclaw; 6Department of Hematology,
Institute of Hematology and Transfusion Medicine, Warsaw; 7Department of Clinical
Transplantology, Medical University of Lublin, Lublin; 8Department of Hematology and
Transplantology, Medical University of Gdansk, Gdansk; 9Department of Hematology,
Medical University of Lodz, Copernicus Memorial Hospital, Lodz; 10Department of Hematology,
St John’s Cancer Center, Lublin, Poland
Background: CML treatment aims to achieve long-term remission without treatment (TFR).
The mechanisms that are responsible for CML relapses remain elusive. It is suggested
that maintaining TFR is not directly related to the total disposing of the gene transcript
BCR-ABL1, but it might be a result of the immune surveillance restoration in CML.
Aims: To determine immune factors responsible for elimination or control over residual
CML cells, which may be crucial for achieving long-term relapse-free survival (RFS)
and TFR after imatinib discontinuation.
Methods: Peripheral blood mononuclear cells from 63 CML patients were analyzed using
flow cytometer analysis at two-time points: at discontinuation of imatinib and 3 months
after withdrawal. The major populations of the immune system that have a potential
effect on the maintenance of the patient in remission were assessed. BCR-ABL1 transcript
were analyzed using two molecular biology methods: standard real-time quantitative
polymerase chain reaction (RQ-PCR) and droplet digital PCR (ddPCR).
Results: We found a significant increase in the percentage of HLA-DR+ cells (median
1.25 vs 1.90%, p<0.0001), mDC (median 0.20 vs 0.28%, p<0.01), pDC (median 0.10 vs
0.15%, p<0.01) and a significant decrease in PD-1 expression on mDC cells (median
35.81 vs 32.23%, p=0.03) in 3 months after treatment discontinuation.
There was a significantly reduced level of NKT cells (median 14.45 vs 12.80%, p=0.02),
a significantly increased PD-1 expression on CD161+ cells (median 21.02 vs 21.28%,
p=0.02) and a significantly decrease of PD-1 expression on CD56brightCD16- cells (median
2.31 vs 2.08%, p=0.04).
Analyses of T cells showed a significant decrease in frequencies of CD4+CD25+ T cells
(median 18.01% vs 13.42%, p<0.0001), CD4+CD25+FOXP3+ (median 3.00 vs 2.89%, p=0.02)
and a significantly lower expression levels of PD-1 on CD4+ cells (median 21.59 vs
19.34%, p=0.02) after 3 months compared to moment of withdrawal.
We found a weak negative correlation between age and CD19+ cell depletion (p<0.01,
R=-0.39) and a moderate positive correlation between age and PD-1 expression on CD19+
cells (p<0.001, R=0.43) at the moment of withdrawal. In addition, we found a weak
positive correlation between age and HLA-DR+ cell count (p=0.01, R=0.32) and a weak
negative correlation with CD19+ cell count (p=0.04, R=-0.26) in 3 months after stopping
treatment.
Significantly higher levels of NK cells: CD56dimCD16+ (median 8.93 vs 14.23%, p<0.01)
and CD56brightCD16- (median 0.33 vs 0.52%, p=0.02) were found in men compared to women
3 months after withdrawal.
Analysis of the BCR-ABL1 transcript by both RQ-PCR and ddPCR methods in relation to
the immune subpopulations showed significant differences between the percentage of
HLA-DR+ cells and the depth of the molecular response: in RQ-PCR greater than or equal
to MR4.0 at the moment of withdrawal vs loss of MR4.0 after 3 months (median 1.24
vs 1.87, p=0.03); greater than or equal to MR4.0 at the moment of withdrawal vs greater
than or equal to MR4.0 after 3 months (median 1.24 vs 1.92, p<0.001).
Summary/Conclusion: The demonstrated changes in the percentage of populations of immune
cells show the importance of immunocompetent cells’ involvement in maintaining the
patient in TFR. The characterization of the immune system, which probably plays the
most important role in achieving long-term relapse-free response and maintaining remission
without treatment, may help define the group of patients who could safely discontinue
imatinib.
Funding: NCN 2018/31/B/NZ6/03361
P693: DIAGNOSTIC ROLE OF CD26+ LEUKEMIC STEM CELLS IN CHRONIC MYELOID LEUKEMIA
S. Tiwari1,*, J. Dass1, G. Vishwanathan1, R. Dhawan1, M. Agarwal1, P. Kumar1, T. Seth1,
S. Tyagi1, M. Mahapatra1
1Hematology, All India Institute of Medical Sciences, New Delhi, India
Background: Diagnosis of CML consists of persistent leucocytosis with the demonstration
of the Philadelphia (Ph) chromosome abnormality, the t(9;22)(q34;q11), by routine
cytogenetics, fluorescence in situ hybridization (FISH) or by molecular studies such
as RT-PCR.These conventional methods suffer from a longer turn around time, higher
costs and requirement of labs with adequate expertise. CD26 is a highly specific marker
expressed in CML stem cells, which is not present on normal hematopoietic stem cells
or on Leukemia stem cells (LSCs) of other myeloid neoplasms. A flow-cytometry based
assessment of CD26+LSCs may prove to be an optimal biomarker for the diagnosis and
monitoring of CML patients.
Aims: 1)Assessment of CD 26 expression in suspected cases of CML-chronic phase
2)Correlation of CD 26+ stem cells with clinico-pathological parameters at baseline
and its kinetics on tyrosine kinase inhibitor (TKI) treatment on further follow-up
at 12 months.
Methods: Suspected patients of CML-CP were included in the study. Peripheral blood
were utilized for flowcytometric assessment of CD26+ LSCs. The results were correlated
with conventional diagnostic tests of CML. Patients with myeloid neoplasms other than
CML who proved negative for BCR-ABL1 by RT-PCR and normal HSCs donors treated with
granulocyte-colony stimulating factors (G-CSF), were included as negative controls.
All samples were processed using standard stain -lyse-wash method within 24 hr of
collection. To reach a sensitivity of 10−5, acquisition of at least 1.0 × 106 cells
was performed on FACS Canto II flow cytometer data a FACS DIVA 8.0.3 software (BD,
Biosciences).CD26 expression was evaluated by a sequential gating strategy. After
exclusion of doublets and debris CD45 dim to intermediate/CD34+/CD38- population was
gated sequentially and then the expression of CD 26+ was studied on this population.
All the samples were simultaneously assessed for BCR-ABL1 transcript by standard RT-PCR
analysis. Patients on treatment were also followed up at 12 months to detect levels
of CD26+ LSCs and correlated with BCR -ABL1 levels by qPCR.
Results: A total of 86 patients of suspected cases of CML were included in the study
from June 2020 to Dec 2021.The On flowcytometry analysis, CD 26 expression was present
in 75 cases. All the cases with positive CD26+ LSCs were found to be positive on RT-PCR
for BCR-ABL transcript. Eleven cases which were scored negative for CD26+ LSCs were
also found to be negative for any BCR-ABL transcript. These negative cases included
8 cases of primary myelofibrosis, 2 cases of chronic myelomonocytic leukemia and 1
case of Polycythaemia Vera. The median percentage of CD26+ LSCs was 39.8% (Range-0.5-100).
There was no significant correlation of the CD26 levels with age (p value=0.96), sex
(p value=0.7), percentage of blasts (p value = 0.88), total leucocyte counts (p value=0.32),
basophils (p value = 0.19), Sokal score (p value = 0.99) and ELTS score (p value =
0.99). Follow-up data was available for 7 patients and at 12 months all of these patients
had achieved major molecular remission on TKIs. Two patients scored negative for CD26+LSCs
while five patients showed significant fall in the level of CD 26+ LSCs however it
was still detectable by flowcytometry (p value= 0.04).
Summary/Conclusion: Flowcytometry assessment of CD26 + LSCs is a rapid and cheap diagnostic
tool for diagnosis of CML with high diagnostic efficacy. Falling levels on TKIs warrants
further research to prove it as a surrogate for monitoring response by conventional
methods.
P694: INVESTIGATING THE ROLE OF CELL ADHESION MOLECULES IN TUNNELING NANOTUBES FORMATION
IN CHRONIC MYELOID LEUKEMIA MICROENVIRONMENT
L. Turos-Korgul1,*, A. Zieminska1, L. Parker2, K. Piwocka1
1Laboratory of Cytometry, Nencki Institute of Experimental Biology PAS, Warsaw, Poland;
2Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota,
Minneapolis, United States of America
Background: The cross-talk and interactions between leukemia cells and their microenvironment
play an important role in the development of drug resistance. Tunneling nanotubes
(TNTs) represent a novel type of direct cell-cell communication way allowing for direct
transfer of different types of cargo between distant cells. Our previous studies (Kolba
et al., 2019) have shown that these structures are formed between bone marrow stromal
HS-5 cells and chronic myeloid leukemia (CML) K562 cells. Significantly, TNT-mediated
transfer of membrane vesicles from stromal to leukemic cells resulted in the stroma-mediated
protection of CML cells from imatinib-induced apoptosis. Despite the efforts made
to understand the molecular mechanisms regulating TNTs formation, they still remain
unclear - especially in the leukemia microenvironment. We have shown that TNTs are
formed between stromal and leukemic cells, following direct cell-cell interaction
and cell dislodgement. Thus, we have focused our attention on cell adhesion molecules,
as potential key players in tunneling nanotubes formation and TNT-mediated vesicles
transfer between leukemic and stromal cells.
Aims: The aim of presented studies was to elucidate the role of CD44 and ITGβ1 - one
of the most important cell adhesion molecules, in TNTs formation between leukemic
and stromal cells and TNT-mediated exchange of vesicles.
Methods: To examine this, we established the HS-5 stromal cell line with silenced
CD44 protein expression. Then, we co-cultured control (wt) or CD44-silenced HS-5 cells
with leukemic K562 cells. For ITGβ1 studies, we used blocking anti-ITGβ1 antibody.
Using confocal microscopy, three types of TNTs in co-cultures were counted: homotypic
TNTs formed between stromal cells, homotypic TNTs formed between CML cells and TNTs
formed between stromal and CML cells (heterotypic). Moreover, the TNT-mediated transfer
of cellular vesicles from stromal to leukemic cells was assessed in these co-cultures
using flow cytometry. Since CD44 expression increases in CML cells after co-culture
with stroma, we examined its potential role in the drug resistance in CML acceptor
cells. To do this, rhodamine-123 efflux assay was used.
Results: We found that both, tunneling nanotubes number and uptake of cellular vesicles
by leukemia, decreased after co-culture with CD44-silenced stromal cells and after
blocking with anti-ITGβ1 antibody. Flow cytometry analyses of rhodamine-123 efflux
showed significantly increased efflux pump activity in leukemic acceptor cells.
Summary/Conclusion: Our data suggest that CD44 and ITGβ1 can play an important role
in formation of tunneling nanotubes between leukemic and stromal cells and TNT-mediated
cellular vesicles exchange. Moreover, CD44 presented in acceptor cells is involved
in the development of drug resistance in leukemic cells, by promoting activity of
efflux transporters.
P695: PROGNOSTIC IMPACT OF ASXL1 MUTATIONS IN CHRONIC PHASE CML
A. Bidikian1,*, H. Kantarjian1, E. Jabbour1, N. Short1, F. Ravandi1, G. Issa1, K.
Sasaki1
1Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston,
TX, United States of America
Background: While the clinical impact of mutations in the ABL1 gene on response to
therapy in chronic phase chronic myeloid leukemia (CP-CML) is well established, less
is known about mutations in genes frequently involved in myeloid malignancies.
Aims: To characterize the clinical impact of mutations in non-ABL1 genes in CP-CML.
Methods: We identified 66 patients (pts) with CP-CML where targeted next generation
sequencing was performed at our institution. This panel included 81 genes recurrently
mutated in myeloid malignancies. Event-free survival (EFS) was measured from treatment
initiation to the time of an event defined as loss of hematologic response, loss of
major cytogenetic response (MCyR), transformation to accelerated or blast phase and
lack of MCyR after 12 months of treatment. Failure-free survival (FFS) was measured
similarly with the additional events of drug discontinuation due to lack of response
or adverse effect. We subsequently performed univariate and multivariate analyses
for survival.
Results: The median age of pts was 61 years (18 – 80 years) and 48% were female. The
most common first line therapy in these patients was Dasatinib (52%), followed by
Imatinib (19%), Dasatinib + Venetoclax (16%), Nilotinib (8%), Bosutinib (3%) and Ponatinib
(2%). The distribution of evaluable patients to low, intermediate and high Sokal risk
groups was 22%, 62% and 16%, respectively. Non-ABL1 mutations were found in 22/66
patients (33%), with 5 (8%) of pts with at least 2 mutations. Most common mutations
involved ASXL1 (8/66 pts; 12%), DNMT3A (4/66 pts; 6%) and RUNX1 (3/66 patients; 5%).
Mutations in ASXL2, CALR, EZH2, GNAS, IDH2, SF3B1, SMC3, SRSF2, STAG2 and SUZ12 were
detected at least once. Unlike mutations in other genes, mutated ASXL1 was associated
with worse EFS compared with wild-type with a median of 33 vs 88 months (P = 0.009)
(Figure 1A) and worse FFS with medians of 32 vs 58 months, respectively (P = 0.05)
(Figure 1B). There was no difference in overall survival by mutational status (Figure
1C). In a multivariate analysis including all baseline prognostic variables, mutated
ASXL1 was independently associated with worse EFS with a hazard ratio of 5.94 (95%
CI 1.25 – 28.21; P = 0.03). There was no difference in rates of cytogenetic and molecular
responses by mutational status. However, time to achievement of response was longer
in patients with mutated ASXL1 albeit not statistically significant. (Figure 1D)
Image:
Summary/Conclusion: Mutations in ASXL1 are associated with delayed responses and worse
outcomes when detected in chronic phase CML.
P696: SUBTHERAPEUTIC TKI DOSES FOR THE MAINTENANCE OF CML PATIENTS WITH INTOLERANCE
OR REFUSAL TO DISCONTINUE: A SINGLE CENTER FEASIBILITY STUDY.
A. Borrero1,*, A. Segura1, S. Sanchez1, R. Stuckey1, J. F. López1, C. Bilbao1, M.
T. Gomez casares1
1HOSPITAL UNIVERSITARIO DE GRAN CANARIA DOCTOR NEGRIN, LAS PALMAS DE GRAN CANARIA,
Spain
Background: Discontinuation has become a common practice in patients with CML treated
with TKI who have maintained a deep molecular response. 50-60% of patients successfully
discontinue treatment without relapsing (treatment-free remission, TFR), which implies
that 40-50% will have to remain with TKI treatment for life. Many studies have been
carried out on TKI treatment with reduced doses, often used to manage adverse events
and TKI intolerance. Such studies have shown that most patients maintain MMR and many
even achieve it when reduced doses are administered from diagnosis. There are fewer
cases reported of patients who receive subtherapeutic doses, i.e. doses lower than
those considered in the TKI data sheet. Reasons why subtherapeutic doses may be employed
in clinical practice include the patient’s refusal to discontinue or a failed discontinuation
attempt (loss of TFR), as a maintenance strategy, or in those patients with intolerance.
Although its use is likely, information on patient outcome is scarce.
Aims: We investigated the feasibility of the use of subtherapeutic doses in CML patients.
We studied the reason for the dose reduction and the impact of the dose reduction
on molecular response.
Methods: In this observational, retrospective, single-center study, we searched the
medical records of all patients diagnosed with CML in chronic phase at our hospital
between 2003 and 2020. Patients were identified who were treated with TKIs at subtherapeutic
doses. Subtherapeutic doses were defined as doses lower than those considered in the
TKI data sheet: dasatinib 20 mg, imatinib 100 mg or 200 mg, nilotinib 150 mg, bosutinib
100 mg.
Results: Medical records were searched and 13 CML patients who received infratherapeutic
TKI doses were identified, 7 men (53.9%) and 6 women (46.2%) with a mean age of 75.5
years. Of these patients, 7 (53.8%) received imatinib (6 with 200 mg doses and 1 with
100 mg), 1 (7.7%) received bosutinib 100 mg and 5 (38.5%) received dasatinib 20 mg.
The mean follow-up time from the start of the dose reduction was 60.5 months and 40.7
months from the last reduction. None of the patients presented TKI resistance or progression
during the follow-up period. 3 patients reduced the dose as a practical alternative
for fear of discontinuation; 2 patients discontinued, but after relapse they remained
with the minimum dose to maintain MMR; the rest of the patient reduced doses because
of intolerance. All patients presented MMR at last follow-up and 92.3% deep MMR (≥MR4).
Summary/Conclusion: Our results show that TKI doses can be optimized on an individual
basis to infratherapeutic levels and MMR or better can be maintained. Although only
a small series of patients was studied, no patients developed TKI resistance or progression
while receiving infratherapeutic doses during an average period of 5 years. For intolerant
patients who received infratherapeutic doses, adverse events were resolved and quality
of life improved. The administration of infratherapeutic TKI doses could also be a
good option for patients who relapse after a discontinuation attempt or for those
who do not want to discontinue, as a form of “maintenance” therapy. The use of infratherapeutic
doses, rather than a full TKI discontinuation, would still provide economic savings.
Moreover, it would be interesting to evaluate if long-term use of infratherapeutic
doses can prevent the development of serious off-target TKI toxicities, such as vascular
events. Studies with larger patient cohorts are needed to validate the feasibility
of this clinical practice.
P697: PONATINIB IN A REAL-LIFE SETTING: A RETROSPECTIVE ANALYSIS FROM THE MONITORING
REGISTRIES OF THE ITALIAN MEDICINES AGENCY (AIFA)
M. Breccia1,*, P. P. Olimpieri2, S. Celant2, O. M. Olimpieri2, F. Pane3, A. Iurlo4,
V. Summa2, P. Corradini5, P. Russo2
1Department of Translational and Precision Medicine, Sapienza University; 2Italian
Medicines Agency, AIFA, Roma; 3Federico II University, Napoli; 4Foundation IRCCS Ca’
Granda Ospedale Maggiore Policlinico; 5Università degli Studi di Milano & Divisione
Ematologia, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
Background: The efficacy and safety of ponatinib, a third-generation tyrosine kinase
inhibitor (TKI), are mainly reported in sponsored clinical trials.
Aims: The aim of this study, based on a retrospective analysis from the monitoring
Registries of the Italian Medicines Agency (AIFA wMRs), is to provide real-world data
on daily practice management, treatment modifications and outcome of a large cohort
of chronic myeloid leukemia (CML) patients treated with ponatinib.
Methods: AIFA wMRs is an administrative database whose main scope is monitoring the
appropriateness of drug prescription in Italy. Information collected through the wMRS
included demographic (place of birth, age, and sex) and clinical data (BCR-ABL levels,
mutations and chromosomal abnormalities if evaluated), drug prescription and administration
data (date of prescription and administration, dose administered, dose changes, occurrence
of adverse events as yes/no dichotomic variable, reasons for treatment interruption
and/or discontinuation) and response to treatment (response assessment was requested
every 90 days). Subjects who had not been administered ponatinib for ≥ 120 days were
adjudicated as “discontinued” even in the absence of the “end of treatment form”.
Time to treatment discontinuation (TTD) was defined as the time occurring between
the date of first administration and the date of treatment discontinuation for any
cause, including death.
Results: Overall, 666 CML subjects were eligible for analysis: 515 patients had chronic
phase (CP) CML, 50 accelerated phase (AP) and 101 blast phase (BP). Median age at
baseline was 58.7 years, male prevalence (57.1%). Median time from diagnosis to start
of ponatinib was 2.35 years: 259 (38.9%) subjects had received 2 lines of treatment,
260 (39.0%) 3 lines and 147 (22.1%) 4 or more lines. Mutational status was available
for 58.3% of patients (n=388), with a T315I mutation reported in 46 (6.9%) patients
and other mutations (the most represented being E255K, F317L, Y253H and V299L) in
99 (14.9%) subjects. At baseline, 181 patients (27.2%) had arterial hypertension,
33 (5.0%) a history of arterial and/or venous thromboses, 32 (4.8%) a pre-existing
ischemic heart disease, and 13 (2.0%) a history of congestive heart failure. Overall,
593 patients (89.0%) were evaluable for best response. Ten cases (1.7%, 3 AP/BP and
7 CP) did not achieve molecular response, whereas 58 patients (8.7%, 26 AP/BP patients
and 32 CP) reached a BCR/ABL1 ratio between 1% and 10% IS. A MR2 (less than 1%IS)
was obtained by 82 subjects (12.3%, 20 AP/BP and 62 CP patients); 128 patients (19.2%,
12 AP/BP and 116 CP patients) achieved a molecular response ranged between MR3 (0.1%)
and MR4 (0.01% IS) and 266 (39.9%, 44 AP/BP and 222 CP patients) a deeper molecular
response (< 0.01% IS). With a median follow-up of 14.4 months, 136 subjects (20.4%)
required at least one dose reduction due to adverse events; 309 patients (46.4%) had
their dose decreased in the absence of any evidence of side effects. Treatment discontinuation
occurred in 144 patients (21.6%): intolerance (7.4%), primary resistance (3.5%) and
acquired resistance (5.6%). Kaplan-Meier estimated TTD was 47.2 months (CI 95% 39.3
– NA) and 7.3 months (CI 95% 4.6 – 11.8) for CP and AP/BP patients, respectively.
The probability of treatment discontinuation did not significantly differ for ponatinib
in second, third or subsequent line of therapy (p=0.58).
Summary/Conclusion: This real-life investigation shows that ponatinib dose reductions
were mainly performed in the absence of reported toxicity rather than owing to the
occurrence of adverse reactions.
P698: BOSUTINIB DOSE OPTIMIZATION IN THE SECOND-LINE TREATMENT OF ELDERLY CML PATIENTS:
EXTENDED 3-YEAR FOLLOW-UP AND FINAL RESULTS OF THE BEST STUDY
F. Castagnetti1,2,*, M. Bocchia3, E. Abruzzese4, I. Capodanno5, M. Bonifacio6, G.
Rege Cambrin7, M. Crugnola8, G. Binotto9, C. Elena10, A. Lucchesi11, M. Bergamaschi12,
F. Albano13, L. Luciano14, F. Sorà15, F. Lunghi16, F. Stagno17, M. Cerrano18, A. Iurlo19,
A. R. Scortechini20, S. Leonetti Crescenzi21, R. Spadano22, E. Trabacchi23, M. Lunghi24,
G. Spinosa25, D. Ferrero26, D. Rapezzi27, M. Ladetto28, L. Nocilli29, G. Gugliotta1,
M. Iezza1, M. Cavo1,2, G. Saglio30, F. Pane14, G. Rosti11
1Istituto di Ematologia “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna;
2Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di
Bologna, BOLOGNA; 3Hematology Unit, University of Siena, Siena; 4Hematology Unit,
S. Eugenio Hospital, Tor Vergata University, Roma; 5Hematology Unit, Azienda Unità
Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia; 6Department of Medicine, Section
of Hematology, University of Verona, Verona; 7Internal Medicine Unite, San Luigi Gonzaga
Hospital, Orbassano (TO); 8Hematology Unit, Azienda Ospedaliero-Universitaria di Parma,
Parma; 9Department of Medicine, Hematology and Clinical Immunology, University Hospital
of Padova, Padova; 10Department of Hematology Oncology, Foundation IRCCS Policlinico
San Matteo, University of Pavia, Pavia; 11Hematology Unit, Istituto Scientifico Romagnolo
per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola (FC); 12Hematology Unit,
Policlinico S.Martino-IRCCS, Genova; 13Hematology Unit, Department of Emergency and
Organ Transplantation, University of Bari, Bari; 14Hematology Unit, Federico II University,
Napoli; 15Hematology Unit, Policlinico A. Gemelli, Università Cattolica del Sacro
Cuore, Roma; 16Hematology and Bone Marrow Transplant Unit, San Raffaele Scientific
Institute IRCCS, Milano; 17Hematology Section and BMT Unit, AOU Policlinico V. Emanuele,
Catania; 18Hematology Unit, AOU Città’ della Salute e della Scienza, Torino; 19Hematology
Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano; 20Hematology
Unit, A.O.U. Ospedali Riuniti, Ancona; 21Hematology Unit, San Giovanni - Addolorata
Hospital, Roma; 22Hematology Unit, Casa Sollievo Della Sofferenza, San Giovanni Rotondo
(FG); 23Hematology and BMT Unit, Department of Hematology and Oncology, Guglielmo
da Saliceto Hospital, Piacenza; 24Hematology Unit, AOU Maggiore della Carità, Novara;
25Hematology Unit, Azienda Ospedaliero Universitaria Ospedali Riuniti, Foggia; 26Department
of Molecular Biotechnology and Health Sciences, University of Torino, Torino; 27Hematology
Unit, AO S. Croce e Carle, Cuneo; 28Hematology Unit, Ospedale SSAntonio e Biagio e
Cesare Arrigo, Alessandria; 29Hematology Unit, Azienda Ospedaliera Papardo, Messina;
30Divisione Universitaria di Ematologia e Terapie Cellulari, A.O. Ordine Mauriziano,
Torino, Italy
Background: The median age of CML patients requiring a second-line treatment is older
than 60 years and many of these patients have relevant comorbidities. Bosutinib (BOS)
is a second-generation TKI, with similar efficacy to dasatinib and nilotinib, but
more favorable toxicity profile in the elderly, because of lower incidence of cardiovascular
(CV) adverse events (AEs). BOS safety profile may be an added value in this setting,
but a fixed initial dose of 500 mg OAD may be higher than necessary.
Aims: All TKIs have been approved in CML at a specific initial dose, with dose reductions
only in case of toxicity. The aim of our study was to evaluate the efficacy of low-dose
BOS in the second line treatment of elderly CML patients, with subsequent dose increase
only in patients without optimal molecular response at given timepoints (dose optimization).
Methods: A prospective phase 2 single-arm study has been designed by the GIMEMA CML
Working Party (NCT02810990). Study design: all patients, ≥ 60 yrs old, started BOS
200 mg for 2 weeks (“run-in” period), then the dose was increased to 300 mg; after
3 months, pts with BCR-ABLIS transcript ≤ 1% continued 300 mg, while in pts with transcript
> 1% the dose is furtherly increased to 400 mg, in absence of relevant toxicity. The
primary endpoint was the rate of MR3 at 12 months (core phase), but all pts were followed
for additional 2 years (extension phase) to describe the long-term efficacy and outcome.
Results: Sixty-three pts have been enrolled. Median age: 73 yrs (range 60-90). Reasons
for switching to BOS: intolerance 65%, resistance 35%. First-line TKI: imatinib 83%,
DAS 11%, NIL 6%. The rate of pts in MR3 at 12 mos was 59% (already reported). Median
follow-up 38 mos (range 35-59). Maximum BOS dose: 400 mg, 21%; 300 mg, 73%; 200 mg,
6%. The probabilities of achieving or maintaining MR3, MR4 and MR4.5 by 36 mos were
78%, 54% and 46%, respectively (excluding pts with response at baseline, 73%, 50%
and 44%, respectively); the probabilities were lower in the resistant cohort (64%,
36% and 18%, respectively). Overall, 24%, 33% and 11% of pts had 1 log, 2 logs or
≥ 3 BCR-ABLIS transcript logs reduction from baseline (68% of pts had a molecular
improvement from baseline). At 36 mos, 36 pts (57%) were still on BOS (33 pts in optimal
response, 3 pts in MR2), while 27 pts (43%) discontinued the study drug. Events leading
to permanent treatment discontinuation: 7 CML unrelated deaths, 9 AEs (transaminase
increase in 5 pts, skin rash, myalgia, GI toxicity and renal failure in 1 patient
each), 9 unsatisfactory responses (without progression to advanced phases), 1 treatment-free
remission attempt, 1 other reason. The overall survival probability was 81%. Pts with
CV AEs: acute coronary syndromes, 6 pts; pericarditis, 2 pts; peripheral arterial
thrombosis, 1 pt (all pts had CV risk factors). No pleural effusions were observed.
The incidence of GI toxicity was lower than reported elsewhere. Thirty-six pts were
still on BOS at the last contact: 6% on 400 mg, 50% on 300 mg, 44% on 200 mg.
Image:
Summary/Conclusion: A progressive dose increase, based on molecular response, in elderly
CML patients treated with second-line bosutinib produced high response rates; few
patients required a dose increase to 400 mg, while most pts remained on 300 mg or
less. Not unexpectedly, given the old age, unrelated CV AEs and deaths occurred, but
the study drug was generally well tolerated. An initial use of low dose TKIs, with
dose optimization in case of absence of optimal response, is a promising strategy
in elderly patients.
P699: REAL-LIFE OUTCOMES OF PONATINIB TREATMENT IN PATIENTS WITH CHRONIC MYELOID LEUKEMIA
(CML) OR PHILADELPHIA CHROMOSOME-POSITIVE ACUTE LYMPHOBLASTIC LEUKEMIA (PH+ALL): 5-YEAR-DATA
FROM A BELGIAN REGISTRY
T. Devos1,2,*, V. Havelange3, K. Theunissen4, S. Meers5, F. S. Benghiat6, A. Gadisseur7,
G. Vanstraelen8, H. Vellemans9, B. Bailly10, N. Granacher11, P. Lewalle12, A. De Becker13,
K. Van Eygen14, M. Lejeune15, M. Janssen16, A. Triffet17, I. Vrelust18, D. Deeren19,
D. Mazure20, M. Beck21, H. Sebti21, D. Selleslag22
1Department of Hematology, University Hospitals Leuven; 2Department of Microbiology
and Immunology, Laboratory of Molecular Immunology (Rega Institute), Leuven; 3UCL
Saint-Luc, Woluwe-Saint-Lambert; 4Jessa Ziekenhuis, Hasselt; 5Algemeen Ziekenhuis
Klina, Brasschaat; 6Hôpital Erasme, Bruxelles; 7Universitair Ziekenhuis Antwerpen,
Edegem; 8CHR Verviers, Verviers; 9CHU UCL Namur, Site Godinne, Yvoir; 10Hôpital de
Jolimont, Haine-Saint-Paul; 11Ziekenhuis Netwerk Antwerpen Stuivenberg, Antwerpen;
12Institut Jules Bordet, Université Libre de Bruxelles, Bruxelles; 13Universitair
Ziekenhuis Brussel, Jette; 14Algemeen Ziekenhuis Groeninge, Kortrijk; 15Centre Hospitalier
Universitaire de Liège (Sart-Tilman), Liège; 16Ziekenhuis Oost-Limburg, Genk; 17Centre
Hospitalier Universitaire Charleroi Vésale, Charleroi; 18Algemeen Ziekenhuis Sint-Elisabeth,
Turnhout; 19Algemeen Ziekenhuis Delta, Roeselare; 20Universitair Ziekenhuis Gent,
Gent, Belgium; 21Incyte Biosciences Benelux B.V., Amsterdam, Netherlands; 22Algemeen
Ziekenhuis Sint-Jan Brugge, Brugge, Belgium
Background: Ponatinib is a third-generation tyrosine kinase inhibitor (TKI) indicated
for adult patients with chronic, accelerated or blast phase CML resistant or intolerant
to nilotinib or dasatinib or with Ph+ALL resistant or intolerant to dasatinib or for
patients with the T315I mutation. Real-world data on ponatinib treatment are limited
but are important to evaluate the effectiveness and safety of ponatinib and optimize
its use in daily practice.
Aims: To report 5-year-Belgian registry data on ponatinib use in CML and Ph+ALL patients
in routine clinical practice.
Methods: This ongoing prospective multi-center registry (NCT03678454) includes ≥18-year-old
patients with CML or Ph+ALL eligible for ponatinib treatment per product label. Data
on demographics, effectiveness (major molecular response [MMR] rate) and safety were
collected for patients enrolled from 01-March-2016 onwards and are presented here
up to 17-May-2021.
Results: In this interim analysis, 77 patients from 21 hospitals were enrolled (50
CML, 27 Ph+ALL). Of the 28 patients (39%; 14 CML, 14 Ph+ALL) with mutations in the
BCR-ABL1 kinase domain, 17 (8 CML, 9 Ph+ALL) had the T315I mutation at entry. Median
age at ponatinib start was 61 years for CML and 56 years for Ph+ALL patients. 88%
of CML and 96% of Ph+ALL patients had received ≥2 prior TKIs. Potential pre-existing
risk factors for TKI cardiovascular toxicity were observed: history of cardiovascular
disease (27 patients), hypertension (23), smoking (15), diabetes (13), hypercholesterolemia
(8), hyperlipidemia (6). The most frequently reported risk factor was history of cardiovascular
disease (36% CML, 33% Ph+ALL). Median follow-up was 545 (14-3190) days for CML and
210 (26-2933) days for Ph+ALL patients. Reasons for starting ponatinib therapy were
intolerance to previous TKIs (26 CML, 11 Ph+ALL), relapse on/refractoriness to previous
TKIs (11 CML, 8 Ph+ALL), T315I mutation (6 CML, 6 Ph+ALL) and disease progression
(7 CML, 2 Ph+ALL). Ponatinib starting doses were: 45 mg/day (66%), 30 mg/day (10%),
15 mg/day (22%) in CML patients (1 patient started with 15 mg every 2 days) and 45 mg/day
(67%), 30 mg/day (15%), 15 mg/day (19%) in Ph+ALL patients. MMR was achieved in 58%
of CML and 52% of Ph+ALL patients. The median time-to-MMR was 170 days in CML and
86 days in Ph+ALL patients. Of the 37 patients who started ponatinib due to intolerance
to previous TKIs, 59% (15 CML, 7 Ph+ALL) achieved MMR. Adverse reactions (ARs) were
reported in 63 patients (82%); the most common were rash (23% of all), constipation
(18% of CML) and headache (11% of Ph+ALL patients). Seventeen patients developed 26
serious AR. Serious cardiovascular ARs were reported in 7 patients (coeliac artery
stenosis [in 2 patients], ischemic stroke [1], worsening hypertension [1], hypertension
[1], deep venous thrombosis [1], possible transient ischemic attack [1]). Eight deaths
were recorded, none related to ponatinib treatment. 56% of CML and 24% of Ph+ALL patients
discontinued ponatinib treatment due to an AR.
Summary/Conclusion: This real-world Belgian registry over 5 years supports the use
of ponatinib in CML and Ph+ALL patients resistant or intolerant to previous TKIs or
carrying the T315I mutation. Results obtained in routine clinical practice are in
concordance with clinical trials, including PACE, in which CML patients had durable
and clinically meaningful responses to ponatinib.
Funding: Incyte Biosciences Benelux BV
Acknowledgements: Medical writing support was provided by Adina Truta (Modis)
P700: TREATMENT-FREE REMISSION (TFR) AFTER DASATINIB IN PATIENTS WITH CHRONIC MYELOID
LEUKEMIA IN CHRONIC PHASE (CML-CP) AND DEEP MOLECULAR RESPONSE (DMR): FINAL 5-YEAR
RESULTS OF DASFREE
N. P. Shah1,*, V. García-Gutiérrez2, A. Jiménez-Velasco3, S. Saussele4, D. Rea5, F.-X.
Mahon6, M. Y. Levy7, M. T. Gómez-Casares8, M. J. Mauro9, O. Sy10, P. Martin-Regueira10,
J. H. Lipton11
1UCSF School of Medicine, San Francisco, United States of America; 2Servicio Hematología
y Hemoterapia, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid; 3Hospital Universitario
Carlos Haya, Malaga, Spain; 4Medizinische Fakultät Mannheim der Universität Heidelberg,
Mannheim, Germany; 5Hôpital Saint-Louis, Paris; 6Institut Bergonié Cancer Center,
Université Bordeaux, Bordeaux, France; 7Baylor Charles A. Sammons Cancer Center, Dallas,
United States of America; 8Hospital Universitario de Gran Canaria Dr. Negrín, Las
Palmas de Gran Canaria, Spain; 9Memorial Sloan Kettering Cancer Center, New York;
10Bristol Myers Squibb, Princeton, United States of America; 11Princess Margaret Cancer
Centre, University of Toronto, Toronto, Canada
Background: Patients (pts) with CML-CP who have a DMR are eligible to discontinue
treatment (Tx) to attempt TFR. DASFREE (NCT01850004) is a single-arm, open-label,
phase 2 trial evaluating dasatinib discontinuation in pts with a stable DMR. At the
2-year (y) follow-up, 46% of pts remained in TFR; in a multivariate analysis, longer
duration of prior dasatinib, first-line (1L) dasatinib, and older age were significantly
associated with TFR maintenance (Shah et al. Leuk Lymphoma 2019).
Aims: To report the final 5-y results assessing TFR maintenance after dasatinib discontinuation.
Methods: Eligible pts were aged ≥18 y, received dasatinib Tx as 1L or subsequent therapy
for ≥2 y, and had a DMR (MR4.5 or BCR-ABL ≥0.0032% on the international scale, confirmed
during pre-screening and with 2 central lab assessments 3 months [mo] apart) for ≥1
y prior to study entry. Pts discontinued dasatinib and were monitored for up to 5
y. All pts provided written informed consent. If loss of major molecular response
(MMR) occurred, pts restarted dasatinib at the same dose as at enrollment. A univariate
analysis was conducted to identify baseline correlates of maintaining TFR. Rate of
TFR maintenance (event-free survival) in the extended follow-up, rate of MMR recapture
after relapse, identification of predictive factors for TFR maintenance, and safety
were all key secondary or exploratory endpoints.
Results: A total of 84 pts discontinued dasatinib. At a minimum follow-up of 60 mo
(database lock, Dec 2021), 44% (n=37) of pts remained in TFR and the remaining 56%
(n=47) had lost MMR and restarted Tx. No relapses occurred later than 39 mo after
discontinuation. Baseline characteristics were balanced between pts who remained in
TFR versus those who restarted Tx, except for age (65% of pts who remained in TFR
vs 85% who restarted Tx were aged <65 y). Among enrolled pts, 24 discontinued the
study early (4 due to drug-related adverse events [AEs]; none due to death); 60 discontinued
as planned per protocol (end of study). Patients who discontinued from the study were
evaluated for response regained before discontinuation. All evaluable pts (n=46; one
patient withdrew from the study 1 month after restarting Tx and did not undergo molecular
assessment) who restarted Tx regained MMR and MR4.5 in a median (range) time of 1.9
mo (0.9–3.7) and 3.3 mo (1.5–29.6), respectively. The 5-y TFR rate was 43.8% (95%
CI, 33.1–54.4; Figure). A univariate analysis of baseline correlates identified older
age and 1L dasatinib as significantly associated with maintaining TFR. Musculoskeletal
and connective tissue disorders (any grade) were experienced by 39% of pts (n=33).
The most common any-grade AEs were arthralgia (18%) and hypertension (13%). There
were no deaths due to CML. Withdrawal events (n=15) were experienced by 9 pts (11%)
after a median (range) of 3.7 (<1–18) mo from dasatinib discontinuation, with no new
events beyond 18 months of study follow-up; 10 events resolved in a median (range)
of 5.98 (<1–70) mo.
Image:
Summary/Conclusion: Discontinuation of dasatinib after 1L therapy and beyond is a
viable option for pts with CML-CP in sustained DMR. About half of the pts who discontinued
dasatinib maintained TFR at 5 y. Re-treatment after relapse was successful; all evaluable
pts who lost MMR regained MMR and MR4.5 after therapy was reinitiated. The overall
safety profile was consistent with the known safety profile of dasatinib and with
the 2-y results.
P701: COVID-19 IN PATIENTS WITH CHRONIC MYELOID LEUKEMIA IN TREATMENT-FREE REMISSION:
DISEASE SEVERITY AND IMPACT ON TFR STATUS
S. Saußele1, N. Evans2, F. E. Nicolini3, E. Chelysheva4, D. Réa5, F. Castagnetti6,
S. Claudiani7, R. Rodrigues8, C. Kok9, T. Hughes2,9,*
1III. Med. Klinik, Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim,
Germany; 2International CML Foundation, London, United Kingdom; 3Hematology department,
Centre Léon Bérard and INSERM1052, Lyon, France; 4National Research Center for Hematology,
Moscow, Russia; 5Hematology department, Saint-Louis Hospital, Paris, France; 6Istituto
di Ematologia “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna,
Italy; 7Department of Haematology, Hammersmith Hospital, Imperial College Healthcare
NHS Trust, London, United Kingdom; 8Departamento de Hematologia, Instituto Português
de Oncologia de Lisboa - IPO, Lisbon, Portugal; 9Precision Medicine Theme, South Australian
Health and Medical Research Institute, Adelaide, Australia
Background: Severe SARS-CoV-2 infections associated with high mortality rates are
reported in a higher percentage of patients (pts) with hematologic malignancies compared
to general population. In chronic myeloid leukemia (CML), pts with uncontrolled disease
have a higher mortality risk. The impact of SARS-CoV-2 infection on CML pts in treatment-free
remission (TFR) has not been studied so far. In particular, as immune control of residual
disease may be important for TFR, the concern is that the infection could induce loss
of TFR.
Aims: To evaluate the outcome of SARS-CoV-2 infection in CML pts in TFR and assess
any impact on maintenance of TFR.
Methods: From March 2020 to December 2021, the CANDID study organized by the international
CML Foundation has collected data on COVID-19 positive CML pts worldwide. Details
on the registry were presented recently (Pagano ASH 2021). For this sub-analysis on
pts in TFR additional information were collected including; molecular remission status
(BCR::ABL1 ratios) before, during and after SARS-CoV-2 infection covering at least
6 months.
For molecular analyses, BCR::ABL1 ratios were classified according to Cross et al
(Leukemia 2015). In addition, ratios of 0% without indication of sensitivity were
allocated as MR4 i.e. 0.01%IS. PCR outlier results were identified using the ROUT
method by nonlinear regression with a maximum false discovery rate (FDR) of 1% (Motulsky
et al 2006). Time to molecular relapse (MR) was measured from the date of COVID-19
diagnosis to the date of MR defined as loss of major molecular remission (MMR, BCR::ABL1
>0.1%IS) or the date of last molecular test. Molecular relapse-free survival (MRFS)
and overall survival (OS) were estimated with the Kaplan-Meier method. The statistical
difference between groups was performed using log-rank test.
Results: By December 2021, 1050 COVID-19 positive CML pts were registered. 95 pts
were in TFR at the time point of SARS-CoV-2 infection of which 89 (93.68%) recovered
and 6 deceased (6.32%). Median age of TFR pts was 57 years, male were 51 (53.68%).
Median time from CML diagnosis to reporting date was 13 years (range 3.7-27.0 years).
TFR duration was 2.83 years in median (range 0.5 months – 10.1 years) including 19
pts with a duration < 1 year.
From the 89 recovered TFR pts, 74 pts completed the 6-month follow up (83%), a further
6 pts with molecular follow-up of 3-5 months after COVID-19 diagnosis were still in
TFR, 9 pts were lost to follow-up.
Of 74 pts with complete reports, 69 pts remained in TFR (93%) and 5 pts lost TFR.
For 71 pts, PCR results were obtained before, during and after infection. With the
ROUT method 10 pts demonstrated outlier PCR tests, 61 pts demonstrated stable PCR
results. There was no statistically significant difference in PCR results before and
during/after infection (p>0.2). MRFS for these 71 pts 15 months after COVID-19 diagnosis
was 86%. Probability of TFR loss was higher in pts with a TFR duration < 6 months
compared to pts with TFR duration >6 months (27% vs 10%, Fig 1A). Additionally, there
were no statistically differences in hospitalization rate (16% vs 23%, p=0.12) and
severity of COVID-19 symptoms (12.6% vs 12%, p=0.87) comparing TFR and TKI treated
pts. OS of COVID-19 positive TFR pts did not differ from COVID-19 positive pts on
TKI therapy (HR 1.1, CI 0.47-2.54) (Fig 1B).
Image:
Summary/Conclusion: In this sub-analysis of the CANDID study, CML pts in TFR had similar
severity and survival to CML pts who were on TKI therapy and there was no evidence
of an increased risk of TFR loss after SARS-CoV-2 infection.
P702: FIRST INTERIM RESULTS OF THE PROSPECTIVE STUDY OF DOSE REDUCTION AND DISCONTINUATION
OF TYROSINE KINASE INHIBITORS (READIT-2020) IN CHRONIC MYELOID LEUKEMIA PATIENTS WITH
DEEP MOLECULAR RESPONSE
M. Gurianova1,*, O. Shukhov1, E. Chelysheva1, A. Petrova1, A. Bykova1, I. Nemchenko1,
E. Kuzmina1, N. Tsyba1, L. Gavrilova2, E. Stepanova1, A. Kohno1, A. Turkina1
1National Research Centre for Hematology, Moscow; 2Republican Clinical Hospital №4,
Saransk, Russia
Background: The feasibility of the one-step tyrosine kinase inhibitors (TKI) dose
reduction before treatment-free remission (TFR) phase in chronic myeloid leukemia
(CML) patients (pts) has been confirmed in several trials.There are no data of the
prospective two-step dose reduction trials of different TKIs followed up with TFR
observation.The experience of resuming TKIs at reduced doses after the molecular relapse
is limited as well.
Aims: To analyze the interim data of the survival without loss of major molecular
response (MMR,BCR::ABL1≤0,1%) after TKI stop in CML pts taking reduced-dose TKI therapy
and the probability of MMR and deep MR (DMR, BCR::ABL1≤0.01%) recovery after resuming
TKIs at reduced doses (Clinicaltrials.gov NCT 04578847).
Methods: The prospective trial had 2 phase:1)TKI dose reduction phase for at least
12 months (mo),2)TFR phase for at least 24 mo.TKI dose reduction consisted of 2 steps
each lasting for 6 mo.The inclusion of pts was possible at any step if they met the
key inclusion criteria:CML in chronic phase,age ≥18 years (y),TKI therapy duration≥
3 y,MMR and DMR duration≥2 y and ≥1 y.The inclusion into TFR was done after the dose
reduction phase in pts with a DMR duration≥2 y and ≥MR4.5 at the time of TKI stop.In
case of MMR loss the TKI dose was increased by +1 level from the dose at which the
MMR loss developed.
Results: A total of 103 CML pts were included from Dec.2019 till Dec.2021 at different
trial phases (Tab 1). Female 60%, Median (Me) age at diagnosis and at inclusion 45
y (23-74 y) and 51 y (23-74 y),respectively;ELTS score 61%:17%:1%, 21% for low,intermediate,high
and unknown risk group.A history of at least one TKI stop was in 29 pts.
Me TKI duration was 7 y (3-19,7 y);Me duration of MMR and DMR was 3,3 y (2-126 y)
and 2,5 y (1,2-10,5 y), respectively.
At baseline 69 (67%) pts received imatinib (IM) and 34 (33%) pts-second-generation
(2G)TKI (Tab 1). The reasons for 2GTKI switch were treatment failure (n=12),drug toxicity
(n=25),others (n=1).
Sixty-four pts completed the 1st dose reduction step lasting for 6 mo. There was no
MMR loss after the 1st dose reduction step. Four pts lost DMR on IM 300 mg,but three
pts restored DMR in 3 mo at the same dose thereafter.
Fifty-seven pts completed the 6 mo 2nd step of dose reduction.Two pts lost MMR on
IM 200 mg.Nine pts lost DMR (without MMR loss):6 pts on IM 200 mg,1 pt on dasatinib
(DAS) 25 mg and 2 pts on nilotinib (NIL) 200 mg.
Forty-nine pts were included in TFR phase,16 pts (32,6%) had a history of at least
one TKI stop.Eight pts had a history of resistance to TKI therapy.Me follow-up after
TKI discontinuation was 10 mo (1-24 mo).
The survival without MMR loss was 61% and 41% in pts with 1st and 2nd TKI stop,respectively
(Fig 1). The survival without MMR loss was 54% after 12 mo in total group.
TKI therapy was resumed at reduced doses after MMR loss:IM 200 mg(n=12),NIL 200 mg
(n=4),DAS 25 mg (n=2);BOS 200 mg (n=3).The probability of recovery of MMR and DMR
was 86% and 83% after 6 mo resumption of TKIs at reduced doses (Fig 2).
Image:
Summary/Conclusion: The 2-step dose reduction of TKIs before TFR phase is a promising
approach for CML pts.Use of the reduced TKI doses in pts with DMR lasting for≥1 y
is a safe treatment option under the strict molecular monitoring. The 61% survival
rate without MMR loss in CML pts having the 1st TKI discontinuation after the dose
reduction phase is encouraging.The high probability of MMR and DMR recovery after
resuming TKIs at reduced doses allows to consider this possibility in case of the
molecular relapse.
P703: ASSOCIATION BETWEEN BARIATRIC SURGERY AND OUTCOMES IN PATIENTS WITH CHRONIC
MYELOID LEUKEMIA TREATED WITH ORAL TYROSINE KINASE INHIBITORS
F. Haddad1,*, H. Kantarjian1, E. Jabbour1, N. Short1, A. Bidikian1, J. Ning2, L. Xiao2,
N. Pemmaraju1, K. Marx1, F. Ravandi1, K. Sasaki1, G. Issa1
1Leukemia; 2Statistics, The University of Texas MD Anderson Cancer Center, Houston,
United States of America
Background: Bariatric surgery is widely used in patients (pts) with morbid obesity
with great success in reduction of associated metabolic complications. However, it
can also alter the pharmacokinetics of oral medications. Oral tyrosine kinase inhibitors
(TKIs) represent the mainstay of chronic myeloid leukemia (CML) therapy. The impact
of bariatric surgery on CML treatment outcomes is largely unknown.
Aims: To evaluate the clinical impact of bariatric surgery on outcomes of CML pts
treated with TKIs.
Methods: In a retrospective analysis, we screened pts with CML treated at our institution
and identified those who had any type of bariatric surgery including gastric bypass,
gastric sleeve or gastric banding. We then compared their responses and outcomes to
a control cohort of pts without history of bariatric surgery, using propensity score
matching for Sokal Risk and body mass index (BMI) at a 2:1 ratio. In addition to cytogenetic
and molecular responses, we assessed times to achieving responses and BCR::ABL1 halving
times to investigate differences in response dynamics. We assessed comorbidities using
the Charlson Comorbidity Index. Event-free survival (EFS) was measured from treatment
start to loss of response, progression, or death, whereas failure-free survival (FFS)
additionally accounted for therapy discontinuation for other reasons such as intolerance.
Overall survival (OS) was measured from treatment start date to death or censored
at last follow-up. Univariate and multivariate analyses (MVA) were used to assess
the association between characteristics and survival outcomes.
Results: We identified 28 pts with CML and a history of bariatric surgery (22 pts
had their surgery before the diagnosis of CML) and 56 pts as a matched control. Their
baseline characteristics are summarized below.
Among pts with bariatric surgery, 61% required >1 TKI throughout the course of their
disease due to intolerance or resistance, compared with 25% of the control group (P=0.002).
BCR::ABL1 halving time was higher in the bariatric surgery group vs control (26 days
vs 13 days; P<0.0001), suggesting slower response dynamics. The median time to achieve
complete cytogenetic response (CCyR) and major molecular response (MMR) was significantly
longer in pts with history of bariatric surgery: 6 months vs 3 months (P<0.0001) and
12 months vs 6 months (P=0.001), respectively. Bariatric surgery was associated with
inferior EFS (10-year: 31% vs 69%; P=0.009) and FFS (10-year: 16% vs 54%; P<0.0001)
compared with control, with a trend for worse OS (10-year: 65% vs 85%; P=0.09) (Figure
1).
We subsequently conducted univariate and MVA and assessed the impact of bariatric
surgery and covariates such as sex, BMI, Sokal risk, TKI used, age and use of proton
pump inhibitors. Bariatric surgery was the only independent predictor for the risk
of treatment failure (hazard ratio [HR] 5.95, 95% CI 2.38-14.89; P=0.0001) or EFS
(HR 3.6, 95% CI 1.38-7.91; P=0.007) in the MVA but did not independently affect the
risk of death. The Charlson Comorbidity Index was the only independent predictor for
the risk of death (HR 1.65, 95% CI 1.10-2.48; P=0.02).
Image:
Summary/Conclusion: Bariatric surgery is associated with slower responses to TKIs
and higher rates of treatment failure. There is an unmet need to design treatment
strategies for these patients. Although not readily available in the clinical setting,
studies measuring drug level in these patients are needed to assess which TKI has
a better bioavailability, which in turn could translate to improved outcomes.
P704: ASCIMINIB PROVIDES DURABLE MOLECULAR RESPONSES IN PATIENTS (PTS) WITH CHRONIC
MYELOID LEUKEMIA IN CHRONIC PHASE (CML-CP) WITH THE T315I MUTATION: UPDATED EFFICACY
AND SAFETY DATA FROM A PHASE I TRIAL
T. Hughes1,*, J. E. Cortes2, D. Réa3, M. J. Mauro4, A. Hochhaus5, D.-W. Kim6, K. Sasaki7,
F. Lang8, M. C. Heinrich9, M. Breccia10, M. Deininger11, Y.-T. Goh12, J. J. Janssen13,
M. Talpaz14, V. Gómez García de Soria15, P. le Coutre16, S. Kapoor17, S. Cacciatore18,
F. Polydoros18, N. Agrawal18, F.-X. Mahon19
1SA Pathology and South Australian Health and Medical Research Institute, University
of Adelaide, Adelaide, Australia; 2Georgia Cancer Center, Augusta, United States of
America; 3Hôpital Saint-Louis, Paris, France; 4Memorial Sloan Kettering Cancer Center,
New York, United States of America; 5Universitätsklinikum Jena, Jena, Germany; 6Uijeongbu
Eulji Medical Center, Geumo-dong, Uijeongbu-si, South Korea; 7The University of Texas
MD Anderson Cancer Center, Houston, United States of America; 8Goethe University Hospital,
Frankfurt, Germany; 9Portland VA Health Care System and OHSU Department of Medicine,
Division of Hematology and Oncology, Knight Cancer Institute, Portland, United States
of America; 10Policlinico Umberto I-Sapienza University, Rome, Italy; 11Versiti Blood
Research Institute, Milwaukee, United States of America; 12Singapore General Hospital,
Bukit Merah, Singapore; 13Amersterdam University Medical Centers, Amersterdam, Netherlands;
14University of Michigan Rogel Cancer Center, Ann Arbor, United States of America;
15Hospital Universitario La Princesa, Madrid, Spain; 16Charité - Universitätsmedizin
Berlin, Berlin, Germany; 17Novartis Pharmaceuticals Corporation, East Hanover, United
States of America; 18Novartis Pharma AG, Basel, Switzerland; 19Institut Bergonié,
Bordeaux, France
Background: In pts with CML, the BCR::ABL1 T315I mutation is associated with poor
clinical outcomes and confers resistance to previously approved ATP-competitive tyrosine
kinase inhibitors (TKIs). Until recently, ponatinib (PON) was the only TKI available
for these pts, but its use may be limited by associated cardiovascular events. In
the primary analysis of the phase I trial X2101, asciminib—the 1st BCR::ABL1 inhibitor
to Specifically Target the ABL Myristoyl Pocket (STAMP)—demonstrated efficacy and
a favorable safety profile in heavily pretreated pts with CML with T315I. These results
supported the FDA approval of asciminib as a new treatment option for pts with CML-CP
with T315I (NCCN 2021). We report updated efficacy and safety data in these pts (data
cutoff: January 6, 2021).
Aims: Provide updated safety and efficacy data for pts with CML-CP with T315I treated
with asciminib monotherapy 200 mg twice daily (BID) after added exposure.
Methods: Pts with CML-CP with T315I were enrolled if treated with ≥1 prior TKI and
no other effective therapy was available, provided informed consent, and received
asciminib 200 mg BID.
Results: 48 pts with T315I were included; 2 (4.2%) pts had additional BCR::ABL1 mutations
at baseline. Eight (16.7%), 15 (31.3%) and 25 (52.1%) pts received 1, 2, and ≥3 prior
TKIs, respectively. At data cutoff, treatment was ongoing in more than half (27 [56.3%])
of pts; the predominant reason for treatment discontinuation was physician’s decision
(11 [22.9%]), mainly due to lack of efficacy.
Of the 48 pts, 45 were evaluable (BCR::ABL1
IS >0.1% at baseline) for major molecular response (MMR); 3 were excluded for BCR::ABL1
atypical transcripts. Among evaluable pts, 19 (42.2%) achieved MMR by wk 24 and 22
(48.9%) by wk 96; 19 were still in MMR at the cutoff date. Evaluable pts included
26 PON-pretreated and 19 PON-naive pts; 34.6% and 68.4%, respectively, achieved MMR
by the cutoff date (Table). The probability of pts maintaining MMR for ≥96 wks was
84% (95% CI, 68.1-100.0). Thirteen (28.9%) and 11 (24.4%) pts achieved MR4 and MR4.5,
respectively. Twenty (54.1%) and 23 (62.2%) of 37 pts with BCR::ABL1
IS >1% at baseline achieved BCR::ABL1
IS ≤1% by wk 48 and 96, respectively.
The median duration of exposure was 2.08 (range, 0.04-4.13) yrs with more than half
(27 [56.3%]) of pts receiving treatment for ≥96 wks; the median daily dose intensity
was 398.3 (range, 179-400) mg/day. The safety/tolerability profile of asciminib remained
favorable after ≈9 months of added follow-up (Table). The most common (≥5%) grade
≥3 adverse events (AEs) were lipase increase (18.8%, all asymptomatic elevations),
thrombocytopenia (14.6%), and vomiting, ALT increase, abdominal pain, hypertension,
anemia, neutropenia, and neutrophil count decrease (6.3% each). Arterial occlusive
events occurred in 4 (8.3%) pts; none led to dose adjustment/interruption/discontinuation.
AEs leading to discontinuation were reported in 2 new pts since the previous data
cutoff; both pts discontinued and died due to COVID-19. These were the only study
deaths reported in this pt population.
Image:
Summary/Conclusion: Asciminib monotherapy 200 mg BID exhibited a sustained, favorable
safety profile after added exposure with no new safety signals in pts with CML-CP
with T315I—a population with high unmet medical need. The clinical efficacy of asciminib
is demonstrated by the high proportion of pts achieving durable MMR and BCR::ABL1
IS ≤ 1%. The updated analysis confirms asciminib as a treatment option for pts with
CML-CP with T315I, including those for whom treatment with PON has failed.
P705: MULTI-CENTER CHART REVIEW STUDY EXAMINING TREATMENT PATTERNS AND CLINICAL OUTCOMES
AMONG PATIENTS WITH CHRONIC PHASE (CP) CHRONIC MYELOID LEUKEMIA (CML) TREATED IN THIRD-LINE
(3L) OR LATER IN FRANCE
F. E. Nicolini1, G. Etienne2, F. Huguet3, M. Gu4, C. Bouvier1, A. Yocolly5, R. Favier6,
M. Trancart6, L. Huynh4,*
1Centre Leon Berard, Lyon; 2Institut Bergonié, Bordeaux; 3Institut Universitaire de
Cancérologie de Toulouse, Toulouse, France; 4Analysis Group, Inc., Boston, MA; 5Novartis
Services, Inc., East Hanover, NJ, United States of America; 6Novartis Oncologie, Rueil-Malmaison,
France
Background: The standard of care for CML has substantially evolved over a relatively
short period of time; pharmacologic inhibition of ABL1 with adenosine triphosphate
(ATP)-competitive tyrosine kinase inhibitors (TKIs) remains the cornerstone of modern
treatment. Clinical guidelines for CP-CML recommend that patients who have failed
≥2 prior TKIs switch to an alternative 2nd or 3rd generation TKI. However, up to 65%
of these patients have prior exposure to ATP-competitive TKIs and history of either
TKI intolerance or resistance.
Aims: This retrospective, multi-center, chart review study aimed to characterize the
disease burden, treatment patterns, and clinical outcomes of CP-CML patients who have
failed ≥2 TKIs.
Methods: De-identified demographic and clinical data for adult patients diagnosed
with CP-CML treated in 3L or later at three reference centers in France were abstracted
from medical charts using electronic case report forms. Descriptive statistics were
summarized for patient characteristics, clinical outcomes during 3L treatment, and
adverse events. Molecular data were standardized and expressed in the international
scale (IS). The cumulative incidence of patients achieving molecular response (major
molecular response [MMR, 0.01% IS ≤0.1%] or deep molecular response [MR4.0, 0.0032%
< BCR–ABL1IS ≤0.01% or MR4.5, BCR–ABL1IS ≤0.0032%]) were summarized at specific time
points. Progression-free survival (PFS) and overall survival (OS) from 3L initiation
were examined using the Kaplan–Meier (KM) method.
Results: Medical data for 157 CP-CML patients were assessed; the mean follow-up was
66.9 months, the median age at 3L initiation was 62.1 years, 56% were male, and 90%
had major BCR-ABL1 rearrangement (among the 89 patients with mutation status assessed,
7 [8%] had BCR-ABL1 T315I mutation, 3 [3%] had M244V, and 3 [3%] had F359I). According
to EUTOS long-term survival score, 40% of patients had low risk, 25% had intermediate
risk, and 11% had high risk of death due to CML. Cardiovascular diseases were present
in 55% of patients at 3L; mean systolic and diastolic blood pressure were 140.9 and
78.9 mmHg, respectively. Mean ± SD low-density lipoprotein cholesterol was 141.0 ±
54.0 mg/dl. Median duration of 3L therapy was 17.0 months. TKIs received in 3L were
dasatinib (32%), nilotinib (19%), imatinib (18%), ponatinib (17%), and bosutinib (14%).
Treatment-free remission was initiated by 16 (10%) patients. In patients with documented
responses, 42% patients achieved MMR, 27% achieved MR4.0, and 14% achieved MR4.5 at
12 months. Although median PFS and OS were not reached as of data collection, 19 (12%)
patients died due to disease progression (n=7), toxicity (n=1), or other reasons (n=10)
and 1 patient had an unknown cause of death. Approximately 50% of patients discontinued
treatment; 37% had ≥4 lines of treatment and 16% had ≥5 lines. The primary reasons
for discontinuation (not mutually exclusive) were intolerance (54/78 [69%]), resistance
(18/78 [23%]), and signs of ineffectiveness (14/78 [18%]). AEs were documented for
139/157 patients (89%). The mean number of AEs per patient was 2.7; infections (18%)
and asthenia (13%) were the more commonly documented AEs.
Summary/Conclusion: CP-CML patients continue to experience a substantial disease burden
and poor prognosis after 3L treatment with available TKIs, underscoring the need for
novel therapies that are well tolerated and can achieve durable responses.
P706: ASCIMINIB USE IN CML: THE UK EXPERIENCE
A. Innes1,2,*, V. Orovboni3, S. Claudiani1, F. Fernando1, A. Khan1, J. Byrne4, P.
Gallipoli5, M. Copland6, G. Horne6, C. Arnold7, A. Collins8, N. Cunningham7, A. Danga9,
R. Frewin10, P. Garland11, G. Hannah12, S. Hassan13, S. Makkuni14, K. Rothwell15,
L. Foroni2, C. Hayden3, J. Apperley2, D. Milojkovic1
1Department of Haematology, Hammersmith Hospital, Imperial College Healthcare NHS
Trust; 2Centre for Haematology, Faculty of Medicine, Department of Immunology and
Inflammation, Imperial College London; 3SIHMDS (Molecular Laboratory), Hammersmith
Hospital, Imperial College Healthcare NHS Trust, London; 4Nottingham University Hospitals
NHS Trust, Nottingham; 5Cancer Research UK Barts Centre, London; 6Institute of Cancer
Sciences, Paul O’Gorman Research Centre, Gartnavel General Hospital, Glasgow; 7Belfast
City Hospital, Belfast; 8Norfolk and Norwich University Hospital NHS Foundation Trust,
Norwick; 9The Hillingdon Hospital, London; 10Gloucestershire Hospital NHS Trust, Gloucester;
11Princess Royal University Hospital; 12King’s College Hospital NHS Foundation Trust,
London; 13Queen’s Hospital, Romford, London; 14Mid and South Essex NHS Foundation
Trust, Essex, London; 15Calderdale and Huddersfield NHS Foundation Trust, Huddersfield,
United Kingdom
Background: Asciminb is an allosteric BCR-ABL1 inhibitor that has been available for
patients with chronic myeloid leukaemia (CML) in the United Kingdom under a managed
access program (MAP) from Novartis since 2016.
Aims: The aim of this project was to share real-world experience of asciminib use
in the UK.
Methods: Using a national survey proforma (REC reference: 21/HRA/2157), we collated
baseline, outcome and toxicity data on 44 patients treated with asciminib in the UK.
Responses were assess using BCR-ABL1 IS values and categorised by ELN criteria. Intolerances
were reported using CTCAEv5.0 criteria.
Results: The median age at treatment was 58 [22-88] years. Vascular comorbidities
were seen in 22 (50%) patients, the most frequent being hypertension (n=12, 27%),
peripheral vascular disease (n=6, 14%), AF (n=6, 14%) and ischaemic heart disease
(n=6, 14%). Chronic kidney disease was present in 7 (16%) patients. All but one patient
was in chronic phase at the time of treatment, but tyrosine kinase domain mutations
(TKDM) were common prior to treatment initiation (T315I in 11/44 (25%), other TKDM
in 8/44 (18%)). The median number of prior TKIs were 4 [2-5], with 44% of prior lines
stopped for resistance, and 56% for intolerance. The most recent TKI was discontinued
for resistance in 14 (32%) and intolerance in 30 (68%). Eight (18%) patients had developed
pleural effusions on a previous TKIs. Most patients had previously received ponatinib
(n=30, 68%) which has been discontinued for resistance in 8 (27%) and intolerance
in 22 (73%), with 7 (16%) vascular events on treatment. Five (11%) patients had received
prior allogeneic stem cell transplant.
At the time of data cut-off, 25 (57%) patients remained on treatment with a median
treatment duration of 20 [3-51] months. Reason for treatment discontinuation was resistance
(n=7, 16%), intolerance (n= 7, 16%), pregnancy (n=1, 2%), treatment-free remission
(n=1, 2%) and non-compliance (n=1, 2%). Two patients died whilst on treatment of unrelated
causes. The median daily dose delivered was 240mg [20-400] in patients with T315I-TKDM
and 80mg [20-80] in others.
While 11 patients had no significant response (including those with early treatment
discontinuation for intolerance), 23 (53%) patients achieved MR3 or better, with 14
(32%) achieving MR4 or better. Of 9 patients in MR3 or better at asciminib initiation,
3 (33%) did not tolerate treatment, 2 (22%) maintained their response, and 4 (44%)
deepened their response by one category or more. The presence of a non-T315I TKDM
was associated with a lower incidence of achieving MR3, with 1 of 6 (16%) evaluable
patients in this group achieving MR3 or better, compared with 22 of 31 (71%) evaluable
patients with no TKDM or a T315I-only TKDM (p=0.04) (Table1). Haematological toxicity
was seen in 17 (38%) patients, and was grade 3-4 in 6 (14%) (predominantly anaemia
and thrombocytopaenia). Non-haematological toxicity was reported in 19 (43%) patients
(most commonly fatigue (n=4), insomnia (n=3), bone pain (n=2), back pain (n=2), nausea
(n=2), fluid retention (n=2)), with only 2 (4%) patients experiencing grade 3-4 toxicity
(fatigue in both cases). No worsening of baseline vascular co-morbidity or renal dysfunction
was noted. One patient developed a new pleural effusion on the 200mg bd dose, which
did not recur on dose reduction.
Image:
Summary/Conclusion: These data show that asciminib is a well-tolerated, effective
treatment for patients who are resistant to or intolerant of multiple TKIs, many of
whom often have a number of co-morbidities including vascular disease.
P707: DOSE MODIFICATION DYNAMICS OF PONATINIB IN PATIENTS WITH CHRONIC-PHASE CHRONIC
MYELOID LEUKEMIA (CP-CML) FROM THE PACE AND OPTIC TRIALS
J. Apperley1,*, H. Kantarjian2, M. Deininger3, E. Abruzzese4, J. Cortes5, C. Chuah6,
D. J. DeAngelo7, J. DiPersio8, A. Hochhaus9, J. Lipton10, F. Nicolini11, J. Pinilla-Ibarz12,
D. Rea13, G. Rosti14, P. Rousselot15, M. Mauro16, N. Shah17, M. Talpaz18, A. Vorog19,
X. Ren19, E. Jabbour2
1Imperial College London, London, United Kingdom; 2The University of Texas MD Anderson
Cancer Center, Houston, TX; 3University of Utah, Huntsman Cancer Institute, Salt Lake
City, UT, United States of America; 4S. Eugenio Hospital, Tor Vergata University,
Rome, Italy; 5Georgia Cancer Center at Augusta University, Augusta, GA, United States
of America; 6Singapore General Hospital, Duke-NUS Medical School, Singapore, Singapore;
7Dana-Farber Cancer Institute, Boston, MA; 8Washington University School of Medicine,
St. Louis, MO, United States of America; 9Universitätsklinikum Jena, Jena, Germany;
10Princess Margaret Cancer Centre, Toronto, Ontario, Canada; 11Centre Léon Bérard,
Lyon, France; 12H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United
States of America; 13Hôpital Saint-Louis, Paris, France; 14IRCCS Istituto Romagnolo
per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola (FC), Italy; 15Hospital Mignot
University de Versailles Saint-Quentin-en-Yvelines, Paris, France; 16Memorial Sloan
Kettering, New York, NY; 17University of California San Francisco, San Francisco,
CA; 18Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI; 19Takeda
Development Center Americas, Inc., Lexington, MA, United States of America
Background: Ponatinib, a potent oral, third-generation tyrosine kinase inhibitor (TKI),
is FDA approved for treatment of patients with relapsed/refractory CML. In the pivotal
Phase 2 PACE (Ponatinib Ph+ ALL and CML Evaluation) trial (NCT01207440), patients
with resistant/intolerant CP-CML demonstrated deep, lasting responses to ponatinib
45 mg once daily. The Phase 2 OPTIC (Optimizing Ponatinib Treatment in CP-CML) trial
(NCT02467270) prospectively evaluated a response-based dose-reduction strategy in
an attempt to optimize the dose schedule of ponatinib in patients with CP-CML resistant
to second-generation (2G) BCR::ABL1 TKI therapy or with a T315I mutation.
Aims: Since the unique dosing strategies in the PACE and OPTIC trials provide the
opportunity to closely evaluate the dose and schedule of ponatinib, we conducted an
in-depth analysis of dosing dynamics between the 2 trials and compare efficacy and
safety outcomes.
Methods: Adults with resistant/intolerant CP-CML from the PACE and OPTIC trials were
enrolled. In PACE, patients received an initial dose of ponatinib 45 mg once daily,
in OPTIC, patients were randomly assigned (1:1:1) to an initial oral dose of ponatinib
45 mg, 30 mg, or 15 mg once daily. In PACE, proactive dose reductions were mandated
in ≈2 years from initiation of first patient (in 2013) as arterial occlusive events
(AOEs) emerged as notable adverse events (AEs). OPTIC was designed to incorporate
a mandatory response-based dose-reduction strategy; patients in the 45-mg and 30-mg
cohorts in OPTIC achieving ≤1% BCR::ABL1
IS reduced their dose to 15 mg once daily; doses also were reduced to manage AEs.
This analysis includes data from patients with CP-CML in PACE and from the 45-mg starting
dose cohort in OPTIC. Efficacy outcomes include ≤1% BCR::ABL1
IS, progression-free survival (PFS), and overall survival (OS). Safety outcomes include
treatment-emergent adverse events (TEAEs) and treatment-emergent AOE (TE-AOE) rates.
Results: Together, 364 patients with CP-CML had ≥1 prior 2G TKI or had a T315I mutation
and received a starting dose of ponatinib 45 mg (PACE, n=270; OPTIC, n=94). Percentages
of patients receiving ≥2 or ≥3 prior TKIs were 93% and 60% in PACE and 99% and 53%
in OPTIC, respectively. Percentages of patients with vascular disorders were 44% in
PACE and 32% in OPTIC. Median follow-up was 57 months (PACE) and 32 months (OPTIC).
≤1% BCR::ABL1
IS response by 24 months was 52% in PACE and 56% in OPTIC, 2-year PFS was 68% in PACE
and 80% in OPTIC, and 2-year OS was 86% in PACE and 91% in OPTIC. Median time to ≤1%
BCR::ABL1
IS response was 5.6 months in PACE and 6 months in OPTIC. Median duration of response
was not reached in either trial. Median time to dose reduction for AEs was 2.85 months
in PACE and 3.64 months in OPTIC and dose reductions due to AEs occurred in 82% of
patients in PACE and 46% in OPTIC. Median time on therapy was 12.6 months in PACE
and 19.5 months in OPTIC. Per 100-patient years, exposure-adjusted TE-AOEs were 15.8
events at 0 to <1 year in PACE and 7.6 events at 0 to <1 year in OPTIC. Individual
dosing dynamics by safety and efficacy will be presented.
Summary/Conclusion: The response-based dose-reduction strategy in OPTIC resulted in
comparable or better efficacy outcomes, fewer dose reductions related to AEs, fewer
exposure-adjusted TE-AOEs, and longer median time on therapy compared with PACE. These
results further demonstrate the benefit of the response-based dosing regimen used
in OPTIC.
This abstract is an encore from the American Society of Hematology 2021 Annual Meeting.
P708: CANADIAN AND RUSSIAN EXPERIENCES OF ASCIMINIB IN CHRONIC MYELOID LEUKEMIA (CML)
PATIENTS WHO FAILED MULTIPLE LINES OF TYROSINE KINASE INHIBITOR (TKI) THERAPY.
F. Khadadah1,*, A. G. Turkina2, E. Lomaia3, E. V. Morozova4, O. A. Shukhov2, A. Petrova2,
T. Chitanava3, J. J. Vlasova5, E. Kuzmina2, I. Nemchenko2, E. Y. Chelysheva2, A. Bykova2,
M. Gurianova2, A. Xenocostas6, L. Busque7, K. Jamani8, S. Cerquozzi8, P. Kuruvilla9,
R. Kaedbey10, B. Leber10, S. Assouline11, D. D. H. Kim1
1Princess Margaret Cancer Centre, Toronto, Canada; 2National Medical Research Center
for Hematology, Moscow; 3Almazov National Medical Research Centre; 4. Raisa Gorbacheva
Memorial Research Institute of Children’s Oncology, Hematology and Transplantation;
5Raisa Gorbacheva Memorial Research Institute of Children’s Oncology, Hematology and
Transplantation, Saint-Petersburg, Russia; 6London Health Sciences Centre, Victoria
Hospital, London; 7Maisonneuve-Rosemont Hospital, Montreal; 8Tom Baker Cancer Centre,
Calgary; 9William Osler Health System, Brampton; 10Juravinski Cancer Center, Hamilton;
11Jewish General Hospital, Montreal, Canada
Background: Asciminib (ASC) is a novel, first in class ‘specifically targeting the
ABL myristate pocket (STAMP) inhibitor. Recent update of the ASCEMBL phase 3 data
showed a superior cumulative incidence of major molecular rate (MMR) of 33.2% with
ASC over bosutinib (BOS) at 18.6% by 48 weeks and higher MR4 rate (BCR-ABL1 ≤0.01%)
of 14.0% with ASC over 6.6% with BOS at 48 weeks in CML patients (pts) in chronic
phase (CP) who have failed at least two lines of TKI therapy.
Aims: Canadian and Russian groups individually reported their real-world experiences
of ASC therapy under the Managed Access Program in heavily pre-treated CML patients.
Methods: Data was collected, updated and merged from 80 CML pts treated with ASC between
Nov 2018 - Dec 2021 (n=57 in Russia; n=23 in Canada). Median age was 57 years (range
20-92). The median number of previous TKIs was 4 (range 2-6); Imatinib (n=74, 93%),
dasatinib (n=64, 80%), nilotinib (n=58, 73%), bosutinib (n=51, 64%), ponatinib (n=35,
44%), and others (n=13, 16%). Median duration from diagnosis to ASC treatment was
92 mo (months) (range 11-310). 27 (34%) pts had a past history of clinically significant
cardiovascular disease or high risk profile for cardiovascular disease. Of the 80
pts, 27 (34%) had a preexisting T315I mutation and 18 (23%) had a non-T315I mutation.
Pts failed previous TKI therapy due to A)resistance or suboptimal response (n=59;
74%) and B)intolerance (n=21; 26%). BCR-ABL qPCRs were monitored at each institution.
Achievement of MMR and molecular response of 4 log reduction (MR4) was assessed at
6 and 12 mo.
Results: With a median of 9 mo of follow-up (range 1-38), MMR was noted in 15/68 (22%)
and 14/36 pts (39%) evaluated at 6 and 12 mo, respectively. MR4 was noted in 11/68
(16%) and 9/36 pts (25%) at 6/12 mo. The cumulative incidence of MMR and MR4 was 32.6%
(20.9-44.8%) and 15.9% (7.9-26.3%) at 12 mo. The probability of freedom from treatment
failure (FTF) at 12 mo was 42.4% (27.6-56.4%), based on the ELN 2013 failure criteria
for 2nd line therapy or beyond.
In the overall population (n=80), FTF was significantly associated with reason for
switch to ASC (p=0.006), and there was a trend toward association of FTF with disease
phase (p=0.08) and ponatinib therapy (p=0.12), but not with ABL1 kinase domain mutation
(p=0.402), additional cytogenetic abnormality (p=0.908), nor cardiovascular risk profile
(p=0.345).
When analysis was restricted to the patients in CML-CP (n=65), a median duration of
FTF was calculated as 11.6 mo, while the 12 mo FTF rate was 46.8% (28.9-62.8%). The
patients previously treated with ponatinib (n=25) showed a 27.7% (8.4-51.4%) FTF rate
at 12 mo, which was lower than that in those naïve to ponatinib (n=40), 61.7% (34.9-80.2%;
p=0.023). Those switched to ASC for intolerance (n=21) showed higher 12 mo FTF rate
of 81.2% (41.5 -95.2%) compared to 29.4% (11.3-50.2%) in those with resistance (n=44;
p=0.008).
Fifteen pts discontinued ASC due to treatment failure (n=11), progression of disease
to blast phase (n=3) or grade 4 thrombocytopenia (n=1). No cardiovascular event was
noted.
Image:
Summary/Conclusion: This combined Canadian and Russian real-world experience of ASC
study includes heavily pretreated CML pts including 15 pts in advanced phase. The
MMR and MR4 rates were comparable in those in CP, while it was lower in non-CP pts.
The 12 mo FTF rate of 42.4% was comparable to the one reported from the ASCEMBL study,
57.7%, considering that this group includes more patients who had heavily pre-treated
with advanced disease.
*AGT and FK contributed equally as co-first author.
P709: CLINICAL OUTCOME OF ASCIMINIB TREATMENT IN A REAL-WORLD MULTI-RESISTANT CML
PATIENT POPULATION.
C. C. Kockerols1,*, J. J. Janssen2, N. M. Blijlevens3, S. K. Klein4, L. G. van Hussen-Daenen5,
G. N. van Gorkom6, W. M. Smit7, P. van Balen8, B. J. Biemond9, M. J. Cruijsen10, M.
F. Corsten11, P. A. te Boekhorst12, H. R. Koene13, G. L. van Sluis14, J. J. Cornelissen12,
P. E. Westerweel1
1Internal Medicine, Albert Schweitzer Hospital, Dordrecht; 2Hematology, Amsterdam
University Medical Center, location VUMC, Amsterdam; 3Hematology, Radboud University
Medical Center, Nijmegen; 4Hematology, University Medical Center Groningen, Groningen;
5Hematology, University Medical Center Utrecht, Utrecht; 6Hematology, Maastricht University
Medical Center, Maastricht; 7Hematology, Medisch Spectrum Twente, Enschede; 8Hematology,
Leiden University Medical Center, Leiden; 9Hematology, Amsterdam University Medical
Center, location AMC, Amsterdam; 10Hematology, Catharina Hospital, Eindhoven; 11Hematology,
Meander Medical Center, Amersfoort; 12Hematology, Erasmus University Medical Center,
Rotterdam; 13Hematology, St.-Antonius Hospital, Nieuwegein; 14Hematology, Isala, Zwolle,
Netherlands
Background: Asciminib is a first-in-class STAMP (Specifically Targeting the ABL Myristoyl
Pocket) tyrosine kinase inhibitor (TKI). Asciminib was found to have superior efficacy
compared to bosutinib in patients (pts) intolerant or refractory to ≥2 currently registered
TKI with good tolerability. While awaiting its EMA approval, asciminib is made available
by Novartis to pts with high clinical need in an early access programme (EAP) in The
Netherlands.
Aims: We aimed to assess treatment outcomes in this nationwide patient cohort.
Methods: Dutch hematologists who treated CML pts in the asciminib EAP were asked to
provide clinical data of consenting pts. Pts were included if ≥18 years and treated
in this asciminib EAP, thus outside clinical trials. Treatment failure was defined
as progression to advanced stage disease (AP/BC), not reaching a complete cytogenetic
response or BCR-ABL1 <1%IS (CCYR/MR2.0) (=primary) or losing previously achieved responses
(=secondary). Event-free survival (EFS) was defined as time from asciminib start until
allogeneic stem cell transplantation (SCT), progression to AP/BC or death; whatever
came first. Responses were assessed with the cumulative incidence competing risk (CICR)
method considering the competing event of TKI discontinuation for any cause.
Results: All hematologists agreed to participate and 53 out of 56 pts from 14 medical
centers consented and were included. Most pts (72%) had been exposed to ≥3 TKIs prior
to asciminib. The majority (79%) started with asciminib because of multi-resistance
(with or without intolerance) including 15 pts with primary ponatinib-failure, 15
pts harboring the T315I mutation and 3 pts with compound mutations. Two pts were treated
with asciminib post-SCT and were excluded from response analyses.
Median asciminib exposure was 7 months (IQR 3-16). Frequently reported adverse events
(AE) were cytopenia and folliculitis, classified as probably related to asciminib
and manageable generally without dose modification. Only one pt definitely discontinued
asciminib due to persistent anemia, but in the context of also a failing response.
Two vascular events were reported, one recurrent TIA therefore classified as unrelated
and one NSTEMI classified as possibly related to asciminib.
Sixteen pts discontinued asciminib, mostly because of treatment failure (primary n=8,
secondary n=2) or proceeding to SCT (n=5). Six out of 7 pts with AP/BC discontinued
asciminib within 6 months because of treatment failure/SCT.
The CI of maintaining or reaching CCYR/MR2.0 and MMR by 6 months for CP pts were 60%
and 44%, respectively. Of note, only one out of 10 pts in CP with primary ponatinib-failure
reached MR2.0 and none of these patients reached MMR. However, CP pts with secondary
ponatinib-failure responded well with CCYR/MR2.0 or better in 78% (7/9).
During asciminib treatment 5 pts progressed to advanced stage disease, 9 pts eventually
underwent SCT and 7 pts died of whom 6 with a CML-related cause. Overall survival
and EFS were 87% and 72%, with a median follow-up time of 10 and 8 months, respectively.
Image:
Summary/Conclusion: Our results show that asciminib is a well-tolerated novel treatment
option for CML with adequate response rates, even in a heavily pretreated multi-resistant
patient cohort. Most frequently reported AEs were manageable cytopenia and folliculitis.
No patient discontinued asciminib solely due to intolerance. The CI of maintaining
or reaching MR2.0/CCYR at 12 months was 57%. In our cohort, prior primary ponatinib-failure
generally heralded subsequent asciminib treatment failure, both in the presence and
absence of a T315I-mutation.
P710: CLINICAL STUDY ON THE RELATIONSHIP BETWEEN NON-ABL1 KINASE REGION MUTATION AND
TKI DRUG RESISTANCE AND DISEASE PROGRESSION IN CHRONIC MYELOID LEUKEMIA
W. xianwei1, L. xiaodong1,*
1Central Lab, Henan Cancer Hospital, Zhengzhou, China
Background: The emergence of small molecule tyrosine kinase inhibitors (The Tyrosine
Kinase Inhibitors, TKI) has gradually transformed chronic myelocytic leukemia (CML)
from a fatal malignant disease into a chronic disease, while ABL1 kinase Kinase Domain
(KD) mutation is the main reason for poor efficacy or resistance to TKIs. In recent
years, with the clinical application of Next Generation Sequencing (NGS) technology,
it has been found that CML patients are often accompanied by non-ABL1 kinase region
gene mutations, but whether these accompanying mutations are related to the efficacy
of TKIs and disease progression in CML? still uncertain. This study retrospectively
analyzed the correlation between CML with non-ABL1 KD mutation and TKI efficacy and
disease progression, in order to provide basis and scientific guidance for further
accurate diagnosis and treatment of CML patients.
Aims: To investigate the relationship between CML associated with non ABL1 Kinase
Domain (KD) mutation and TKI efficacy and disease progression.
Methods: 108 blood tumor related genes in 120 patients with CML were detected by Next-generation
sequencing (NGS). The non ABL1 KD mutations and their relationship with relevant clinical
information and ABL1 KD mutation were analyzed.
Results: Non ABL1KD mutations were detected in 58 patients (48%), and the mutation
proportion of patients with additional chromosome abnormalities (63%) was significantly
higher than that of patients without additional chromosome abnormalities (34%) (P=0.003).
In the analysis of mutation proportion in patients with different stages of CML, the
mutation proportion in blastic phase(73%), accelerated phase (67%) and chronic phase
(42%) decreased gradually (P=0.031). The proportion of WT1 mutation and transcriptional
regulation related mutations in patients with AP CML (WT1+: 17%, P=0.027; Transcriptional
regulatory mutations 42%, P=0.001) were higher than those in CP. In the analysis of
TKI treatment response, the proportion of mutations detected in the best response
group (30%), warning group (52%) and drug resistance group (61%) increased gradually
(P=0.018). The proportion of RUNX1 mutation in TKI treatment failure group (17%) was
higher than that in warning group (2%) (P=0.029). The proportion of ABL1 KD mutation
in CML patients with non ABL1 KD mutation (60%) was significantly higher than that
in patients without non ABL1 KD mutation (36%) (P=0.022).
Summary/Conclusion: Non ABL1 KD mutations are closely related to TKI efficacy and
disease progression, while WT1 and RUNX1 mutations may be new molecular targets for
accurate diagnosis of CML.
P711: KINETICS OF CITED2 GENE EXPRESSION IN CHRONIC MYELOID LEUKEMIA PATIENTS
B. Atef1,*, S. El-Ashwah1, L. M. Saleh2, H. Gawish3, M. Nasr1
1Hematology Unit, Internal Medicine Department; 2Hematology Unit, Clinical Pathology
Department; 3Diabetes & Endocrinology Unit, Internal Medicine Department, Faculty
of Medicine Mansoura University, Mansoura, Egypt
Background: In chronic myeloid leukemia (CML), BCR-ABL1 oncoprotein induces overexpression
of STAT5 and its target genes. Some of these genes are involved in regulating the
leukemic stem cells (LSCs) cell cycle. CITED2 gene controls the balance between the
proliferation and the quiescence states of the LSCs that resist tyrosine kinase inhibitors
(TKIs). Recently, studies reported that adding peroxisome proliferator‐activated receptor
γ agonists (approved for type 2 Diabetes mellitus treatment) to TKI therapy may decrease
the transcription of STAT5 and its target genes, eliminate the quiescent LSCs and
ameliorate the patients’ response.
Aims: The aim of the study is to clarify the value of combining the glitazones with
the TKIs in the treatment of denovo CML CP patients as regard the patients’ response
and effect of this combination therapy on the expression of CITED2 gene.
Methods: In a context of a clinical trial (approved by the ethical Committee of the
Faculty of Medicine Mansoura University), eligible denovo patients of CML chronic
phase (N=26) were treated with imatinib 400 mg combined and pioglitazone 15 mg (off
label use) daily for 6 months after having their consent. Responders continued on
same TKI and their sequential BCR/ABL Q-PCR was monitored. The non-responder patients
were switched to another line and excluded. Pre and post-treatment samples were collected
to assess the expression levels of CITED2 gene.
Results: The study group included 15 males (57.7%) and 11 females (42.3%) with a mean
age of 43.2 years. Their median BCR-ABL gene expression before treatment was 79% (range
= 0.63-380) that decreased to 3.65% (range = 0.05-20) and 0.750 % (range = 0.002-46)
on 3rd and 6th months post-treatment. Among the study group 100% of the patients achieved
hematological response (HR) at 3 months. At 6 months 65.4% of the patients had response
more than CCyR and 23% of them had MMR at 6 months respectively. The most frequent
complains among patients were increase body weight and edema (p value < 0.001). The
median of CITED2 gene pre-treatment and post-treatment expression levels were: 276.3
(range= 1-241221.7) and 2.6 (range= 0.006-86475.3) with a significant decrease in
expression (p=0.005) after the combined treatment.
Summary/Conclusion: Pioglitazone in combination with imatinib improved the patients’
response and downregulate the expression of the CITED2 overexpressed gene. Longer
follow up is planned and needed to confirm whether it can effectively induce more
deep molecular response among the patients or not.
P712: ASCIMINIB ITALIAN MANAGED ACCESS PROGRAM: EFFICACY PROFILE IN HEAVILY PRE-TREATED
CML PATIENTS
M. Breccia1,*, A. V. Russo Rossi2, B. Martino3, E. Abruzzese4, M. Annunziata5, G.
Binotto6, A. Ermacora7, C. Fava8, L. Giaccone9, V. Giai10, A. P. Nardozza11, P. Coco11,
A. Gozzini12, L. Levato13, A. Lucchesi14, L. Luciano15, M. Maria Cristina16, G. Rege-Cambrin17,
M. Santoro18, B. Scappini19, A. R. Scortechini20, P. Sportoletti21, E. Trabacchi22,
F. Castagnetti23
1Department of Translational and precision medicine-Az, Policlinico Umberto I-Sapienza
University, Rome; 2Hematology and Transplantation Unit, University of Bari, Bari;
3Azienda Ospedaliera “Bianchi Melacrino Morelli”, Reggio Calabria; 4Hematology, Sant
Eugenio Hospital, Rome; 5Division of Hematology, AORN Cardarelli, Naples; 6Hematology
Unit, University of Padova, Padova; 7Department of Internal Medicine, Pordenone General
Hospital, Pordenone; 8Clinical and Biological Sciences, University of Turin; 9Department
of Molecular Biotechnology and Health Sciences, University of Turin, Struttura Semplice
Dipartimentale Allogeneic Stem Cell Transplant, Azienda Ospedaliero-Universitaria
Città della Salute e della Scienza di Torino; 10Division of Haematology, Città della
Salute e della Scienza, Turin; 11Novartis Oncology, Medical Department, Novartis Farma
SpA, Origgio; 12Department of Cellular Therapies and Transfusion Medicine, AOU Careggi,
Florence; 13Department Hematology-Oncology, Azienda Ospedaliera Pugliese-Ciaccio,
Catanzaro; 14Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST)
“Dino Amadori”, Meldola; 15Hematology Unit, AUOP Federico II, Naples; 16Hematology
Department, San Bortolo Hospital, Vicenza; 17Medicina Interna e Ematologia, Ospedale
San Luigi, Università di Torino, Orbassano; 18Hematology, University Hospital Paolo
Giaccone, Palermo; 19Hematology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence;
20Division of Hematology, Azienda Ospedaliero Universitaria Ospedali Riuniti Ancona,
Ancona; 21Institute of Hematology, Centro Ricerche Emato-Oncologiche, Ospedale S.
Maria della Misericordia, University of Perugia, Perugia; 22Hematology Unit and BMT
Center, Ospedale G. Saliceto, Piacenza; 23Dipartimento di Medicina Specialistica,
Diagnostica e Sperimentale, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna,
Italy
Background: Chronic myeloid leukemia (CML) patients who have experienced failure and/or
intolerance to multiple lines of treatment have limited therapeutic possibilities.
Novel treatment options are needed to achieve sustained responses and increased tolerability
in this subset of patients. Asciminib, unlike all approved tyrosine kinase inhibitors
(TKIs) that bind to the ATP site of the BCR-ABL1 oncoprotein, is an allosteric, first-in-class
STAMP (Specifically Targeting the ABL Myristoyl Pocket) inhibitor with a new mechanism
of action that has shown efficacy in heavily pre-treated patients after two TKIs or
more.
Aims: We share our experience on asciminib use in CML patients outside of clinical
trials.
Methods: We describe retrospective data from 34 chronic phase CML patients in their
3rd or later line, treated with asciminib between April 2019 and October 2021 in 22
Italian institutions. The drug was granted by Novartis under a Managed Access Program
(MAP). BCR::ABL1/ABL ratio was expressed as % IS in all centers. Efficacy was analyzed
by comparing the response registered at the latest time point (34 pts evaluable) or
at 3 months (27 pts evaluable – we excluded those patients for whom molecular analysis
was not assessed) vs the response at baseline. Patient recruitment, dosing regimen
and safety were recorded according to the MAP recommendations. The safety information
was extracted from Argus database, a Novartis database where all adverse events related
to Novartis compounds are stored.
Results: Median time of asciminib treatment was 8.3 months (3.3–33.2) for the whole
cohort. Patients’ characteristics are displayed on Fig 1. Twenty-five patients (73.5%)
were pretreated with at least 3 or more TKIs and 50% have reported to have ≥3 comorbidities.
Approximately 59% of patients received prior ponatinib. Switch to asciminib occurred
for resistance in 21 pts (61.8%) and intolerance in 10 pts (29.4%). Ten patients (29.4%)
harbored mutations in ABL kinase domain, and 5 (14.7%) have a T315I mutation. All
the T315I patients had a previous ponatinib exposure.
Nineteen out of 27 (70.4%), 12/27 (44.4%) and 6/27 (22.2%) achieved or maintained
respectively Molecular Response of 2 log reduction (MR2), Major Molecular Response
(MMR) and Deep Molecular Response (DMR) at three months’ time point. Twenty-four out
of 34 (70.5%), 14/34 (41.7%) and 8/34 (23.5%) achieved or maintained respectively
MR2, MMR, DMR response at the last follow up. After 3 months and at the last follow
up, 18/27 (66.7%) and 20/34 (58.8%) of patients, respectively, showed an improvement
of previous baseline response. Approximately 37% and 35%, without MMR response at
baseline, reached at least an MMR at three months and last follow up, respectively.
We also observed that ponatinib treated patients showed a reduced probability of reaching
MR2, MMR and DMR responses compared to ponatinib naive patients at both 3 months and
last follow up time points. Seventy-five percent of patients with a T315I mutation
showed a response improvement, respect to baseline, already after 3 months on asciminib.
The safety profile of asciminib, based on the information spontaneously reported by
physicians, remained consistent with that already reported, with no new safety findings.
Image:
Summary/Conclusion: Asciminib has shown a promising efficacy profile and tolerability
in a setting of CML patients resistant and/or intolerant to 2 or more TKIs and with
a high comorbidity burden. Our results confirmed what observed in sponsored trials
and in the other world-wide programs, strongly suggesting a possible role for this
new agent in the future therapeutic scenario.
P713: REAL-WORLD MANAGEMENT OF PATIENTS WITH CHRONIC MYELOID LEUKAEMIA (CML): FIRST
ANALYSIS FROM THE UK NATIONAL REGISTRY FOR CML
G. Nesr1,*, R. Szydlo1, B. Braithwaite2, S. Frackelton2, P. Mathew2, M. Hashmi2, F.
Fernando1, A. Innes3, S. Claudiani1, A. Khan1, A. Rao4, L. Foroni1, D. Milojkovic3,
J. F. Apperley1, R. E. Clark5
1Centre for Haematology, Imperial College London, London; 2The Royal Liverpool University
Hospitals, Liverpool; 3Department of Clinical Haematology, Imperial College Healthcare
NHS Trust; 4Department of Clinical Haematology, Great Ormond Street Hospital for Children,
London; 5Department of Molecular & Clinical Cancer Medicine, University of Liverpool,
Liverpool, United Kingdom
Background: Analysing data emerging from population based cancer registries (PBCR)
informs current practice. The UK National Registry for chronic myeloid leukaemia (CML)
patients (ptn) is a web-based PBCR, initially established as a regional registry in
2010, then expanded nationally in 2015.
Aims: To understand real world management of UK CML ptn.
Methods: The registry utilizes an online interface to capture baseline and yearly
follow up data from contributing centres (CC). Data are securely transferred using
the N3 backbone of the NHS network. Ptn are consented for enrolment at their respective
centres. A “treatment episode” is defined as the duration during which a ptn received
a specific tyrosine kinase inhibitor (TKI). The 2013 version of the ELN guidelines
was used to define treatment milestones. For each milestone timepoint, treatment switches
were counted if they occurred before the next milestone.
Results: Demographic and baseline disease characteristics data were available for
748 ptn from 43 CC (Figure 1). Of those, 79%, 75% and 94% had available karyotypes,
FISH for BCR-ABL1, and RT-PCR for BCR-ABL1, respectively, at diagnosis and/or follow
up. Follow up and treatment data were available for 570 and 534 ptn, respectively.
Median follow up duration was 4.7 years (range: 0.1-23.7). Median age at diagnosis
was 53.3 years (range: 14.6-95.1) and 53% were males. Karyotype and RT- PCR for BCR
ABL1 at diagnosis were available for >75% of ptn in 40% of CC. Molecular monitoring
data were available for 540 ptn: 342 (63%) had a median testing frequency of ≤ 3 months,
167 (31%) between 3-6 months, 20 (4%) between 6-12 months, and 11 (2%) of > 12 months.
Imatinib (IM), nilotinib (NIL), dasatinib (DAS), bosutinib and ponatinib were prescribed
as 1st or subsequent line therapy in 46%, 23%, 21%, 14%, and 7% of the treatment episodes,
respectively. IM remained the most frequent 1st-line treatment after NICE approval
of 2GTKI in newly diagnosed ptn. The choice of 1st line TKI (IM vs 2G) was not influenced
by the Sokal, Euro, EUTOS or ELTS scores. Of ptn started on 1st line IM, NIL, and
DAS; 94%, 63% and 100% received the licensed doses (400 mg OD, 300 mg BD, and 100 mg
OD), respectively. Optimal responses at 3,6 and 12 months were achieved in 73%, 63%,
and 51% of pts. Failure at 3 months was not evaluable as the registry did not capture
sequential FBCs. Of those with a suboptimal response at 3 months, 15% changed treatment.
At 6 months 28% and 50% of ptn with suboptimal and failure responses, respectively,
switched their TKI. At 12 months, 10% and 28% of ptn in the suboptimal and failure
categories changed TKI. Between 1-6 treatment switches, for failure and/or intolerance,
occurred in 46% of ptn. Treatment switches on IM were more common for resistance than
intolerance and vice versa for 2G and 3G TKI. Of all ptn who switched TKI, 40% underwent
a further TKI switch. Kinase domain mutation analysis was requested in 36% and 33%
of ptn resistant to their TKI at 6 and 12 months, respectively. Five-year overall
survival (OS) probability was 93% for CP-CML ptn. Ptn achieving CCyR had a significantly
higher OS probability than those who did not (p<0.001 by log rank test). However,
differences in OS probability were not statistically significant for ptn attaining
deeper responses (p<0.5 for MR3, p<0.076 for MR4, p<0.141 for MR4.5, and p<0.084 for
MR5).
Image:
Summary/Conclusion: Our results highlight the lack of conformance to the current guidelines
in various aspects of CML management, the excellent OS survival achievable with currently
available TKIs and the importance of achieving CCyR for better OS.
P714: EFFECT OF TYROSINE KINASE INHIBITORS ON MALE FERTILITY IN PATIENTS WITH CHRONIC
PHASE CHRONIC MYELOID LEUKAEMIA
G. Nesr1,*, S. Claudiani1, D. Milojkovic2, I. Caballes2, S. Lovato1, J. Channa3, J.
F. Apperley1
1Centre for Haematology, Imperial College London; 2Department of Clinical Haematology,
Imperial College Healthcare NHS Trust; 3Section of Investigation Medicine, Imperial
College London Faculty of Medicine, London, United Kingdom
Background: With the current availability of 3+ generations of tyrosine kinase inhibitors
(TKIs) and the resultant survival benefit, chronic phase chronic myeloid leukaemia
(CP-CML) is considered a chronic disease for most patients. Consequently, haematologists
are faced more with questions regarding the effects of treatment on fertility and
pregnancy outcomes. Whilst the teratogenic effect of TKIs is well established, data
on their effect on fertility, and especially male fertility, are scarce.
Semen analysis parameters (sperm number, motility and morphology) remain the cornerstone
in assessing male fertility, despite its limitations in comprehensively assessing
sperm functions.
Aims: To compare semen analysis parameters before and after TKI therapy in CP-CML
patients treated in the haematology department of Imperial College Healthcare NHS
Trust (ICHNT).
Methods: In collaboration with the Andrology Department, newly diagnosed men with
CP-CML are offered sperm banking before starting cytoreductive or TKI therapy, and
sperm analyses are routinely undertaken.
Patients treated with TKIs who had sperm banking at diagnosis were invited for repeat
semen analysis.
Treatment history and semen analysis parameters were collected from patients’ paper/electronic
medical records.
Median and range were used to summarize continuous non-parametric data. Paired sample
t-test was used to compare continuous semen analysis parameters before and after TKI
therapy. Double-sided p value of < 0.05 was considered as statistically significant.
SPSS version 25 software was used for statistical analysis.
Results: Twenty-five patients were identified to have a semen analysis performed around
the time of starting TKI therapy and were approached for repeat testing. Of those,
13 refused to participate, 1 was an international patient no longer residing in the
UK and 1 stopped TKI therapy for treatment free remission trial. Therefore, 10 patients
were finally included.
Median age at diagnosis was 35.6 years (range: 22.9-45.4) and patients were diagnosed
between 2001 to 2014.
Six and 2 patients received hydroxyurea and interferon α, respectively, before TKI
therapy. Six patients received 1 TKI (imatinib (IM), n = 4; nilotinib (NIL), n = 1;
bosutinib (BOS), n = 1), 2 received 2 TKIs (IM followed by NIL), and 2 received 3
TKIs (IM, DAS, NIL and IM, NIL, DAS). Switches were for failure and/or intolerance.
Median duration of TKI therapy at the time of repeat semen analysis was 5.9 years
(range: 3.3-16.5). Patients 6 and 10 had the initial semen analysis after 736 and
475 days, respectively, from commencing TKI therapy and all remaining patients had
it before starting. All patients had a deep molecular response at the time of the
repeat semen analysis.
The differences between sperm concentration, % progressive motility and % total motility
were all non-statistically significant (p = 0.457, 0.535, and 0.574, respectively).
In 2 patients the semen parameters were below the reference ranges in both occasions.
Morphology (% normal forms) was less than the reference 4% in the repeat samples of
5 patients (Table 1).
Image:
Summary/Conclusion: In our cohort, no significant differences were found in sperm
concentration or motility before and after treatment with different TKIs for a prolonged
period. The relevance of the finding of a low % of normal forms, in the presence of
normal sperm concentration and motility, in some of the patients in the semen sample
after TKI therapy is not clear. Studies with larger sample size would be needed to
confirm or refute our findings.
P715: MULTICENTER OBSERVATIONAL STUDY OF PONATINIB IN CML PATIENTS IN ARGENTINA. REAL-WORLD
EXPERIENCE.
M. J. Mela Osorio1, M. V. Osycka2, B. Moiraghi2, M. A. Pavlovsky1, A. I. Varela2,
I. Fernandez1, F. Sackmann Massa1, L. Ferrari1, I. Giere1, C. Sighel1, M. Riddick3,
C. Pavlovsky1,*
1HEMATOLOGY, FUNDALEU; 2HEMATOLOGY, Hospital General de Agudos Jose Maria Ramos Mejia;
3HEMATOLOGY, Instituto de Investigaciones Fisicoquímica Teóricas y Aplicadas (INIFTA,
Facultad de Ciencias Exactas, Universidad Nacional de La Plata- CONICET). Departamento
de Matemática, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Buenos
Aires, Argentina
Background: Ponatinib is a 3rd generation tyrosine kinase inhibitor (TKI) indicated
for adults with resistant or intolerant chronic phase (CP), accelerated phase (AP)
or blast phase (BP) chronic myeloid leukemia (CML) as well as in patients carrying
the T315I mutation. There is a lack of data on its use in the real-world setting.
Aims: We aimed to evaluate treatment patterns, outcomes and cardiovascular toxicity
in CML patients treated with ponatinib in Argentina.
Methods: This retrospective study included all patients aged ≥ 18 years with CP, AP
and BP CML who initiated ponatinib in routine clinical practice across 2 medical centers
in Argentina from 2013 to 2021. Demographic, efficacy and safety data were collected
from patient medical charts. Continuous variables were dichotomized. Association was
evaluated through Fisher exact test.
Results: A total of 56 patients were analyzed, 53 in CP, 2 in AP and 1 in BP. One
patient (1.8%) received ponatinib in first-line, 19 (34%) in second-line (2L), 35
(62.4%) in 3L, and 1 (1.8%) in ≥4L. Prior cardiovascular (CV) risk factors were recorded:
hypertension 8/56 14%, body mass index ≥25kg/m2 32/56 57%, hyperlipidemia (>220mg/dl)
13/56 23.2%, smokers 10/56 18% and diabetes mellitus 4/56 7%.
Median age at ponatinib start was 43.5 years (range, 18–73). Of 38 evaluated patients,
23 (60.5%) had a confirmed ABL1 mutation, including 19 (50%) with the T315I mutation.
Starting doses of ponatinib were 45 mg (34%), 30 mg (53.5%), or 15 mg (12.5%). The
median follow-up was 130 months (range, 13-324) and median treatment duration was
38 months (range, 1–105). At baseline, 51 patients (91%) with CP CML had less than
CCyR and 2 (3.5%) were in CCyR. For 3 patients (5.3%), assessment was not available.
At 3 months, 15/51 (29%) evaluable patients with CP CML were in CCyR; at 6 months,
17/44 (38.6%) evaluable patients with CP CML were in CCyR and at 12 months, 19/34
(56%) evaluable patients with CP CML were in CCyR.
Additionally, 21 (37.5%) and 9 (16%) patients with CP CML achieved a major molecular
response (MMR) or a deep molecular response (MR4.0–MR5.0) at least once during follow-up,
respectively.
Progression-free survival rates estimated for patients with CP CML at 12 and 24 months
were 90.3% (95% CI, 82.6–98.8%) and 86.2% (95% CI, 77.3–96.3%), respectively. Corresponding
overall survival rates were 100%, and 96.1% (95% CI, 90.9–100%).
Serious arterial thrombotic adverse events (AT-AEs) occurred in 8 patients (14%),
60% of these were coronary artery disease, 30% cerebrovascular and 10% peripheral
arterial disease. AT-AEs events occurred after a median time on ponatinib of 5 months
(range, 2-48), the majority of the events were severe but resolved, however two were
fatal.
Patients who developed an AT-AE after starting ponatinib were older than 60 years
(p=0.0174), and had higher cholesterol levels (p=0.03979). No associations were found
regarding obesity (p=0.403), previous TKI lines (p>0.5), smoking history (p>0.5) or
ponatinib starting dose ≤30mg vs >30mg (p=0.4248).
Summary/Conclusion: This is the first analysis of ponatinib treated patients in real-world
in Argentina and demonstrates that ponatinib has a favorable efficacy and safety profile
in patients with CML treated in standard clinical practice. Cardiovascular toxicity
of ponatinib in routine clinical practice is increased. Older (>60y) and dyslipemic
patients were at higher-risk of occlusive events. Prompting aggressive control of
CV risk factors as well as reducing doses at optimal time-points may help optimize
the use of ponatinib during daily practice.
P716: OUTCOMES AND PATTERNS OF TREATMENT IN CHRONIC MYELOID LEUKEMIA, A GLOBAL PERSPECTIVE
BASED ON A REAL-WORLD DATA GLOBAL NETWORK
A. Sanz1,*, R. Ayala1, G. Hernández2, N. López1, D. Gil1, R. Gil1, R. Colmenares1,
G. Carreño-Tarragona1, J. M. Sánchez Pina1, R. A. Alonso1, N. García Barrio3, D. Pérez-Rey2,
L. Meloni4, M. Calbacho1, M. Pedrera-Jiménez3, P. Serrano-Balazote3, J. de la Cruz5,
J. Martínez-López1
1Hematology, Hospital 12 de Octubre; 2Biomedical Informatics Group, Universidad Politécnica
de Madrid; 3Data Science Group, Research Institute imas12, Madrid, Spain; 4Trinetx,
LLC, Cambridge, United States of America; 5Research Institute imas12, Hospital 12
de Octubre, Madrid, Spain
Background: The natural history of chronic myeloid leukemia (CML) experienced a major
change in the early 2000s with the development of the first BCR-ABL1 specific tyrosine
kinase inhibitors (TKIs); however, there is limited supporting evidence from electronic
health records (i.e., real-world data [RWD]). TriNetx is a global health research
platform that provides researchers access to electronic medical records from healthcare
organizations (HCOs) for conducting research studies. It allowed us to analyze trends
in survival in CML patients throughout the years from a global perspective, as well
as examine relevant comorbidities and long-term toxicities.
Aims: The objective of this study is to better define the current survival landscape
of CML and the long-term toxicities, so that we can have a better and updated understanding
of the disease and the actual clinical practice in local, regional and global settings.
Methods: We analyzed data from more than 7500 patients diagnosed with CML from four
research networks (Hospital 12 de Octubre (H12O), the EMEA network, the US network,
the Global Network) covering more than 16 million subjects. Propensity score matching
for age and gender was applied, comparing samples equal in size (CML and controls).
This study was conducted with anonymized data accessed via the TriNetX platform, which
provided diagnoses, procedures, medications, laboratory values, and genomic information
from approximately 6,000 CML patients treated with TKIs from 57 health care organizations.
Results: Survival at 20 years was not significantly different between patients with
CML treated with TKIs and non-cancer patients (63·4% vs 69·05%, HR 1.095 (0·924 -
1·298) p = 0·295, Figure 1). No significant change in survival probability was seen
comparing patients treated 2001-2010 and 2011-2020 (HR 1·113 (0·858 - 1·443), p =
0.42) or comparing imatinib with second-generation TKIs (imatinib vs dasatinib HR
1·017 (0·895 - 1·157), log-rank p = 0·80; imatinib vs nilotinib HR 1·101 (0·925 -
1·310).
When comparing the occurrence of second malignancies, an increased incidence was seen
in CML patients (64·4% vs 17·2%, OR 8·72 (7·955, 9·549)). In the case of cardiovascular
diseases, CML patients again showed a higher incidence than non-CML patients (74·2%
vs 60·5%, p<0·001, OR 1·879 (1·729, 2·041)). No specific TKI was associated with the
increased risk of any of these conditions.
Regarding TKI usage and switching patterns, dasatinib was the most common second-line
treatment for both H12O and the US, but not in the EMEA where it was nilotonib. Ponatinib
assumed a larger role in third-line and beyond.
Image:
Summary/Conclusion: From this large real-world analysis, we conclude that in the TKI
era, the CML population has the same survival as a non-cancer population. However,
the main TKI toxicities and comorbidities remained high in the CML population, and
despite them not seeming to impact survival, it could affect the quality of life.
Therefore, strategies to stop lifelong TKI use should be prioritized to reduce adverse
events.
P717: BOSUTINIB IN NEWLY DIAGNOSED CHRONIC MYELOID LEUKEMIA: GASTROINTESTINAL, LIVER,
EFFUSION AND RENAL SAFETY CHARACTERIZATION IN THE BFORE TRIAL
J. E. Cortes1,*, D. Milojkovic2, C. Gambacorti-Passerini3, V. García-Gutiérrez4, M.
J. Mauro5, E. Leip6, S. Purcell7, A. Viqueira8, T. H. Brümmendorf9
1Georgia Cancer Center, Augusta, United States of America; 2Hammersmith Hospital,
London, United Kingdom; 3University of Milano-Bicocca, Monza, Italy; 4Universitario
Ramón y Cajal, Ramón y Cajal Health Research Institute, Madrid, Spain; 5Memorial Sloan
Kettering Cancer Center, New York; 6Pfizer Inc, Cambridge, United States of America;
7Pfizer Ltd, London, United Kingdom; 8Pfizer SLU, Madrid, Spain; 9Universitätsklinikum
RWTH Aachen, Aachen, Germany
Background: Bosutinib is approved for the treatment of Philadelphia chromosome–positive
(Ph+) chronic myeloid leukemia (CML) resistant/intolerant to prior therapy and newly
diagnosed Ph+ chronic phase (CP) CML. Efficacy and safety of bosutinib vs imatinib
in patients with newly diagnosed CP CML was assessed in the phase 3 BFORE trial (NCT02130557).
Aims: The safety profile of bosutinib after 5 years follow-up, with a focus on gastrointestinal,
liver, effusion and renal treatment-emergent adverse events (TEAEs), was characterized.
Methods: Patients who received ≥1 dose of bosutinib (n=268) or imatinib (n=265) 400 mg/day
in BFORE were included. Adverse events (AEs) of special interest were analyzed by
selecting prespecified MedDRA terms to generate TEAE clusters. This analysis is based
on the final database lock: June 12, 2020.
Results: Median duration of treatment was 55 months for patients receiving bosutinib
or imatinib; respective median (range) dose intensity was 393.6 (39–583) vs 400.0
(189–765) mg/d. Any grade TEAEs occurred in 98.9% and 98.9% of bosutinib- vs imatinib-treated
patients. The most common newly occurring TEAEs (any grade) after 12 months were increased
lipase (9.0%) with bosutinib, and diarrhea (8.3%) with imatinib. In bosutinib- vs
imatinib-treated patients, 25.4% vs 14.3% had AEs leading to permanent treatment discontinuation;
the majority discontinued in year 1 (14.2% vs 10.6%). The most frequent AEs leading
to discontinuation were increased ALT (overall, 4.9%; year 1, 4.5%) with bosutinib
vs thrombocytopenia (overall, 1.5%; year 1, 1.5%) with imatinib.
Gastrointestinal, liver, effusion and renal TEAEs, respectively, occurred in 79.9%,
44.0%, 6.0% and 10.4% (maximum grade 3/4 [G3/4]: 9.0%, 26.9%, 1.1% and 2.2%) of bosutinib-
vs 61.5%, 15.5%, 2.3% and 9.8% (G3/4: 1.1%, 4.2%, 0.4% and 0.8%) imatinib-treated
patients. One grade 5 renal TEAE occurred in the bosutinib arm and was not considered
related to treatment. Cumulative rates per treatment year are shown in the Table.
The most common gastrointestinal TEAEs were diarrhea (bosutinib vs imatinib: 75.0%
vs 40.4% [G3/4: 9.0% vs 1.1%]) with bosutinib, and nausea (37.3% vs 42.3% [G3/4: 0%
vs 0%]) with imatinib. In both arms, the most common liver, effusion and renal TEAEs,
respectively, were increased ALT and/or AST (34.0% vs 8.3% [G3/4: 22.0% vs 2.3%]),
pleural effusion (5.2% vs 1.9% [G3/4: 0.7% vs 0.4%]) and increased blood creatinine
(6.7% vs 8.3% [G3/4: 0.4% vs 0.4%]). Gastrointestinal, liver, effusion and renal TEAEs
infrequently led to treatment discontinuation (1.9%, 7.8%, 0.7% and 0.7% vs 1.1%,
0.8%, 0% and 0.4%).
Image:
Summary/Conclusion: The safety profiles of bosutinib and imatinib in BFORE were distinct,
with no new safety signals identified after 5 years of follow-up. The onset of TEAEs
occurred primarily during year 1 (e.g., gastrointestinal and liver), with an increased
incidence of some TEAEs (e.g., effusion and renal) in later years. Discontinuations
due to AEs generally occurred early into treatment, with low rates of discontinuation
due to gastrointestinal, liver, effusion and renal AEs. These long-term safety results
further support the use of first-line bosutinib as a standard of care in patients
with CP CML.
P718: ASCIMINIB MANAGED-ACCESS PROGRAM (MAP) IN RUSSIA
O. Shukhov1,*, A. Turkina1, E. Lomaia2, E. Morozova3, A. Petrova1, T. Chitanava2,
Y. Vlasova3, E. Chelysheva1, E. Kuzmina1, I. Nemchenko1, A. Bykova1, M. Gurianova1,
A. Kohno1, E. Parovichnikova1
1National Medical Research Center for Hematology, Moscow; 2Almazov National Medical
Research Centre; 3Raisa Gorbacheva Memorial Research Institute of Children’s Oncology,
Saint-Petersburg, Russia
Background: Asciminib is aa ABL kinase inhibitor that works by Specifically Targeting
the ABL Myristoyl Pocket (STAMP) that has shown efficacy and a good safety profile
in phase I and III studies in patients with Ph-positive leukemia failing prior TKIs.
In Russia, asciminib is available under the Novartis approved Managed Access Program
(MAP).
Aims: to present interim results of the use of aciminib in clinical practice under
the MAP program in Russia.
Methods: From September 2019 to January 2022, 68 patients (pts) with CML were enrolled
in the MAP program from three Russian clinics. We analyzed therapy results from 50
pts. Eleven pts who received asciminib for less than 3 months and 7 pts who underwent
bone marrow transplantation were excluded from the analysis. Pts recruitment, dosing
regimen, response monitoring, and toxicity control were performed according to the
MAP treatment plan. Complete cytogenetic response (CCyR), major molecular response
(MMR) and deep molecular response (MR4) were assessed by cumulative incident function
(CIF) with Gray’s test for comparison responses in subgroups. Multivariate analysis
was performed to find independent factors for MMR. Differences were considered significant
if the p ≤ 0.05
Results: Baseline characteristics: female 66%; Ме age 53 years (range 26-81); median
duration of CML before asciminib 8 years (range 1-24); 42 pts were in chronic phase
(CP) CML, 7 and 1 pts had a history of accelerated phase (AP) and myeloid blast crisis
(MBC), respectively. Twenty nine (58%) pts had BCR::ABL1 mutations, 20 pts (40%) had
BCR::ABL1
T315I, 8 (16%) pts had at least two mutations. Eight (16%) pts had additional chromosomal
abnormalities (ACAs). Among the thirty three (66%) pts who received ≥4 TKIs, 20 (40%)
had a history of ponatinib treatment. Me follow-up period was 11 months (range 4-30),
7(14%) pts discontinued asciminib (5 due to resistance, 2 due to progression). One-year
survival rate without treatment discontinuation was 92%. In 30 (60%) pts, The initial
dose was 40 BID in 30 (60%) pts and 200 mg BID in 20 (40%) pts
The 2-year overall survival was 96%, two (4%) pts progressed to advanced phase and
died. CIF of CCyR, MMR and MR4 at 12 month was 37.5% (CI:24-58), 32% (CI:20-52) and
14% (CI:7-31), respectively. Univariate analysis was performed for the following factors:
history of advanced phases, initial dose of asciminib, presence of BCR::ABL1 mutations
and ACAs, best molecular response on previous TKIs, molecular response at the time
of asciminib starting, history of ponatinib treatment, number of prior TKIs and duration
of TKI therapy before asciminib. Best molecular response to previous TKIs, molecular
response at the time of asciminib starting, number of TKIs prior to asciminib, and
history of previous ponatinib treatment were identified as predictors of MMR at 12
months (Table 1). Best molecular response on previous TKIs (≤1% vs 1-10% vs ≥10%)
was found to be independently significant factor due to multivariate analysis (p=0,0072,
hazard ratio 7,6 (1,7-33))
Twenty two (44%) patients experienced adverse events (AEs) of any grade and 8 (16%)
had AEs of grade 3-4 (Table 2), reported during treatment, regardless of the connection
with the drug. No one stopped treatment because of toxicity. No differences in toxicity
were found between doses of 80 and 400 mg/day.
Image:
Summary/Conclusion: Asciminib shows promising results for the treatment of highly
pre-treated CML patients and should be considered as an therapeutic options for pts
with failure to previous TKIs
P719: EFFICACY AND SAFETY OF BOSUTINIB IN LATER-LINE PATIENTS WITH CHRONIC MYELOID
LEUKEMIA: A SUB-ANALYSIS FROM THE PHASE 4 BYOND TRIAL
C. Gambacorti-Passerini1,*, T. H. Brümmendorf2, T. Ernst3, E. Leip4, S. Purcell5,
A. Viqueira6, F. J. Giles7, G. Rosti8, A. Hochhaus3
1University of Milano-Bicocca, Monza, Italy; 2Universitätsklinikum RWTH Aachen, Aachen;
3Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany; 4Pfizer Inc,
Cambridge, United States of America; 5Pfizer Ltd, London, United Kingdom; 6Pfizer
SLU, Madrid, Spain; 7Developmental Therapeutics Consortium, Chicago, United States
of America; 8IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”,
Meldola, Italy
Background: Bosutinib is approved for patients with Philadelphia chromosome-positive
chronic myeloid leukemia (CML) resistant or intolerant to prior therapy and newly
diagnosed patients in chronic phase. The efficacy and safety of bosutinib in patients
with CML resistant or intolerant to prior tyrosine kinase inhibitors (TKIs) was evaluated
in the phase 4 BYOND trial (NCT02228382; Gambacorti-Passerini et al, Blood, 2021).
Aims: The efficacy and safety of bosutinib in resistant and intolerant patients with
CML treated with 2 (3L) and 3 (4L) prior TKIs was characterized.
Methods: The BYOND trial included 163 patients with CML resistant or intolerant to
prior TKIs. A sub-analysis of 48 patients treated with 2 (3L) and 3 (4L) prior TKIs,
categorized by resistance or intolerance to the last received TKI, was performed.
This sub-analysis is based on the final November 23, 2020 database lock.
Results: There were 18 and 30 patients resistant or intolerant to the last received
TKI who entered the study without complete cytogenetic response (CCyR) or major molecular
response (MMR), respectively. Median (range) treatment duration was 10.6 months (1.6–48.5)
vs 28.3 months (0.2–48.6) and median (range) dose intensity was 447.1 mg/d (131.3–520.4)
vs 288.8 mg/d (79.7–500.0) for resistant vs intolerant patients. Prior TKIs included
imatinib (88.9% vs 100.0%), dasatinib (88.9% vs 83.3%), and nilotinib (66.7% vs 63.3%)
for resistant vs intolerant patients. Overall, 61.1% vs 66.7% of resistant vs intolerant
patients discontinued bosutinib, mostly commonly due to adverse events (AEs) in 27.8%
vs 16.7% patients; 16.7% vs 6.7% discontinued bosutinib due to insufficient clinical
response. Rates of CCyR and MMR are shown in the Table. Among responders (resistant
vs intolerant patients), median (range) time to CCyR was 5.1 months (2.8–8.8) vs 3.0
months (2.7–6.1); median (range) time to MMR was 5.8 months (2.8–9.4) vs 3.2 months
(2.8–9.3). In resistant vs intolerant patients, any grade treatment-emergent AEs (TEAEs)
were reported by 100.0% vs 96.7% patients; grade 3/4 TEAEs were reported by 72.2%
vs 83.3% patients. Grade 3/4 TEAEs >10% in resistant patients were thrombocytopenia
(22.2%) and neutropenia (11.1%), and in intolerant patients were increased alanine
aminotransferase (26.7%), diarrhea (23.3%), pleural effusion (13.3%), and rash (13.3%).
Image:
Summary/Conclusion: This sub-analysis of resistant and intolerant patients without
baseline CCyR or MMR shows bosutinib was active in heavily pretreated patients with
resistance or intolerance to the last received TKI. Despite a difference between resistant
and intolerant patients with CML, efficacy outcomes, though lower than the overall
BYOND population, are encouraging, and safety was generally consistent with previous
reports.
P720: PROACTIVE DASATINIB DOSE REDUCTION IN THE ALLG CML 12 DIRECT STUDY BASED ON
TROUGH LEVELS MINIMISE TOXICITY AND PRESERVE EFFICACY
D. Yeung1,2,*, A. Grigg3, N. Shanmuganathan1, A. Soltenbeck4, D. White2, S. Branford5,
N. Viiala6, P. Rowlings7, A. Mills8, J. Shortt9, C. Tiley10, D. Ross11, D. Kipp12,
R. Harrup13, I. Cunningham14, J. Kwan15, R. Eek16, H. Mutsando17, K.-S. Tan18, K.
Burbury19, M. Wright20, T. Hughes1,2, On Behalf of the ALLG21
1Haematology, Royal Adelaide Hospital; 2Precision Medicine, SAHMRI, Adelaide; 3Haematology,
Austin Hospital; 4Statistical Revelations, Melbourne; 5Molecular Pathology, SA Pathology,
Adelaide; 6Haematology, Liverpool Hospital, Sydney; 7Haematology, Calvary Mater Hospital,
Newcastle; 8Haematology, Princess Alexandra Hospital, Brisbane; 9Haematology, Monash
Medical Centre, Melbourne; 10Haematology, Gosford Hospital, Gosford; 11Haematology,
Flinders Medical Centre, Adelaide; 12Haematology, Barwon Health, Geelong; 13Haematology,
Royal Hobart Hospital, Hobart; 14Haematology, Concord Hospital; 15Haematology, Westmead
Hospital, Sydney; 16Haematology, Border Medical Oncology, Albury; 17Haematology, Toowoomba
Hospital, Toowoomba; 18Nephrology, Princess Alexandra Hospital, Brisbane; 19Haematology,
Peter MaCallum Cancer Centre, Melbourne; 20Haematology, Finoa Stanley Hospital, Perth;
21ALLG, Melbourne, Australia
Background: Dasatinib treatment leads to excellent molecular responses in chronic
phase chronic myeloid leukemia (CP-CML) but at 100mg leads to a substantial risk of
pleural effusions, which may be higher in the elderly. Rousselot et al (BJH 2021)
suggested this risk may be mitigated through dose modifications as guided by trough
levels (Cmin).
Aims: The CML12 (DIRECT) study, run by the ALLG, is a single arm phase II study, aiming
to minimise dasatinib related toxicity whilst preserving efficacy, through dose adaptation
as guided by dasatinib trough levels. We report here the primary end point - cumulative
incidence of pleural effusion (PEf) at 24 months (mos) and key efficacy secondary
end points.
Methods: DIRECT enrolled newly diagnosed CP-CML pts. The first 34 patients (pts) were
aged >60 years (yrs), after which the protocol was amended to include pts aged >18
yrs at the recommendation of the trial management committee. All pts started on dasatinib
100mg QD, with Cmin taken at 7, 28, 56 & 90 days, then every 3 mos hence. Pts sequentially
dose reduced to 70mg, then to 50mg QD, for Cmin >3nM, within the first 2 mos, prior
to assessment of early molecular response at 3 mos. Doses <50mg QD were permitted
temporarily only for toxicity management. Chest x-ray and transthoracic echocardiograms
were performed at baseline & 24 mos.
Results: Eighty pts were enrolled from 14 centres, with a median follow up of 36 mos
(range 24-60). Median age was 64 yrs (range 21-86) and 46% were female. The ELTS score
was low and intermediate in 54% and 34% respectively, high in 5% and missing in 8%.
Cmin and treatment assigned at various timepoints are detailed in Table 1. Older patients
had higher Cmin, particularly prior to dose adjustments. The majority of pts were
dose reduced to 50mg QD. Fifteen cases of pleural effusion (PEf) occurred, 5 within
the first mo, and 12 in total by 24 mos; 3 were asymptomatically detected on chest
x-ray. The cumulative incidence of PEf by 24 mos was 15% overall (95% CI 8-25%, Fig
1); with values of 7.6%, 14.3% and 45.5% respectively in patients <60 yrs, 60-75 yrs
and ≥75 yrs. The Cmin prior to the PEf was >3nM in 8/15 pts.
At 24 mos, 59 (74%) of pts remained on dasatinib. Reasons for discontinuation were
intolerance / adverse events (AE) (n=15, 19%); treatment failure (n=4, 5%); death
(n=1, 1%) and consent withdrawn (n=1, 1%). AEs leading to discontinuation were PEf
(n=6, 7.5%) pulmonary hypertension (n=4, 5%; 2 reported as SAEs), pericardial effusion
(n=2, 2.5%) and cardiac failure (n=2, 2.5%). There were no cases of peripheral vascular
disease or strokes.
An early molecular response (BCR::ABL1 <10% IS at 3 mos) was achieved in 77 pts (96%).
Cumulative incidence of MMR (<0.1%) by 12 and 24 mos were 75% (95% CI 66-84%) and
92% (95% CI 86-97%), respectively; MR4.5 by 24 and 48 mos were 48% (95% CI 38-60%)
and 76% (95% CI 64-90%), respectively. No patient progressed to AP/BC on or off study.
Three pts have died of non CML-related causes (infection n=2; biliary carcinoma n=1).
Image:
Summary/Conclusion: High rates of MMR/MR4.5 were achieved, despite significant dose
reductions, according to target drug levels over the first 2 months. Toxicity profile
was also broadly favourable. However, such adaptive approaches had limited capacity
to influence early development of PEf. A strategy using lower starting doses, with
escalation for failure to achieve molecular targets, may further minimise toxicity,
especially in the elderly.
P721: PREDICTIVE SCORING SYSTEMS FOR MOLECULAR RESPONSES IN PERSONS WITH CHRONIC MYELOID
LEUKEMIA RECEIVING INITIAL IMATINIB THERAPY
X. Zhang1,*, Z. Li1, M. Zhang1, P. G. Robert2, X. Huang1, Q. Jiang1
1Peking University People’s Hospital, Beijing, China; 2Centre for Hematology Research,
Department of Immunology and Inflammation, Imperial College London, London, United
Kingdom
Background: Nowadays more patients have benefited from tyrosine kinase inhibitors
(TKIs) therapy to achieve a stable major molecular response (MMR) and even deep molecular
responses (DMR, at least MR4), which prevented disease progression and allows for
pursuing treatment-free remission (TFR). However, there were still lacking of robust
prediction models for molecular responses including MMR or DMR to TKI-therapy.
Aims: Develop and validate predictive scoring systems for MMR and MR4 in persons with
newly-diagnosed chronic phase chronic myeloid leukemia (CML) receiving imatinib.
Methods: Data from 1364 consecutive subjects were randomly-assigned 2:1 to training
(n=909) and validation (n=455) datasets. Co-variates significantly associated with
MMR and MR4 in multi-variable analyses in the training dataset were used to develop
a predictive scoring system based on standardized regression coefficients (beta [β])
and the model tested in the validation dataset. Time-dependent area under the receiver-operator
characteristic curves (AUROC), calibration plots and decision curve analyses (DCAs)
were used to evaluate predictive accuracy.
Results: In the training dataset, male sex (β=-0.350, HR=0.7 [95%CI: 0.6, 0.8]), WBC
≥ 130 ×10E+9/L (β=-0.538, HR=0.6 [0.5, 0.7]), hemoglobin < 110 g/L (β=-0.490, HR=0.6
[0.5, 0.8]) and EUTOS long-term survival (ELTS) risk score (intermediate-risk: β=-0.464,
HR=0.6 [0.5, 0.8]; high-risk: β=-0.743, HR =0.5 [0.3, 0.7]) were significantly-associated
with lower cumulative incidence of MMR. Each co-variate was assigned 1 point in the
predictive scoring system for MMR except WBC ≥ 130 ×10E+9/L and ELTS high-risk assigned
2 points. Using the predictive scoring system, subjects were classified into (1) low-
(score=0-1; n=429, 47%), (2) intermediate- (score=2-4; n=340, 38%); and (3) high-
(score=5-6; n=140, 15%) risk subgroups. 5-year cumulative incidences of MMR were 86%
(84, 89%), 61% (55, 65%) and 34% (27, 43%) (p for trend <0.001; Figure A). Comparable
5-year cumulative incidences in validation dataset were 86% (82, 90%), 69% (64, 74%)
and 31% (22, 40%) (p for trend<0.001; Figure B). AUROC values were 0.77 and 0.74 in
the training and validation datasets. Similar analyses for MR4 in training dataset
identified male sex (β=-0.282, HR=0.8 [0.6, 0.9]), WBC ≥ 120 ×10E+9/L (β=-0.747, HR=0.5
[0.4, 0.6]), hemoglobin<110 g/L (β=-0.496, HR=0.6 [0.5, 0.8]) and ELTS risk score
(intermediate-risk: β=-0.509, HR=0.6 [0.4, 0.8]; high-risk: β=-1.212, HR=0.3 [0.1,
0.6]) were significantly-associated with lower cumulative incidence of MR4. Male sex
was assigned 1 point; ELTS intermediate-risk and hemoglobin < 110 g/L, 2 points; WBC
≥ 120 ×10E+9/L, 3 points; and ELTS high-risk, 4 points in the predictive scoring system
for MR4. Subjects in the training dataset were classified into (1) low- (score=0-2;
n=416, 46%), (2) intermediate- (score=3-7; n=348, 38%); and (3) high- (score=8-10;
n=145, 16%) risk cohorts. 5-year cumulative incidences of MR4 were 71% (70, 73%),
35% (31, 39%) and 16% (10, 22%) (p for trend<0.001, Figure C). Similar 5-year cumulative
incidences in validation dataset were 66% (61, 71%), 45% (38, 52%) and 21% (12, 30%)
(p for trend<0.001, Figure D). AUROC values were 0.76 and 0.72. Calibration plots
and DCAs showed good performance for predicting MMR and MR4.
Image:
Summary/Conclusion: We developed the predictive scoring systems for MMR and MR4 in
persons with chronic phase CML receiving imatinib. These data may help physicians
make an appropriate therapy decision.
P722: IS THE SECOND-GENERATION TYROSINE KINASE INHIBITOR A BETTER THERAPY THAN IMATINIB
FOR PERSONS WITH CHRONIC MYELOID LEUKEMIA PRESENTING IN ACCELERATED-PHASE?
X. Zhang1,*, S. Yang1, Y. Liu1, Q. Jiang1
1Peking University People’s Hospital, Beijing, China
Background: Although most patients with chronic myeloid leukemia (CML) were presenting
in the chronic phase at diagnosis, approximately 5% presented with features of accelerated
phase. The features of accelerated phase were variably defined by different criteria,
such as MD Anderson and European LeukemiaNet (ELN) criteria most often used. Regardless
of these criteria used to defined accelerated phase, only scant data were available
regarding the prognostic co-variates for outcome. Moreover, evidence that patients
could benefit more from the second generation (2G-) tyrosine kinase inhibitor (TKI)
than imatinib as initial therapy was still extremely limited.
Aims: Explore whether patients with chronic myeloid leukemia (CML) presenting in accelerated-phase
could benefit from initial second-generation tyrosine kinase inhibitor (2G-TKI) therapy.
Methods: We interrogated data from consecutive patients with CML presenting in accelerated
phase at diagnosis by the MD Anderson criteria (n = 325) or ELN criteria (n = 275).
Propensity score matching (PSM) analysis was used to compare cytogenetic and molecular
responses and outcomes in those receiving initial imatinib versus a 2G-TKI. COX multi-variable
regression model was performed to identify the prognostic co-variates.
Results: In the cohort defined by MD Anderson criteria, PSM analysis showed that patients
receiving initial 2G-TKI (n = 89) had higher probability of achieving a CCyR (p =
0.01), MMR (p = 0.04) and MR4.5 (p = 0.004) than those receiving imatinib (n = 156),
however, probabilities of FFS, PFS and survival were similar. Multi-variable analyses
showed that lower platelets and higher blasts in blood or bone marrow were significantly-associated
with poor FFS, PFS and/or survival (Figure A). In an attempt to determine whether
choice of therapy contributed to the differences in responses and outcomes, platelets
< 600 × 10E+9/L and blood / bone marrow blasts ≥ 5% were identified as the prognostic
categorical co-variates by ROC analyses. The entire cohort were classified into low-
(possessing none of these co-variates, n = 57), intermediate (possessing any 1 of
these co-variates, n = 127) or high-risk (possessing all of these co-variates, n =
61) subgroups. There were significant differences in probabilities of outcomes among
these risk subgroups (all p values < 0.001, Figure B). In each risk subgroup, patients
receiving initial 2G-TKI had the similar outcomes to those receiving imatinib except
higher incidences of responses including CCyR, MMR and MR4.5. The same results were
observed in the cohort defined by ELN criteria.
Image:
Summary/Conclusion: Initial 2G-TKI lead to similar outcomes as imatinib in patients
with CML presenting in accelerated-phase except higher therapy responses.
P723: LOSS OF THE NK MOTIF AND ANKYRIN REPEAT DOMAIN 1 (KANK1) LEADS TO LYMPHOID COMPARTMENT
DYSREGUATION IN MICE
M. Almosailleakh1,*, S. Narcisi1, C. Roger Michel Côme1, H. Luche2, K. Grønbæk1,3
1Biotechnology Research and innovation Center, University of Copenhagen, Copenhagen,
Denmark; 2Centre d’Immunophénomique, Parc Scientifique et Technologique de Luminy,
Marseille, France; 3Department of Hematology, Regishospitalet, Copenhagen, Denmark
Background: We identified a novel germline loss of heterozygosity (LOH) mutation encompassing
the NK Motif And AnKyrin Repeat Domain 1 (KANK1) gene in a young Myelodysplastic syndrome
(MDS) patient and his healthy father, with no additional MDS-related mutations or
chromosomal abnormalities. Clinical investigation suggested development of MDS following
severe infection, likely triggered by an autoimmune mechanism. KANK1 protein plays
a role in the control of cytoskeleton formation by regulating actin polymerization.
A t(5;9) translocation resulting in a fusion of the platelet-derived growth factor
receptor beta gene (PDGFRB) and KANK1 was detected in a patient with myeloid neoplasm
(MN) characterized with severe thrombocythemia. The protein is thought to have a tumour
suppressor function, as its expression is downregulated or missing in several tumor
tissues, while over-expressing the protein was reported to inhibit the proliferation
of tumor cells in solid cancer models. In addition, defects in cytoskeletal proteins
are reported to contribute to immunological dysfunction and disruption of normal immune
processes.
Aims: To investigate the role of KANK1 in the regulation of normal hematopoiesis and
the development of myeloid disorders.
Methods: We generated a new genetically modified KANK1 (Kank1
-/-) knockout mouse model and analysed the phenotypic and developmental changes in
hematopoiesis in young and aged mice.
Results: We confirmed the loss of Kank1 mRNA and protein expression by q-PCR and targeted
protein mass-spectrometry respectively. The mice are viable and fertile, and reproduced
according to mendelian ratio. Over the two years observation period, Kank1
-/- mice did not exhibit any signs of disease, and their peripheral blood parameters
remained within the normal range. Flow cytometry analysis of bone marrow (BM) cells
from 17-20 weeks kank1
-/- old mice showed minor increase in CD3+ T-cell population, while the expression
of the double positive B220/CD19 B-cells population was reduced in Kank1
-/- compared to Kank1
+/+. Comprehensive flow cytometry analysis of spleen and thymus cells from young (n=8
each, 6-7 weeks old) and aged (n=8, 68-71 weeks old) Kank1
-/- mice showed further dysregulation of the lymphoid compartment when compared to
Kank1
+/+. In young mice, a slight decrease of Treg cells is noticed in the spleen, coupled
with a slight decrease of activated CD4 in the peripheral blood. In the spleen of
aged Kank1
-/- mice, the proportions of macrophages and Treg cells were decreased, while the
central memory CD4+ T-cells were increased. Total protein analysis from BM cells of
Kank1
-/- (n=3) compared to Kank1
+/+ mice (n=3) showed a significant reduction (FC >0.5, adj.p. <0.05) in proteins
associated with cytoskeleton formation such as Myosin1 (MYH1), Tropomysin-1 (TDM1)
and Annexin (ANXA2), and extracellular matrix components such as Collagena(CO1A1)
and Matrix metalloproteinase9 (MMP9). We also detected a significant increase in expression
of the DNA licensing factor minichromosome maintenance complex component 7 (MCM7),
and the mitochondrial membrane associated transporter ATP binding Cassette Subfamily
B Member 10 (ABCBA).
Summary/Conclusion: Dysregulation of the immune system has been widely recognized
to play a role in the development of MDS. Based on our results, we propose a role
for KANK1 in regulating the development of immune cells in the mouse through it invovlement
in cytoskeleton regulation. In vivo infectious challenge experiments using our model
will futher elucidate the importance of KANK1 in the development of MDS.
P724: THE BONE MARROW MESENCHYMAL CELL-DERIVED EXTRACELLULAR MATRIX IS SIGNIFICANTLY
ALTERED IN PATIENTS WITH MELODYSPLASTIC SYNDROMES
A. Bains1,*, L. Behrens Wu2, M. Richter1, V. Magno3, S. Rother4, M. Cross1, C. Werner3,
M. Wobus2, U. Platzbecker1
1Medical Clinic and Policlinic I, Hematology and Cellular Therapy, University Hospital
Leipzig, Leipzig; 2Department of Medicine I, University Hospital Carl Gustav Carus;
3Leibniz Institute of Polymer Research, Technische Universität, Dresden, Dresden;
4Center for Molecular Signaling, Saarland University School of Medicine, Homburg (Saar),
Germany
Background: Myelodysplastic syndromes (MDS) are characterised by ineffective hematopoiesis
and peripheral cytopenia in one or more hematopoietic lineages. MDS is driven by genetic
and epigenetic lesions in the hematopoietic clone itself, but also by changes in the
bone marrow microenvironment. Here, the mesenchymal stromal cells (MSCs) regulate
hematopoietic cell behaviour through cell-cell contact, paracrine signalling and metabolic
support. The MSC-derived Extracellular matrix (ECM) plays a key role in coordinating
both physical and biochemical interactions but the involvement of ECM in MDS has yet
to be determined.
Aims: We aimed to characterise the relevant parameters of the mesenchymal cell-derived
ECM generated by MSCs from healthy donor and MDS patient bonemarrow.
Methods: MSC from low risk (LR) and high risk (HR) MDS patients and age-matched healthy
donors were cultured for 10 days in DMEM Glutamax medium supplemented with 10% FBS
on glass slides pre-coated sequentially with poly-octadecene-alt-maleic anhydride
(POMA) and human fibronectin. The confluent monolayers were washed in PBS, decellularized
by washing with 20mM ammonia and were subject to DNase treatment. The resulting ECM
were stored for up to 4 weeks at 4oC. Matrix structure was analysed by scanning electron
microscopy (SEM) and atomic force microscopy (AFM), glycosaminoglycan (GAG) content
by staining with peanut and wheat germ agglutinin and collagen composition by immunostaining
with antibodies to collagens 1 and 4. Total collagen content was quantified by Sircol
assay. In parallel, the expression of matrix-related genes was measured by real time
PCR of RNA extracted from the MSCs.
Results: In SEM, the ECM network generated by MSCs derived from both low risk (LR)
and high risk (HR) MDS was thicker and of a higher density than that produced by MSCs
from healthy donors. AFM analysis showed ECM of MDS-derived MSCs also had a significantly
reduced rigidity. An increase in overall collagen content in MDS-derived ECM was detected
by sircol assay and confirmed by immunostaining for the most prevalent collagen subtypes
1 and 4. Lectin staining revealed disease stage-specific differences in GAG composition:
The levels of both GAGs carrying N-acetyl glucosamine and those carrying N-acetyl
galactosamine sugars were increased in ECM from LR-MDS, while ECM from HR-MDS retained
high levels of N-acetyl glucosamine but carried only low levels of N-acetyl galactosamine
GAGs. Finally, the pattern of hyaluronan synthase (HAS) gene expression differed markedly
between MSCs from healthy donor, LR- and HR-MDS. Specifically, HAS1 was strongly increased
in LR-MDS and remained significantly above control levels in HR-MDS, while HAS3 was
down-regulated specifically in HR-MDS. Consistent with this, increased levels of low
molecular weight hyaluronic acid was observed in ECM from LR-MDS
Summary/Conclusion: We report both structural and compositional differences between
the MSC-derived ECM from healthy donors, LR-MDS and HR-MDS. The thicker and softer
structure of the MDS matrix may be both a consequence and a driver of changes in the
MSC phenotype. Increase in the major collagens 1 and 4 are consistent with an increase
in overall ECM, but it remains unclear whether the decreased rigidity of ECM in MDS
is due to differences in protein cross-linking or in the levels of hydrated glycosaminoglycans.
MDS-associated changes in hyaluronic acid expression suggest that MSC-derived HA may
contribute to the inflammatory bone marrow state characteristics of LR-MDS.
P725: METABOLIC PROPERTIES OF MESENCHYMAL STROMAL CELLS IN MYELODYSPLASTIC SYNDROMES
E. Balaian1,2, K. Sockel1,2, M. Cross3, A. Funk4, M. Bornhäuser1,5, T. Chavakis4,5,
M. Wobus1,2,*
1Medical Department 1, University Hospital Carl Gustav Carus Dresden, Dresden; 2German
Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ),
Heidelberg; 3Medical Department I - Hematology and Cell Therapy, University of Leipzig
Medical Center, Leipzig; 4Institute for Clinical Chemistry and Laboratory Medicine,
University Hospital Carl Gustav Carus Dresden, Dresden; 5National Center for Tumor
Diseases, Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg,
Germany
Background: The crosstalk between the mutant hematopoietic stem and progenitor cells
and the bone marrow microenvironment plays an important role in the pathogenesis of
myelodysplastic syndromes (MDS). Mesenchymal stromal cells (MSC) represent one of
the key cellular elements in this context. The functional properties of MDS-derived
MSCs including impaired hematopoietic support, as well as increased senescence and
defective osteogenic differentiation have been extensively studied. Additionally,
the detrimental effect of high erythropoietin (Epo) concentration on MDS-MSCs has
been demonstrated by our group. However, whether these deficiencies are associated
with cellular metabolic changes has not been hitherto addressed.
Aims: We aimed to assess the basal metabolic characteristics in MDS-derived MSCs and
the effect of Epo on their energetic profile in vitro.
Methods: MSC from bone marrow aspirates of patients with MDS (low risk n=3, MDS del5q
n=1, chronic myelomonocytic leukemia n=2, high risk n=3) or age-adjusted healthy donors
(n=3) were isolated by Ficoll density centrifugation and plastic adherent culture.
Metabolic measurements were performed before and after incubation with Epo (50 IU/ml)
for 2 days. Seahorse X96 Flux Analyzer (Seahorse Biosciences, Billerica, MA, USA)
was used to analyse glucose metabolism by oxygen consumption rate (OCR) and extracellular
acidification rate (ECAR). Metabolite concentrations were measured in the culture
supernatants using NMR spectroscopy.
Results: MSC from healthy donors demonstrated a higher OCR/ECAR ratio compared to
MDS counterparts (6.19±4,71 vs 1.14±0.28, p=0.005), suggesting enhanced glycolysis
engagement of MDS-MSCs. This was confirmed by increased levels of lactate in the culture
medium from MDS-MSCs after 48 hours of cultivation (healthy: 3.78±0.28 mM vs MDS:
6.75±2.35 mM, p=0.049), whereas the concentrations of pyruvate, alanine and acetate
did not differ. Treatment with Epo resulted in a significant increase of the OCR/ECAR
ratio (from 1.14±0.28 to 3.25±1.75, p=0.003) in MDS-MSCs, whereas changes in healthy
MSCs were not significant (from 6.19±4,71 to 3.81±1.86, p=0.46).
Summary/Conclusion: In contrast to healthy MSCs which rely mainly on mitochondrial
respiration, MDS-MSCs preferentially utilize the glycolytic pathway under normoxic
conditions in vitro. Epo treatment leads to increased mitochondrial respiration in
MDS-MSCs, but not in healthy counterparts. Whether this effect has a causative link
to the disturbed function of MDS-MSCs remains to be clarified.
P726: MYELOID-DERIVED SUPPRESSOR CELLS AS POTENTIAL TARGETS FOR IMMUNE THERAPY IN
CCUS AND LOW RISK MDS
S. Bentivegna1,*, M. O. Holmström2, M. H. Andersen2, K. Grønbæk1,3
1Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen; 2National
Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University
Hospital, Herlev; 3Department of Hematology, Rigshospitalet Copenhagen University
Hospital, Copenhagen, Denmark
Background: Myeloid-derived suppressor cells (MDSC) are powerful immune suppressing
cells that infiltrate the bone marrow (BM) microenvironment in myelodysplastic syndrome
(MDS) and suppress both a potential immune response against the malignant HSPC and
the normal hematopoiesis (Chen et al., 2013). Different groups reported an expansion
of MDSC in MDS, and an increased number of MDSC is associated with disease progression
(Chen et al., 2013; Kittang et al., 2016). MDSC mediate their immunoregulatory effects
releasing immunosuppressive cytokines, such as TGFβ, and enzymes such as indoleamine
2,3-dioxygenase (IDO) and arginase (ARG) (Mondanelli et al., 2017). The presence of
self-reactive T cells that specifically target immunosuppressive cells in both healthy
donors and cancer patients has previously been described (Andersen, 2017). These cells,
termed anti-regulatory T cells (anti-Tregs), recognize epitopes derived from immunosuppressive
proteins such as IDO, ARG, which are expressed by MDSC (Andersen, 2018). We speculate
that the stimulation of anti-Tregs cells using peptide-based vaccines can lead to
the depletion of the immunosuppressive cells in the BM, and it will help to prevent
disease progression in patients with clonal cytopenia of undetermined significance
(CCUS) or MDS.
Aims: The aim of this work was to characterize the immune cell composition of blood
and BM samples from CCUS/MDS patients and investigate the presence of circulating
anti-Treg cells in blood samples and their reactivity towards immunogenic peptides.
Methods: Using flow cytometry we analyzed blood and BM samples from 20 patients (pts)
with CCUS/MDS (10 CCUS and 10 low risk-MDS) and 6 elderly healthy controls (EHC).
The presence of anti-Tregs was investigated in 7 pts, by ELISPOT assay on patient
T cells that had been stimulated with epitopes derived from the immunosuppressive
proteins ARG, IDO or TGFβ.
Results: Although there were no significant differences in the percentage of MDSC
in BM samples between controls and pts, the proportion of IDO+ and ARG+ MDSC in pts
was significantly higher compared to EHC. In addition, there was a significantly higher
percentage of CTLA-4+ and PD-1+ T cells present in the BM samples from pts compared
to EHC. Interestingly, in the ELISPOT assay, 4 out the 7 pts showed a significant
response towards the ARG peptide, 2 showed a response towards IDO peptide and in 4
of the patients there was a significant response towards the TGFβ peptide. Overall,
of the 7 pts analyzed, 6 harbored T cells specific to one or more of the immunosuppressive
proteins ARG, IDO and TGFβ (figure 1).
Image:
Summary/Conclusion: There is an increased proportion of activated MDSC in BM in pts
with CCUS/MDS compared to EHC. As the immune checkpoint molecules are expressed after
T cell activation, the increased percentage of CTLA-4+ and PD-1+ cells in pts BM suggests
that there is a T cell immune response in the BM of pts with CCUS/MDS. The ELISPOTs
showed that circulating anti-Tregs are present in CCUS/MDS patients. These results
suggest, that the tumor specific immune response in patients with CCUS/MDS is present,
albeit suppressed by (amongst others) MDSC. We speculate that therapeutic cancer vaccination
targeting immunosuppressive cells such as MDSC by vaccination using e.g. IDO, ARG
or TGFβ derived epitopes will incite an anti-regulatory immune response, which may
break the local immune suppression in the BM and allow effector immune cells to exert
killing of transformed cells.
P727: NK-CELL IMMUNOEDITING BY MESENCHYMAL STROMAL CELLS IN MDS
V. Bisio1,2,*, B. Schell1,2, L.-P. Zhao1,2, E. Lereclus1,2, M. Boy1,2, A. Bonaud1,2,
A. Caignard1,2, A. Toubert1,2,3, P. Fenaux4, M. Espéli1,2, K. Balabanian1,2, L. Adès4,
N. Dulphy1,2,3
1Université de Paris, Institut de Recherche Saint Louis, EMiLy, Inserm U1160; 2OPALE
Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis;
3Laboratoire d’Immunologie et d‘Histocompatibilité, Assistance Publique des Hôpitaux
de Paris (AP-HP), Hôpital Saint-Louis; 4Université de Paris, Department of Hematology,
AP-HP, Paris, France
Background: Disorders of the immune system contribute to the pathophysiology and progression
of myelodysplastic syndrome (MDS). Cytotoxic Natural Killer (NK) lymphocytes have
been extensively studied for their anti-leukemic activity. However, whether the dysplastic
bone marrow (BM) in MDS can directly imprint NK cells and hence alter immunosurveillance
is still controversial. Extensive characterizations of MDS patients uncovered deep
perturbations in the phenotype and function of cytotoxic lymphocyte subsets and of
the stromal compartment, in particular mesenchymal stromal cells (MSC), linked with
disease predisposition, initiation and progression. However, few studies have been
undertaken to characterize the interactions established within the MDS immune microenvironment
between immune and stromal cells.
Aims: Recent studies on MDS BM ecosystem suggest a possible symbiotic relationship
between cancer, immune and stromal cells. Our project thus aims to underpin the influence
of the pathological MSC from MDS BM over NK cells to create a leukemia-permissive
microenvironment allowing disease development and progression.
Methods: We performed an extensive in vitro characterization of the NK and MSC interactions
by co-culture system. Healthy donor (HD) NK cells were co-cultured either with MSC
isolated from HD or MDS patient BM samples and were analyzed by high-parametric flow-cytometry,
qRT-PCR, secretome and imaging assays.
Results: Analyses of supernatants from MSC/NK co-cultures unveiled altered secretion
of soluble factors related to inflammation and BM homing such as IL6, CCL2 and CXCL10
in the pathologic condition compared to the healthy one. MDS MSC did not support efficiently
NK-cell viability. Moreover, NK cells cultured with MDS MSC were less efficient at
eliminating cancer cells compared to NK cells cultured with HD MSC. Finally, metabolic
analyses uncovered an altered NK cell energetic profile dictated by the different
MSC that could contribute to their poor survival and reduced cytolytic function.
Summary/Conclusion: Altogether, our results suggest that MSC from MDS BM edit NK cells
to promote an immunosuppressive environment facilitating MDS escape and progression.
Further analyses are ongoing to underpin the pivotal role of immune-microenvironment
interactions in the treatment response. We expect this study to identify new biomarkers
to forecast disease progression and open an avenue for new combination therapies targeting
the tumour microenvironment.
P728: GENE EXPRESSION PROFILE REVELS T-LYMPHOCYTES ACTIVATION THROUGH CELL CYCLE PROGRESSION
AND MITOCHONDRIAL METABOLISM REGULATION IN MYELODYSPLASTIC SYNDROMES WITH DEL(5Q)
AFTER LENALIDOMIDE TREATMENT
M. del Rey1,*, L. A. Corchete2, T. González1, F. López-Cadenas3, E. Lumbreras1, S.
M. Toribio1, B. Xicoy4, J. Sánchez-García5, T. Bernal6, G. F. Sanz7, P. Fenaux8, J.
M. Hernández-Rivas9, M. Diez-Campelo10
1Cancer Research Center - University of Salamanca; Instituto de Investigación Biomédica
de Salamanca (IBSAL); 2Instituto de Investigación Biomédica de Salamanca (IBSAL);
Hematology Department, Hospital Clínico Universitario de Salamanca; Centro de Investigación
Biomédica en Red de Cáncer (CIBERONC); 3Hematology Department, Hospital Clínico Universitario
de Salamanca, Salamanca; 4Hematology Department, Institut Català d’Oncologia-Hospital
Germans Trias i Pujol; Josep Carreras Leukemia Research Institute, Universitat Autònoma
de Barcelona, Badalona; 5Hematology Department, Hospital Reina Sofía, Córdoba; 6Hematology
Department, Hospital Universitario Central de Asturias, Oviedo; 7Hematology Department,
Hospital Universitario y Politécnico La Fe, Valencia, Spain; 8Hôpital Saint-Louis,
Paris, France; 9Hematology Department, Hospital Clínico Universitario de Salamanca;
Cancer Research Center - University of Salamanca; Instituto de Investigación Biomédica
de Salamanca (IBSAL); 10Hematology Department, Hospital Clínico Universitario de Salamanca;
Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
Background: Lenalidomide is a potent drug whose mechanisms of action in del(5q) clone
from Myelodysplastic Syndromes (MDS) have been extensively described. Lenalidomide
also induces immune modulation independent of del(5q) and has multiple effects on
T-cell signaling; however, the molecular and cellular determinants that mediate this
immunomodulatory function remain elusive.
Aims: To better understand the immunomodulatory activity of lenalidomide in MDS patients
with del(5q) by in vivo sequential gene expression profile studies of T lymphocytes
in response to the treatment.
Methods: Our study was conducted in patients included in the Sintra-REV Clinical Trial
(phase III). Sequential study was carried out in 26 samples from 13 paired patients.
Seven out 13 were treated with lenalidomide and achieved a major erythroid and cytogenetic
response. Peripheral blood (PB) samples were collected before and one month after
treatment in treated patients and at the same time points for non-treated patients.
CD3+ cells were collected from PB samples and total RNA was isolated. SureSelect Strand
Specific RNA library (Agilent Technologies) was applied to study changes in RNA levels.
Raw reads were aligned against the Human genome GRCh37 using the STAR aligner. Counts
were assigned to Ensembl gene IDs through HTseq using its UNION version. Differential
gene expression was determined with DESeq2, considering as statistically significant
those genes with FDR < 0.05. Pathway over-representation analysis (ORA) was conducted
in the Webgestalt suite.
Results: 332 genes were differentially expressed in CD3+ lymphocytes one month after
lenalidomide treatment in our cohort of patients; of note, none of them were observed
after one month in non-treated patients. The ORA revealed significant differences
in the gene expression profile of sixteen cytokines (e.g. IL10, TNFSF10, IFNG, IL6
and MBP6). These genes could contribute to activation of an antileukemic immune response,
help to correct the anemia as well as attenuate inflammation signaling in MDS patients
with del(5q). The ORA also showed an enrichment of genes involved in cell cycle (35
genes). The most represented up-regulated genes related to this pathway were: cyclines
(CCNB1, CCNB2, CDK1), centromere genes (CENPE, CENPM, CENPU), kinesin family members
(KIF18A, KIF23, KIF2C) and genes involved in cell division (CDC6, CDC7, CDC25A). It
has been described that lenalidomide inhibits CDC25A selectively in the del(5q) clone
resulting in G2/M arrest and apoptosis. By contrast, our study showed that this gene
was upregulated in T-lymphocytes promoting cell cycle and proliferation of these cells.
In addition, 456 genes were differentially expressed between non-treated and lenalidomide
treated samples; 30 of them were related to mitochondrial metabolism (e.g HIBCH, ECHS1,
DECR1, ACSM1, HADH, COQ3, COQ5, SDHAF3, SDHAF4) and mitochondrial translation (e.g.
MRPL15, MRPL24, MRPL48, MRPL51, MRPS33). Thereby, T lymphocytes adaptive responses
as well as sustained killing by cytotoxic T cells could be mediated by mitochondria
in treated patients.
Summary/Conclusion: To our knowledge, this is the first report describing RNA expression
profiles in PB CD3+ lymphocytes collected from lenalidomide-treated del(5q) patients,
contributing to overall understanding of lenalidomide action. A better understanding
of the molecular targets that mediate the immunomodulatory activity is needed to harness
the full potential of this agent. Functional experiments trying to test the new biological
mechanisms described above are currently conducted.
P729: DEREGULATION OF NONCODING RNAS DERIVED FROM KDM GENES IS ASSOCIATED WITH AZACITIDINE
RESPONSE IN PATIENTS WITH MYELODYSPLASTIC SYNDROMES AND ACUTE MYELOID LEUKEMIA
M. Dostalova Merkerova1,*, A. Hrustincova1, I. Trsova1, Z. Krejcik1, D. Kundrat1,
J. Klema2, K. Szikszai1, A. V. Le2, J. Cermak3, A. Jonasova4, M. Belickova1
1Department of Genomics, Institute of Hematology and Blood Transfusion; 2Department
of Computer Sciences, Czech Technical University; 3Laboratory of Anemias, Institute
of Hematology and Blood Transfusion; 4First Department of Medicine, General University
Hospital, Prague, Czechia
Background: Prediction of response to azacitidine (AZA) treatment is an important
challenge in the management of higher-risk myelodysplastic syndromes (MDS) and acute
myeloid leukemia (AML). To date, no clinical, cytogenetic, or molecular markers of
AZA treatment outcome have been validated to support clinical decisions. Moreover,
little is known about how AZA efficiency can be affected by various noncoding RNAs
(ncRNAs), such as long ncRNAs (lncRNAs) and circular RNAs (circRNAs). KDM genes encode
demethylases of histone lysine residues (e.g., KDM1 mediates demethylation of H3K4
and H3K9, and KDM4 acts on H3K9, H3K36, and H1K26). KDMs are often differentially
expressed in leukemia and they cooperate with transcription factors to activate or
repress gene expression.
Aims: Our aims were to provide biological insight into the contribution of ncRNAs
to AZA mechanisms of action and to propose novel disease biomarkers that would be
able to predict future response to AZA treatment. Based on our results, we particularly
focused on expression and function of KDM-related ncRNAs.
Methods: Whole transcriptome RNA sequencing (RNA-seq) was performed in CD34+ bone
marrow cells from 26 patients with MDS or AML with myelodysplasia-related changes
(AML-MRC) before AZA treatment (11 responders and 16 nonresponders), and 9 healthy
controls. Expression of protein coding genes (PCGs), lncRNAs and circRNAs was bioinformatically
processed.
Results: Overall, differential expression analysis between AZA responders and nonresponders
identified significant deregulation of 202 PCGs, 34 lncRNAs, and 21 circRNAs. Surprisingly,
machine learning data showed that predictive potential of ncRNAs was stronger than
that of PCGs. Within the data, deregulation of several KDM-related ncRNAs was observed,
e.g., CTC-482H14.5 (antisense RNA to KDM4B gene, downregulated in AZA responders),
hsa_circ_0003889 (circRNA processed from KDM1A, downregulated in AZA responders),
and hsa_circ_0001580 (circRNA processed from KDM1B, upregulated in AZA responders).
By contrast with these noncoding transcripts, the levels of corresponding PCGs (namely
KDM1A/1B/4B) were not altered between AZA responders and nonresponders, suggesting
that the deregulated ncRNAs play specific roles independent of their host genes. Further,
the best predictor genes were defined by machine learning using RNA-seq data and pathway
analysis linked these markers to cellular processes related to MDS/AML. Interestingly,
CTC-482H14.5 was identified as one of the strongest predictors of AZA treatment response.
Expression of this KDM-related ncRNA was coregulated with expression of DNMT1, EZH2,
and multiple MCM genes, associating this transcript with epigenetic regulation and
recombinational repair pathways.
Summary/Conclusion: Several ncRNAs processed from KDM genes seem to be closely related
to the responsiveness of MDS/AML-MRC patients to AZA treatment. In this study, novel
functions were predicted for these ncRNAs, pointing to possible mechanisms by which
their deregulation could affect AZA treatment response.
Acknowledgements: Supported by GA CR (No. 20-19162S) and MH CZ-DRO (UHKT, 00023736).
P730: GENETIC LANDSCAPE OF SOMATIC MYELOID MUTATIONS IN THE PRESENCE OF RARE TERT
VARIANTS AND THEIR RELATION WITH MYELOID NEOPLASIA
A. Ferrer1,2,*, A. Mangaonkar1, T. Lasho1, M. Wylam3, C. Finke1, J. Fernandez1, R.
He4, D. Viswanatha4, M. Patnaik1,2
1Division of Hematology; 2Center for Individualized Medicine; 3Division of Pulmonary
and Critical Care Medicine; 4Department of Hematopathology, Mayo Clinic, Rochester,
United States of America
Background: Genetic variants in TERT, the gene that encodes telomerase reverse transcriptase,
can result in accelerated telomere attrition. Impaired telomere maintenance is implicated
in the pathogenesis of myelodysplastic syndromes (MDS), with mechanisms remaining
to be elucidated.
Aims: To explore the clinical and genetic associations of TERT variants in an unselected
cohort of patients screened with a myeloid mutation next generation sequencing panel
due to clinical suspicion of a myeloid neoplasia.
Methods: Using a research protocol approved by the Mayo Clinic Institutional Review
Board, we performed a retrospective chart review to identify a cohort of unselected
individuals within all historical data at our institution that had undergone somatic
mutational analysis for suspected myeloid neoplasia using our insitutional amplicon
based next generation sequencing panel (42 genes, including exons 2 to 16 of TERT,
with a read depth of at least 250X). TERT variants were classified into those with
CADD score >20 and <20 in line with previous literature to identify variants within
the top 1% according to their predicted deleteriousness. Additionally, we identified
within our cohort individuals that received a diagnosis of MDS, clinical and survival
information.
Results: We identified 55 different TERT variants (46 missense, 3 in canonical splice
sites, 4 synonymous and 2 small inframe deletions) in 148 individuals tested from
April of 2016 to November of 2021 (Fig. 1A&B). The five most recurrent variants were
c.1234C>T (48 patients), c.1323_1325del (29 patients), c.3164C>T (5 patients), c.604G>A
(4 patients) and c.969G>A (4 patients), all with CADD<20. None of these patients were
diagnosed with a bona fide telomere biology disorder and no clinical information about
telomere length status was available. All variants were considered germline since
their allele frequency was near 50%, although no germline confirmation was performed.
All variants were clinically classified as uncertain significance according to the
American College of Medical Genetics guidelines. Of the 148 total patients, 17 (12%)
carried TERT variants with CADD>20 that were distributed thorough all the gene domains
with no clustering or presence of hotspots (Fig.1A&B). 59% (10 patients) of CADD>20
variants carriers were diagnosed with MDS: 5 patients with <5% bone marrow blasts
(4 with normal karyotype) and 5 patients with 5-19% blasts, all with complex karyotype.
This percentage was significantly higher compared to CADD<20 variant carriers (34%,
49 patients, p=0.048 by Chi-square test, Fig. 1C). However, no major differences were
found between the two groups in terms of age at diagnosis (median 70 vs 73 years old,
p = 0.2305) or AML-free survival (median 10 vs 15.5 months since diagnosis, p=0.4674).
Similarly, the percentage of patients presenting additional somatic variants in other
myeloid-related genes was similar between CADD<20 and CADD>20 TERT variant carriers
(60% vs 64%). The two most frequenly mutated genes in CADD>20 carriers were TP53 (22%),
TET2 (18%), RUNX1 (9%), U2AF1 (9%) and BCOR (9%) while in the CADD<20 carriers were
TET2 (14%), ASXL1 (11%), SRSF2 (9%), TP53 (7%) and JAK2 (6%). Our data showed that
TP53 pathogenic variants were significantly enriched in CADD>20 TERT variant carriers
compared to CADD<20 carriers. (22.7% vs 7.4%, p=0.0169, Fig. 1D).
Image:
Summary/Conclusion: Carriers of TERT variants with CADD score of >20 were more likely
to be associated with a diagnosis of MDS and with somatic TP53 mutations in comparison
to those with CADD score <20, findings that we plan to validate functionally and prospectively.
P731: INTERPLAY BETWEEN INFLAMMATION AND EPIGENETICS IN TUMOR REPROGRAMMING OF MESENCHYMAL
STROMAL CELLS (MSCS) IN MYELODYSPLASTIC SYNDROMES (MDS)
C. Giallongo1,*, I. Dulcamare2, D. Tibullo3, D. Pieragostino4, M. C. Cufaro4, M. A.
Amorini5, G. Lazzarino6, A. Romano7, G. Scandura2, T. Zuppelli2, M. Di Rosa8, A. Duminuco2,
G. Broggi9, R. Caltabiano9, R. Floresti10, R. Motterlini11, F. Di Raimondo7, G. A.
Palumbo1
1Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia;
2Division of Hematology, AOU Policlinico; 3Department of Biomedical and Biotechnological
Sciences, Section of Biochemistry, University of Catania, Catania; 4Department of
Innovative Technologies in Medicine and Dentistry, ‘‘G. d’Annunzio’’, University of
Chieti-Pescara, Chieti; 5Department of Biomedical and Biotechnological Sciences, Section
of Biochemistr, University of Catania, Catania; 6UniCamillus - Saint Camillus International
University of Health Sciences, University of Rome, Rome; 7Department of General Surgery
and Medical-Surgical Specialties; 87Department of Biomedical and Biotechnological
Sciences, Human, Histology and Movement Science Section; 9Department of Medical and
Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Anatomic Pathology,
University of Catania, Catania, Italy; 10University Paris-Est Créteil, INSERM, IMRB,
University Paris-Est; 11University Paris-Est Créteil, INSERM, IMRB, University Paris
Est, Paris, France
Background: The alteration of hematopoiesis in MDS patients is deeply associated with
microenvironment alterations, in particular in MSCs. Stromal cells are epigenetically
reprogrammed to function in cooperation with leukemic cells and propagate the disease
as a “tumor unit”. Notably, dysfunctions of MDS-MSCs persist following expansion ex
vivo suggesting a hereditable epigenetic dysregulation which endures despite removal
of disease-associated microenvironment factors.
Aims: Here, we investigated the role of histone H2A variant MacroH2A1 (mH2A1) in promoting
pro-tumorigenic inflammation and metabolic reprogramming of MSCs.
Methods: MSCs were collected from BM of MDS patients (n=18) and matched healthy controls
(n=8; HC). HS-5 cells were used as model of healthy MSCs. Real time, western blot,
immunohistochemistry and immunofluorescence were used. Proteomic and metabolic analysis
were performed.
Results: MDS patients had significant higher signature of mH2A1 in bone marrow slides
compared to age-matched control. Investigating mH2A1 expression in stromal cells,
MDS-MSCs had increased levels of expression in respect of HC-MSCs. In accordance with
the aberrant inflammation described in MDS microenvironment, MDS-MSCs showed higher
levels of the sum of NO2+ and NO3+ associated to increased GSH and NADP+/NAPH. TLR4
also was upregulated and positively correlated to mH2A1 expression. To better investigate
the relationship between mH2A1 and TLR4 in MDS-MSCs, we induced mH2A1 overexpression
in HS-5 cells (mH2A1-OE) by mH2A1-CT-MYC plasmid. Our data showed that mH2A1-OE upregulated
TLR4 and increased NFkB nuclear translocation compared to cells transfected with empty
vector (CTL). Proteomic analysis confirmed upregulation of intracellular serine protease
inhibitors (SerpinB2, B8, B6) strongly induced during inflammation and important for
the maintenance of TLR4 activation. Moreover, proteomic approach identified upregulation
of several proteins associated to hypermethylation of DNA and histones in mH2A1-OE.
In particular, S-adenosylhomocysteine hydrolase which regulates the concentration
of S-adenosylhomocysteine (SAH), a strong inhibitor of methyltransferase reactions
and of the methyl donor S-adenosyl-methionine (SAM), resulted overexpressed. HPLC
analysis showed higher SAM/SAH ratio associated to a significant reduction of SAH
in mH2A1-OE, confirming the increase of the methylation index. In addition, the higher
levels of CBX3, a suppressive epigenetic mark which recognizes histone H3K9me3, contributes
to the maintenance of the heterochromatin. The higher levels of H3K9me3 in mh2A1-OE
were confirmed by western blot analysis. Overexpression of mH2A1 also induced metabolic
reprogramming with the acquisition of a more glycolytic metabolism characterized by
decreased levels of NAD+/NADH, upregulation of LDHA and MCT4. As LDHA could translocate
into the nucleus, we evaluated if it used a noncanonical enzymatic activity in mh2A1-OE.
Data showed increased nuclear localization of LDHA producing histone H3K79 hypermethylation.
Nuclear LDHA was also observed in MDS-MSC. Finally, we treated ex-vivo HC- and MDS-MSCs
with azacytidine founding a significant reduction both of mH2A1 and TLR4 associated
to lower levels of nuclear NFKB.
Summary/Conclusion: Our data provide a key role of mH2A1in driving the crosstalk between
inflammation, metabolism and epigenetic signatures in MDS-MSC. Future experiments
will define the contribution of mH2A to ineffective hematopoiesis and leukemic evolution.
P732: TH17/TREG IMBALANCE IN LOW RISK MDS PATIENTS
M. Boada1, N. Trias2, A. Brugnini2, C. Guillermo1, S. Grille1,2,*
1Catedra de Hematologia; 2Laboratorio de Citometria y Biologia Molecular. Depto. Básico
de Medicina, Hospital de Clinicas, Montevideo, Uruguay
Background: Myelodysplastic Syndromes (MDS) are a heterogeneous group of clonal haematopoietic
stem/progenitor cells (HSPC) disorders characterized by ineffective haematopoiesis,
peripheral cytopenia, and risk of acute myeloid leukemia (AML). MDS pathogenesis is
complex and depends on the interaction between HSPC and microenvironment. Many studies
have highlighted the role of different immune cells in immune dysregulation leading
to pathogenesis of MDS and progression of disease to AML. Immune response differs
between risk groups favouring a pro-inflammatory profile in low risk, whereas in high-risk
patients there is a trend towards immunosuppressive environment. Regulatory T cells
(TRegs) and T helper 17 cells (Th17) are CD4 T cell functional subsets and their balance
(TH17/TReg) is essential for maintaining immune haemostasis
Aims: To study T cell subsets and cytokines plasma levels in low risk MDS patients
and healthy controls.
Methods: This is a case control study conducted in Hospital de Clinicas Montevideo,
Uruguay. Patient inclusion criteria were Low Risk MDS (R-IPSS <4.5) and control group
were healthy blood donors. We assessed peripheral blood T cell subsets by multiparametric
flow cytometry using the following monoclonal antibodies: CD45, CD3, CD4, CD8, CXCR3,
CCR6, CCR4, CD25 and CD127. Subpopulations were defined as reported by Maecker et
al (1). Cytokine’s plasma levels were determined by Cytometric Bead Array (BD). We
studied pro-inflammatory, TH1, TH2 and TH17 cytokines (IL-2, IL-4, IL-6, IL-10, INFg,
TNFa, and IL-17). The protocol was authorized by the Ethics Committee of the institution
and all subjects signed an informed consent.
Results: We included 24 MDS patients and 19 controls with a median age of 68.5 (47-78)
and 48 (32-55) years respectively. Male:Female ratio was 1.3:1 in controls and 0.9:1
in patients (p˃0.05). According to R-IPSS we included 13 (30.2%) Very Low Risk, 11
(25.2%) Low risk and 19 (44.2%) Intermediate patients. MDS patients showed lower proportion
of Treg cells (p<0.001) and higher TH17/Treg ratio (p<0.001) than controls (Figure
1). There were no statistical differences in total T CD4+ cells and TH1/TH2 ratio.
IL-2, IL-6 and IFN-γ levels were higher in patients than controls. Median levels of
IL-2 were 0.2 (0.1-4.5) pg/mL in patients and 0.1 (0.0-2.6) pg/mL in controls, p=0.002.
Median levels of IL-6 were 9.4 (0.5-10.5) pg/mL in patients and 0.5 (0.2-2.9) pg/mL
in controls, p=0.001. Median levels of IFN-γ were 2.2 (1.1-3.3) pg/mL in patients
and 0.1 (0.0-2.6) pg/mL in controls, p=0.003. There was no statistical difference
in IL-4, IL-10, TNFa, and IL-17A between groups.
Image:
Summary/Conclusion: Our results showed that TH17/Treg balance is impaired, revealing
a predominant proinflammatory state in in MDS Low risk patients. This occurs in concordance
with elevated pro-inflammatory cytokines (IL-2, IL-6 and IFN-γ). These findings agree
with previous reports that evidenced TH17/Treg ratio was higher in low risk MDS patients
when compared with high risk. However, those studies highlighted the importance of
TReg expansion and its prognostic impact in high risk. Evidence comparing TH17/TReg
balance between low risk MDS patients and controls is scant. We believe that these
results support the importance of a pro-inflammatory response in low-risk patients.
1. Maecker HT et al. Standardizing immunophenotyping for the Human Immunology Project.
Nature Reviews Immunology. 2012;12(3):191-200. doi:10.1038/nri3158
P733: DHX9 HELICASE DYSFUNCTION CONTRIBUTES TO DISEASE PROGRESSION IN PATIENTS WITH
MYELODYSPLASTIC SYNDROMES
N.-F. Huang1, C.-K. Chang1, Q. He1, L.-Y. Wu1, Z. Zhang1, X. Li1, F. Xu1,*
1Department of Hematology, ShangHai Jiaotong University Affiliated Sixth People’s
Hospital, ShangHai, China
Background:
DHX9 is a member of the DEAH (Asp-Glu-Ala-His) helicase family, and participates in
regulating DNA replication and RNA processing. DHX9 dysfunction promotes tumorigenesis
in several solid cancers. However, the role of DHX9 in myelodysplastic syndromes (MDS)
is still unknown.
Aims: to investigate the role of DHX9 in the pathogenesis of myelodysplastic syndromes.
Methods:
DHX9 expression was analyzed in 120 MDS patients and 42 non-MDS benign controls by
quantitative PCR. Clinical significance of changes in DHX9 expression was investigated
in disease progression and survival. Lentivirus-mediated DHX9 knockdown experiments
were performed to investigate its biological function. The mechanistic involvement
of DHX9 in MDS development was explored in cell functional assays, gene microarray
and pharmacological intervention. Considering the importance of DHX9 in regulation
of R-loop, we also investigated the role of DHX9 and R-loop in MDS.
Results: We found that overexpression of DHX9 is frequent in MDS patients, and associated
with poor survival and high risk of AML transformation. Cell functional assays showed
that DHX9 is essential for the maintenance of malignant proliferation of leukemia
cells, and knockdown of DHX9 results in increased cell apoptosis and growth inhibition.
Gene expression profile and bioinformatics analysis reveal that DHX9 may be involved
in PI3K-AKT signaling pathways. Validating experiments show that DHX9 is indispensable
for the activation of PI3K-AKT signaling as knockdown of DHX9 inactivates the PI3K-AKT
signaling, reduces the expression of anti-apoptosis genes such as CCND2, MYC and BCL2.
In addition, we found that DHX9 knockdown leads to R-loop accumulation in MDS cells,
and induces R-Loop-dependent DNA damage through ATR-Chk1 activation. It suggests that
overexpressed DHX9 could counteract cell growth defect caused by R-Loop which is quite
common in MDS patients especially in those with splicing genes mutations.
Summary/Conclusion: Our data suggest that DHX9 contributes to disease progression
by activation of PI3K-AKT signaling and correcting R-loop-mediated growth defect in
MDS, and may serve as a novel prognostic marker for AML transformation and therapeutic
target in MDS.
P734: DOWN-REGULATION OF RING1 AND YY1 BINDING (RYBP) LEADS TO EPIGENETIC OVEREXPRESSION
OF ANTI-APOPTOTIC PROTEIN BCL2 IN MYELODYSPLASTIC SYNDROMES
F. Xu1,*, X. Li1, Q. He1, L.-Y. Wu1, Z. Zhang1, C.-K. Chang1
1Department of Hematology, ShangHai Jiaotong University Affiliated Sixth People’s
Hospital, ShangHai, China
Background: Excessive activation of oncogenes plays a crucial role in apoptosis resistance
and disease progression to leukaemia in myelodysplastic syndromes (MDS). RYBP is a
member of BCOR complex for epigenetic transcription repression, and also considered
as an apoptosis-associated protein. However, the role of RYBP is still unclear in
the myelodysplastic syndromes.
Aims: To investigate the role of RING1 and YY1 binding (RYBP) in the myelodysplastic
syndromes.
Methods: The expression of RYBP mRNA and its clinical significance was analyzed in
MDS patients. Lentivirus-mediated RYBP knockdown and overexpression were performed
to investigate its biological function. The mechanism of RYBP in MDS development was
investigated by cell functional assays, gene microarray and chromatin immunoprecipitation.
Results: In this study, we found that reduced RYBP expression is observed in MDS patients
and associated with high AML transformation. Overexpression of RYBP inhibits cell
proliferation, and results in an increased cell apoptosis along with p53 activation
in myeloid cell lines. However, knockdown of RYBP promotes cell proliferation, induces
apoptosis resistance to etoposide and decitabine, and simultaneously reduces H2AK119ub1
level. Gene expression profile analysis identifies BCL2 as a possible target of RYBP.
Further functional analysis shows that depletion of RYBP leads to transcription activation
of BCL2 through removal of H2AK119ub1 at the promoter region of BCL2 gene. BCL-2 specific
inhibitor ABT-199 significantly induces cell apoptosis in RYBP-depleted cells, and
restores the apoptosis sensitivity to etoposide and decitabine.
Summary/Conclusion: Reduced RYBP expression is frequent in MDS patients and associated
with high AML transformation. RYBP inhibition leads to epigenetic overexpression of
anti-apoptotic protein BCL2 in MDS, which contributes to apoptotic resistance of clonal
cells and disease progression. BCL2 inhibitor may be considered as a candidate agent
in advanced MDS.
P735: THE ROLE OF SF3B1 MUTATIONS IN MYELODYSPLASTIC SYNDROMES
S. Huber1,*, S. Hutter1, M. Meggendorfer1, G. Hoermann1, W. Walter1, C. Baer1, W.
Kern1, T. Haferlach1, C. Haferlach1
1MLL Münchner Leukämie Labor GmbH, München, Germany
Background:
SF3B1 is one of the most commonly mutated genes in patients with myelodyplastic syndromes
(MDS), associated with ring sideroblasts (RS) and an indolent disease course. Malcovati
et al. proposed MDS with mutated SF3B1 fulfilling certain criteria as distinct MDS
entity (Malcovati et al., Blood, 2020).
Aims: Study the SF3B1 mutation in MDS with respect to the genomic landscape, AML transformation
rate and clinical outcome.
Methods: We analyzed 734 patients diagnosed with MDS based on cytomorphology, cytogenetics
and molecular genetics and classified according to WHO classification 2017. Amplification-free
WGS was performed with a median coverage of 103x (Illumina, San Diego, CA). Reads
were aligned to the human reference genome (GRCh37, Ensembl annotation, Isaac aligner)
and variants were called using Strelka Somatic Variant Caller v2.4.7.
Results:
SF3B1 mutations were identified in 231 of 734 (31%) MDS patients and were mainly found
in MDS-RS (171/200; 86%; MDS-RS-SLD: 43/51, 84%; MDS-RS-MLD: 128/149; 86%). In addition,
13% (37/300) of MDS with excess blasts (MDS-EB1/2) and 20% of MDS with isolated del(5q)
harbored SF3B1 mutations. In the total cohort SF3B1 mutations were associated with
better outcome (median OS: 79 vs. 53 month; p<0.0001). Within the different MDS entities
SF3B1 mutations were favorable in MDS-RS-SLD (median OS: 106 vs. 25 month; p=0.009),
MDS-RS-MLD (median OS: 82 vs. 64 month; p=0.049) and MDS-EB-2 (median OS: 129 vs.
25 month; p=0.011), but were associated with shorter survival in MDS with isolated
del(5q) (median OS: 69 vs. 79 month; p=0.044). 144/231 (62%) SF3B1 mutated samples
fulfilled the criteria proposed by Malcovati et al. (SF3B1ent). These cases were associated
with longer survival compared to SF3B1 mutated samples not falling into the proposed
SF3B1 entity (SF3B1nent). Within SF3B1ent 47% (67/144) did not harbor any additional
mutation resulting in an average of 1.8 mutations (including SF3B1) in this group.
SF3B1nent patients showed on average more mutations (MDS with isolated del(5q): 1.9;
MDS-EB: 2.7; MDS-RS: 3.1). The most frequent additional mutations in all SF3B1 mutated
patients were TET2 (29%), DNMT3A (16%) and ASXL1 (9%). Regarding cytogenetic abnormalities,
69/231 (30%) SF3B1 mutated samples showed aberrant karyotypes (SF3B1ent: 27/144, 19%;
SF3B1nent: 42/87, 48%). Of SF3B1 mutated patients 7% (15/231) progressed to AML compared
to 15% (75/503) of SF3B1 wildtype patients (median follow-up: 112 months). Notably,
80% of SF3B1nent showed AML transformations in comparison to 20% of SF3B1ent (median
follow-up: 122 and 112 months). Within the 15 SF3B1 mutated patients progressing to
AML 47% showed additional RUNX1 and 27% DNMT3A or TET2 mutations at MDS diagnosis.
In AML-transforming patients with a low SF3B1 variant allelic frequency (VAF, n=2)
additional spliceosome mutations (SRSF2 or ZRSF2) were identified at the time of diagnosis.
Two other SF3B1 mutated patients showing disease progression to AML harbored MECOM
rearrangements when MDS was diagnosed. During progression one patient gained a MECOM
rearrangement and two patients other chromosomal aberrations present in the AML state.
Image:
Summary/Conclusion:
SF3B1 mutations occur in several MDS entities, are associated with good clinical outcome
and mainly co-occurred with TET2 mutations. Patients showed a better prognosis (longer
OS, lower AML transformation rate) when categorized into the proposed SF3B1 entity.
AML transformation of SF3B1 mutated MDS patients is determined by the entire genomic
landscape indicating RUNX1 as potential driver.
P736: BCOR MUTATION REGULATES MYELODYSPLASTIC SYNDROMES PROGRESSION THROUGH REPRESSING
AUTOPHAGY FLUX MEDIATED BY HDAC6 ACTIVATION
C. Jianan1, Y. dongqin2, J. jiacheng1, W. LINGYUN1,*
1xueye, shanghai no.6 hospital; 2xueye, shanghai no.8 hospital, shanghai, China
Background: Past studies have revealed that mutations in bcor are involved in various
human cancers including MDS and AML. Autophagy plays a critical role in cancer progression.
However, the role of bcor mutations in autophagy and MDS progression remains subject
to debate.
Aims: This study investigated the role of bcor mutations in autophagy and MDS progression.
Methods: Cell lines with bcor
p483L stable overexpression were established with a recombinant lentiviral vector.
The effects of bcor mutations on cell proliferation, cell cycle progression, and cell
apoptosis were detected by CCK8 assay, colony formation assay, and flow cytometry.
We detected changes in autophagy levels by Western blot (WB), quantitative polymerase
chain reaction (qPCR), and most importantly, transmission electron microscopy. The
function of histone deacetylase 6 (HDAC6) was inhibited by Tubasatin A (TBA), and
We detected the consequent changes in the functional phenotype of the cells by WB
and qPCR. The interactions between related proteins were analyzed by a co-immunoprecipitation
assay. To test cell lines for mitochondrial dysfunction, bcor
p483L mutant cells, negative control cells, and wild-type cells were double-stained
with MitoTracker Green and MitoTracker Red, or single-stained with MitoSOX followed
by fluorescence-activated cell sorting (FACS) analysis. The changes in pyroptosis
level caused by bcor
p483L mutation were detected by WB and qPCR.
Results: The cell proliferative ability of bcor
p483L mutant cells was repressed compared to the negative control cells. Besides,
bcor
p483L mutation could promote cell apoptosis and block the cell cycle progression to
the G2 phase. Notably, bcor
p483L mutation repressed autophagic flux in cultured cells, as corroborated by decreased
expression of autophagy marker genes (e.g., ATG7, ATG10) and reduced numbers of autophagosomes
or autolysosomes, which were accompanied by upregulation of HDAC6, downregulation
of FoxO1 and ac-FOXO1. HDAC6 was required for inhibiting the deacetylation of FoxO1.
Enhanced HDAC6 expression and repression of FOXO1 were required for bcor
p483L -mutation-deduced autophagy. Depression of HDAC6 by TBA was sufficient for increasing
the expression of FOXO1, ac-FOXO1, and inducing autophagic flux. Notably, we examined
the direct interaction between HDAC6 and FOXO1, FOXO1 and ATG7. Bcor
p483L mutated cells accumulated dysfunctional, non-respiring mitochondria and increased
mitochondrial ROS production compared to negative control cells. And we found upward
adjustment of pyroptosis level and DNA damage.
Image:
Summary/Conclusion:
Bcor p.P483L mutation could disrupt BCOR protein structure and function, therefore
reducing the transcriptional repressive effect of BCOR on HDAC6. HDAC6 was upregulated
and decreases the acetylation of FOXO1, further repressing autophagy. We further verified
the protein: protein interaction between HDAC6 and FOXO1, FOXO1 and ATG7. Defects
in the clearance of damaged mitochondria by bcor
p483L-mutation-regulated autophagy contribute to the accumulation of cellular reactive
oxygen species(ROS), thus promoting cell apoptosis and repressing cell proliferation.
The escalation in pyroptosis levels was found after bcor mutations and may be partly
due to increased ROS levels. We also found increased gene damage and increased likelihood
of gene mutations in bcor
p483L mutant cells, which would promote MDS transformation into AML. These findings
indicate a completely novel BCOR-HDAC6-FOXO1-ATG7 axis in the regulation of autophagy
and MDS progression, providing potential possibilities for MDS treatment.
P737: FUNCTIONAL ROLES OF DDX41 MUTATIONS IN THE DEVELOPMENT OF MYELOID MALIGNANCIES
A. Kon1,*, M. Nakagawa1, K. Kataoka2, H. Makishima1, M. Nakayama3, H. Koseki4, Y.
Nannya1, S. Ogawa1
1Dept. Pathology and Tumor Biology, Kyoto University, Kyoto; 2Division of Molecular
Oncology, National Cancer Center Research Institute, Tokyo; 3Department of Technology
Development, Kazusa DNA Research Institute, Chiba; 4Laboratory for Developmental Genetics,
RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
Background:
DDX41 is a newly identified leukemia predisposition gene encoding an RNA helicase,
whose germline mutations are tightly associated with late-onset myeloid malignancies.
Importantly, germline DDX41 mutations were also found in as many as ~3-7 % of sporadic
cases with MDS/AML, in which a germline protein-truncating allele is compounded by
a somatic missense mutation affecting the helicase domain (p.R525H) in the remaining
allele in typical cases. However, little is known about the molecular mechanism by
which DDX41 mutations lead to myeloid neoplasms.
Aims: This study aimed to clarify the molecular pathogenesis of DDX41 mutated myeloid
neoplasms.
Methods: We generated mice models carrying conditional/constitutive Ddx41 knock-out
(KO) and conditional R525H knock-in (KI) alleles in various combinations. By crossing
these mice with Rosa26-CreERT2 transgenic mice, we obtained heterozygous Ddx41 KO
(Ddx41
+/-), homozygous Ddx41 KO (Ddx41
-/-), heterozygous Ddx41 R525H mutation (Ddx41
R525H/+), and Ddx41 R525H/KO (
Ddx41
R525H/-) mice, in which expression of the mutant allele was induced by tamoxifen administration.
We then investigated the effect of these alleles on hematopoietic cells.
Results: First, we assessed cell intrinsic effects of these Ddx41 alleles, in noncompetitive
transplantation experiments. Shortly after tamoxifen administration, most of the recipient
mice that were reconstituted with bone marrow (BM) from Ddx41
-/- or Ddx41
R525H/- mice died within a month after CreERT2 induction due to severe BM failure
(BMF). By contrast, the mice transplanted with BM from Ddx41
+/- mice showed significantly reduced WBC counts and myeloid skewing in long-term
observation.
We also assessed the reconstitution capacity of BM cells from different Ddx41 mutant
mice using peripheral blood (PB) chimerism after competitive transplantation with
Ddx41
+/+ cells. The PB donor chimerism of Ddx41
-/- and Ddx41
R525H/- mice-derived cells was markedly reduced compared to that of Ddx41
+/+ cells. In contrast, Ddx41
+/- and Ddx41
R525H/+ mice-derived cells showed a slightly reduced PB chimerism compared to Ddx41
+/+ mice-derived cells. Of interest, when Ddx41
R525H/--derived BM cells were co-transplanted with Ddx41
+/--derived BM cells, some of the recipient mice showed impaired hematopoiesis, suggesting
the presence of a non-cell autonomous effect of Ddx41
R525H/- cells on Ddx41
+/- cells.
Transcriptome analysis of stem cells (Kit+Sca-1-Linlow cells) from different Ddx41
mutant mice revealed significant changes in gene expression and splicing patterns
in many genes in all mutant mice, with larger changes for Ddx41
R525H/- than Ddx41
+/- and Ddx41
R525H/+ cells. Notably, Ddx41
R525H/--derived stem cells exhibited a significant upregulation of genes involved
in innate immunity, including an upregulation of cGAS-STING innate immunity pathways,
as well as an enhanced Trp53 pathway, whereas there was a downregulation of genes
related to RNA metabolism and ribosome biogenesis.
Summary/Conclusion: Our results suggest that monoallelic Ddx41 loss leads to age-dependent
impaired hematopoiesis, while compound biallelic loss-of-function and R525 alleles
showed early development of severe BM failure, where activated innate immunity and
impaired ribosome functions may play important roles. The role of DDX41 mutations
in myeloid neoplasms, however, need further evaluation.
P738: EXPRESSION OF TRANSPOSABLE ELEMENTS IN CD34+ CELLS IN MYELODYSPLASTIC SYNDROMES
Z. Krejcik1,*, A. Hrustincova1, I. Trsova1, D. Kundrat1, K. Szikszai1, M. Belickova1,
J. Cernak2, A. Jonasova3, M. Dostalova Merkerova1
1Department of Genomics; 2Laboratory of Anemias, Institute of Hematology and Blood
Transfusion; 3First Department of Medicine, General University Hospital, Prague, Czechia
Background: Myelodysplastic syndromes (MDS) are a heterogenous group of malignant
hematopoietic stem cell (HSC) disorders characterized by genomic instability resulting
in aberrant differentiation of HSCs, peripheral blood cytopenia, and a tendency toward
leukemic transformation. Transposable elements (TEs) are common in the human genome
and, with regard to their ability to change the position in the genome, may represent
one of the sources of this instability. Although TE dysregulation has previously been
reported in different hematological malignancies, e.g., in acute myeloid leukemia
(AML), information about TEs in MDS is rare. In the past, Colombo et al. demonstrated
suppressed TE expression in higher risk MDS in a small cohort of patients. They proposed
this suppression as a mechanism for immune escape in MDS/AML because the higher TE
expression can potentially trigger immune-mediated cell clearance of cancer cells
via “viral mimicry” pathways.
Aims: Our aims were to identify, quantify and compare TE expression in different stages
of MDS on a larger cohort of patients to provide a deeper insight into their occurrence
and possible roles in MDS.
Methods: We prepared, sequenced, and analyzed rRNA-depleted RNA libraries of CD34+
HSCs in a cohort of 75 MDS patients, and 13 healthy controls, using the SalmonTE tool
for the TE detection.
Results: Comparison between healthy controls and MDS patients revealed significantly
upregulated class I LTR (Long Terminal Repeat) retrotransposons (p = 0.0006, logFC
= 0.040), especially then ERV1 (Endogenous Retrovirus) class (p = 0.0002, logFC =
0.052), in MDS patients. Detailed analysis revealed 11 significantly (FDR < 0.05)
differentially expressed TEs, nine upregulated (ERV1: HERV-Fc1, MER51E, LTR27E, MER65C,
HUERS-P1, PABL_A, ERV3: MER54B, CR1: X5A_LINE, SINE (Short Interspersed Element):
MIR3) and only two downregulated (ERV1: LTR71A, LTR26E) in the group of MDS patients.
After stratification of patients into lower (IPSS-R score ≤ 3.5, n = 48) and higher
risk (IPSS-R score ≥ 4, n = 27) categories, we observed significant dysregulation
of non-LTR retrotransposons in general, and in particular of SINE (p = 6.2x10-14,
logFC = -0.21) and LINE1 (Long Interspersed Element) (p = 3x10-7, logFC = 0.06) clades.
Detailed differential expression analysis revealed that 18 out of 21 most significantly
(FDR < 0.001) deregulated TEs were suppressed in higher risk MDS, namely AluY, AluYb8,
AluYd8, AluYe2, AluSq4, AluYa1, AluYa5, AluYb3a2, AluYc1, AluYbc3a, AluYe5, AluYf2,
SVA_B and SVA_F (all SINE), L1HS (LINE1), LTR12E and LTR1A2 (both ERV1); and only
three TEs were upregulated (HARLEQUIN, LTR24C, PABL_AI (all ERV1)).
Summary/Conclusion: To our knowledge, this is the first report on TE expression and
MDS-risk specific dysregulation in a larger cohort of MDS patients, revealing their
importance in the pathogenesis of MDS. Significant suppression of TEs observed in
higher risk MDS patients supports the previously formulated potential mechanism of
immune escape in these MDS patients.
Acknowledgements: Supported by AZV CR (NU20-03-00412) and MH CZ-DRO (UHKT 00023736).
Reference: Colombo et al. Suppression of transposable elements in leukemic stem cells.
Sci Rep. 2017. 1;7(1):7029. doi: 10.1038/s41598-017-07356-9.
P739: THE EPI-GENOMIC LANDSCAPE OF MONOSOMY 7 IN ADULT MDS/AML
A. G. Lema Fernandez1,*, C. Nardelli1, V. Di Battista1, M. Quintini1, F. Pellanera1,
C. Matteucci1, V. Pierini1, B. Crescenzi1, M. Moretti1, V. Bardelli1, P. Gorello1,
C. Mecucci1
1Department of Medicine, University of Perugia, Perugia, Italy
Background: Monosomy 7 is one of the most frequent aneuploidies in myeloid malignancies
often occurring in the context of high-risk Myelodysplastic Syndrome (MDS) and Acute
Myeloid Leukemia (AML) and associated to poor prognosis. Despite several studies have
focused on the pathogenetic role of genes mapping at chromosome 7q, its molecular
landscape remains undetermined.
Aims: The aim of this study was to characterize biological features of adult MDS/AML
with isolated monosomy 7 by investigating the epi-genomic landscape.
Methods: Epi-genomic characterization included karyotype, Fluorescent In Situ Hybridization
(FISH), Single Nucleotide Polymorphism Array (SNPa), Targeted Genomic Sequencing,
RNA sequencing and multiplex Enhanced Reduced Representation Bisulfite Sequencing
(mERRBS). This comprehensive approach was used in analyzing a cohort of three adult
MDS/AML cases with isolated monosomy 7 against a cohort of three non-neoplastic cytopenias
and against dic(1;7)(q10;p10) cases previously characterized by our group (Leukemia,
2019).
Results: Karyotype identified the presence of isolated monosomy 7 in all cases, interphase-FISH
showed clonality as accounting for 65-85% of the total bone marrow population. Targeted
sequencing showed multiple variants affecting myeloid genes (mean 5.3 events; from
2 to 9 per case) but did not identify a common mutational marker. Gene expression
profile (FDR<0.1, log2FC>I1I) provided us with a specific signature differentiating
monosomy 7 from both controls [632 differentially expressed genes (DEGs): 405 up and
227 down] and dic(1;7) [1.706 DEGs: 832 up and 874 down]. In keeping with monosomy,
a strong gene dosage effect at chromosome 7 genes emerged. Functional pathways analysis
specifically identified the presence of active stemness programs in monosomy 7, through
deregulation of mTOR pathway, Rho-GTPases and hematopoietic stem cell markers. Deregulation
of genes involved in apoptosis escape, included in TP53 and heat-shock proteins signaling,
supported the undifferentiated phenotype. Interestingly, PTCH1 tumor suppressor downregulation
was a differential feature of monosomy 7 with respect to the other groups and it was
validated in an independent cohort of MDS/AML with isolated monosomy 7. Global methylation
further delineated specific boundaries between monosomy 7 and dic(1;7) cases showing
a methylation gradient that defined monosomy 7 as the most hypermethylated group.
Private localization of differentially methylated regions in monosomy 7 emerged at
intergenic enhancers, enriched for homeo-domain transcription factors binding sites,
and at CTCF binding sites linked to promoters. A subset of hypermethylated enhancers
in monosomy 7 was in accordance with repression of their respective target genes,
almost totally located on chromosome 7 (ARPC1A, CCZ1, IKZF1, PMS2, RBAK, RBM33, RHEB,
RNF216P1, ZNF12 and ZNF853). Their downregulation was related not only to the chromosome
loss but also to the effect of enhancer hypermethylation in the retained chromosome,
supporting a combined cytogenetic and epigenetic mechanism in determining biallelic
deregulation. Among these deregulated genes, IKZF1 and its target genes accounted
for > 6% of the whole typical expression profile.
Summary/Conclusion: In conclusion our results first identified a specific signature
of DNA methylation and an active epi-genomic program reminiscent of stem cells in
MDS/AML with monosomy 7, providing new insights to the knowledge of biological targets
in this aggressive aberration.
P740: IMPACT OF MAGROLIMAB IN COMBINATION WITH AZACITIDINE ON RED BLOOD CELLS IN PATIENTS
WITH HIGHER-RISK MYELODYSPLASTIC SYNDROME (HR MDS)
J. Y. Chen1,*, L. Johnson2, K. M. McKenna2, T. S. Choi2, J. Duan2, D. Feng2, J. M.
Tsai3, N. Garcia-Martin4, K. Sompalli2, R. Maute2, P. Vyas4, R. Majeti5, C. H. M.
Takimoto2, J. Liu1, G. Ramsingh2, M. P. Chao2, J.-P. Volkmer2, I. L. Weissman5
1Stanford University School of Medicine, Stanford; 2Gilead Sciences, Inc., Foster
City; 3Brigham and Women’s Hospital, Boston, United States of America; 4Molecular
Hematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford,
Oxford, United Kingdom; 5Ludwig Cancer Center and Institute for Stem Cell Biology
and Regenerative Medicine, Stanford University School of Medicine, Stanford, United
States of America
Background: Magrolimab is a monoclonal antibody that blocks CD47, a “don’t eat me”
signal expressed on cancer cells, to escape immune surveillance and macrophage-mediated
clearance. Prior preclinical studies have shown that CD47 is critical to red blood
cell (RBC) homeostasis, with CD47 deficiency decreasing RBC half-life. Fc-mediated
opsonization also depletes RBCs, raising concerns that potential on-target anemia
could result from the use of anti-CD47 agents via multiple mechanisms. Nonetheless,
several clinical trials have demonstrated that magrolimab can be safely administered
as monotherapy, with an initial lower “priming” dose yielding transient anemia with
compensatory reticulocytosis and no anemia observed at subsequent higher maintenance
doses. However, the mechanism underlying this observed protection has not been fully
defined.
Aims: To describe manageable anemia in patients with HR MDS treated with magrolimab
in combination with azacitidine (AZA) in a phase 1 clinical trial (NCT03248479) and
further investigate the underlying mechanisms in preclinical models.
Methods: In a multicenter prospective study, complete blood counts (CBCs), peripheral
blood, and bone marrow (BM) were collected at prespecified time points from patients
with HR MDS (n=57) treated with magrolimab in combination with AZA. CBCs were measured,
and blood and BM samples were analyzed by flow cytometry for expression of CD47 on
RBCs and white blood cells (WBCs). Magrolimab was initially administered as a priming
dose (1 mg/kg) followed by an initial weekly maintenance dose (30 mg/kg) before transitioning
to maintenance dosing every 2 weeks. AZA 75 mg/m2 was administered on days 1-7 of
each 28-day cycle. Preclinical modeling studies were conducted with intact and Fc-deficient
anti-mouse CD47 (MIAP410) and anti-human CD47 (magrolimab) antibodies in murine models,
including C57BL/6J B-hSIRPA/hCD47 mice.
Results: Combination treatment with magrolimab and AZA resulted in tolerable anemia
that correlated with rapid, near-complete loss of CD47 in RBCs but not WBCs. The initial
1-mg/kg priming dose was sufficient for this CD47 loss, which persisted with subsequent
30-mg/kg maintenance doses. Both findings are consistent with prior clinical observations
of magrolimab monotherapy in patients with solid tumors and magrolimab in combination
with rituximab in patients with lymphoma. Our preclinical studies with mouse models
revealed that CD47 removal is mechanistically independent of previously described
RBC antigen modulation mechanisms and cellular compartments. Instead, this CD47 loss
requires anti-CD47 cross-linking between RBCs and non-RBCs.
Summary/Conclusion: Overall, these results support the idea that on-target magrolimab-mediated
anemia is mitigated by a near-complete loss of RBC CD47. Patients with HR MDS treated
with magrolimab in combination with AZA had tolerable anemia with the use of priming
and maintenance doses.
P741: DDX41 MUTATIONS DEFINE A UNIQUE SUBTYPE OF MYELOID NEOPLASMS.
H. Makishima1,*, R. Saiki1, Y. Nannya2, S. Korotev3, C. Gurnari4, J. Takeda1, Y. Momozawa5,
S. Best6, P. Krishnamurthy6, T. Yoshizato1, Y. Atsuta7, Y. Shiozawa1, Y. Iijima-Yamashita8,
K. Yoshida1, Y. Shiraishi9, Y. Nagata1, N. Kakiuchi1, M. Onizuka10, K. Chiba9, H.
Tanaka9, A. Kon1, Y. Ochi1, M. Nakagawa1, R. Okuda1, T. Mori1, H. Itonaga11, Y. Miyazaki11,
M. Sanada8, H. Tsurumi12, S. Kasahara13, C. Müller-Tidow14, A. Takaori-Kondo15, K.
Ohyashiki16, T. Kiguchi17, F. Matsuda18, J. Jansen19, C. Polprasert20, S. Miyano9,
L. Malcovati21, T. Haferlach22, M. Kubo5, M. Cazzola21, A. Kulasekararaj6, L. Godley3,
J. Maciejewski4, S. Ogawa1
1Department of Pathology and Tumor Biology; 2Kyoto University, Kyoto, Japan; 3Departments
of Medicine and Human Genetics, Section of Hematology/Oncology, The University of
Chicago, Chicago; 4Department of Translational Hematology and Oncology Research, Taussig
Cancer Institute, Cleveland Clinic, Cleveland, United States of America; 5Laboratory
for Genotyping Development, Center for Integrative Medical Sciences (IMS), RIKEN,
Yokohama, Japan; 6King’s College Hospital NHS Foundation Trust, London, United Kingdom;
7Department of Healthcare Administration, Nagoya University Graduate School of Medicine;
8Department of Advanced Diagnosis, Clinical Research Center, Nagoya Medical Center,
Nagoya; 9Division of Genome Analysis Platform Development, National Cancer Center
Research Institute, Tokyo; 10Department of Hematology and Oncology, Tokai University
School of Medicine, Isehara; 11Department of Hematology, Atomic Bomb Disease and Hibakusha
Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki; 12Department
of Hematology, Gifu University; 13Department of Hematology, Gifu Municipal Hospital,
Gifu, Japan; 14Heidelberg University Hospital, Heidelberg, Germany; 15Department of
Hematology, Kyoto University, Kyoto; 16Department of Hematology, Tokyo Medical University,
Tokyo; 17Chugoku Central Hospital, Fukuyama; 18Center for Genomic Medicine, Kyoto
University, Kyoto, Japan; 19Department of Laboratory Medicine, Laboratory of Hematology,
Radboud University Medical Center, Nijmegen, Netherlands; 20Department of Medicine,
Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital,
Bangkok, Thailand; 21Department of Molecular Medicine, University of Pavia, Pavia,
Italy; 22MLL, Munich Leukemia Laboratory, Munich, Germany
Background:
DDX41 was identified as a causative gene for late-onset familial myelodysplastic syndromes
(MDS) and acute myeloid leukemia (AML). While DDX41 is thought to be one of the most
frequent targets of germline mutations responsible for sporadic cases with AML/MDS
and other myeloid neoplasms (MN), the entire spectrum of pathogenic DDX41 variants
and their effect size therein are still to be elucidated. The clinical and genetic
pictures of DDX41-mutated myeloid neoplasms have not fully been investigated in a
large cohort of patients, either.
Aims: In this study, we enrolled a total of 9,081 sporadic and familial cases with
different MNs from different ethnicities, in which germline and somatic mutations
in DDX41 and other major driver genes in AML/MDS were analyzed using next generation
sequencing.
Methods: To estimate the size of the risk of MN development associated with pathogenic
germline DDX41 variants, we calculated their enrichment in 3,672 MN cases as compared
with a control cohort (n=20,238). We also estimated the penetrance of pathogenic DDX41
germline variants for the development of MNs.
Results: We identified 292 cases with pathogenic or likely pathogenic DDX41 mutations
with conspicuous ethnic diversity, of which 159 (54%) accompanied somatic mutations
as well, while the remaining 53 cases had somatic mutations alone. Among 292 germline
variants, 197 were truncating variants, whereas only 2.6% of somatic mutations were
truncating (P<0.0001). Pathogenic germline mutations were significantly more frequent
in Asian than in Caucasian cases (odds ratio (OR)=1.64), consistent with a significantly
higher frequency of pathogenic germline DDX41 variants in the former cases. We observed
a significant enrichment of 5 DDX41 variants in myeloid neoplasms, including p.A500fs,
p.E7*, p.Y259C, p.E256K, and p.S363del, compared with 20,238 control cases, with a
mean odds ratio of 10.6. To estimate the penetrance of NMs associated with pathogenic
germline variants, we calculated cumulative incidence of MN in a cohort of 384 first-degree
relatives (kin-cohort) of DDX41-mutated NM cases. Penetrance of pathogenic DDX41 germline
variants was only negligible under 40 years of age, but rapidly elevated thereafter,
reaching as high as 38.5% at the age of 85, confirming late onset of DDX41-mutated
MNs.
DDX41 variants were significantly more common in higher risk MDS (7.1%) and secondary
AML (9.7%), compared with other myeloid neoplasms (1.9%). Patients with MDS having
both germline and somatic mutations were more likely to classified in refractory anemia
with excess blasts (RAEB), compared with those with germline or somatic alone (P=0.029).
DDX41 variants were significantly associated with lower WBC and hypocellular bone
marrow. Most frequently co-occurring mutations included those in ASXL1, SRSF2, TET2,
CUX1, and DNMT3A, of which only CUX1 mutations were statistically significant. Among
MDS cases, DDX41 mutations were associated with a significantly higher incidence of
leukemic evolution (P=0.006) than DDX41-unmutated cases, which was virtually confined
to those with truncating DDX41 variants. However, despite this, DDX41-mutated cases
had a better OS than un-mutated cases, particularly when treated with hypomethylating
agents (HMAs) (P<0.001).
Summary/Conclusion: In summary, DDX41-mutated myeloid neoplasms define a distinct
entity of myelodysplasias associated with higher incidence of leukemic evolution and
better responsiveness to hypomethylating agents.
P742: TRANSCRIPTOMIC ANALYSIS OF ISOLATED CD4+ AND CD8+ T CELLS REVEALS DIFFERENCES
IN TCR REPERTOIRE IN PATIENTS WITH HIGH RISK MDS COMPARED TO AML.
K. Katsiki1, A. Tasis1, A. Filia1, E. Lamprianidou1, K. Liapis1, I. Kotsianidis1,
I. Mitroulis1,*
1Department of HEmatology, Democritus University of Thrace, Alexandroupolis, Greece
Background: Myelodysplastic syndrome (MDS) is a group of clonal disorders of hematopoietic
progenitor cells, which often progresses towards acute myeloid leukemia (AML). Even
though MDS is a clonal hematopoietic disease, bone marrow microenvironment and more
specifically inflammation has been linked to its progression. Among other cell types,
CD4+ and CD8+ T cells are major players in the regulation of the microenvironment
in malignancies.
Aims: In this study, we aimed to investigate the molecular signature of bone marrow
CD4+ and CD8+ T cells from patients with high-risk MDS, AML and chronic myelomonocytic
leukemia (CMML).
Methods: RNA sequencing was performed on isolated CD4+ and CD8+ T cells from the bone
marrow of six patients with high risk MDS (RAEB-II), six patients with CMML and four
patients with AML, using cell sorting. Except from the analysis of gene expression,
we performed TCR repertoire analysis using VDJtools, an open-source Java/Groovy-based
framework that can analyze immune repertoire sequencing (RepSeq) data and allows applying
a diverse set of post-analysis strategies. To do so, RNA sequencing data were used
as input for the V-(D)-J junction mapping software and analysis was performed on the
MiXCR platform. The output of MiXCR wereused as input for the VDJtools after running
a convert routine with -S mixcr argument to prepare datasets in a format for VDJtools
analysis.
Results: Analysis of CD8+ T cells from patients with high risk MDS and AML identified
38 differentially expressed genes. We observed that the expression of the TCR variants
TRAV20 and TRAV13-1 was increased in patients with AML compared to high risk MDS (Figure
1A). Further analysis of the TCR repertoire revealed that the diversity of TCR was
higher in CD8+ T cells from patients with AML (Figure 1B), whereas multidimentional
scaling analysis revealed a clear separation of the two groups (Figure 1C). To the
same direction, clustering of the usage of TRAV-TRAJ further confirmed this finding
(Figure 1D). On the other hand, we did not observe any difference in the TCR diversity
of the variant usage when we compared CD8+ T cells from patients with CMML and AML
(Figure 1E-F), or in the comparison between CD4+ T cells from patients with high risk
MDS and AML (Figure 1G-I).
Image:
Summary/Conclusion: CD8+ T cells are a major cell population that exerts anti-tumour
properties in cancer. Herein, we compared the TCR repertoire of CD8+ and CD4+ T cells
in the bone marrow of patients with high risk MDS and AML and observed that despite
the common features of these two disorders, there are significant differences in the
TCR repertoire specifically in CD8+ T cells, especially concerning TCR diversity.
These findings imply that there is more active T cell response in the bone marrow
in AML compared to MDS. However, it is not clear to what degree this T cell response
targets AML neo-antigens.
P743: THE PIVOTAL ROLE OF GLUTAMINOLYSIS IN MYELODYSPLASTIC SYNDROME (MDS): A NOVEL
STRATEGY FOR THE TARGETED THERAPY OF MDS
S. Okabe1,*, Y. Tanaka1, A. Gotoh1
1Department of Hematology, TOKYO MEDICAL UNIVERSITY, Tokyo, Japan
Background: Myelodysplastic syndromes (MDS) is indicated by bone marrow failure and
increased risk of transformation to acute myelogenous leukemia (AML). MDS is more
common with advancing age and a combination of comorbidities is associated with significantly
worse overall survival. Moreover, iron overload (IOL) starts to develop in MDS patients
by transfusion in many cases. Because the outcome of MDS is poor, alternative therapeutic
strategies are required to improve the survival of MDS and AML patients.
Aims: Glutamine is the most abundant amino acid in blood. Glutaminase (GLS) is the
initial enzyme in glutamine metabolism and glutaminolysis contributes to tumor growth.
Because GLS is frequently activated in various types of cancer, GLS inhibitor could
suppress MDS and AML cells in combination with BCL2 inhibitor.
Methods: In this study, we investigated whether GLS was involved in MDS progress.
We also investigated the efficacy of GLS inhibitor (CB-839 or IPN-60090) and BCL2
inhibitor, venetoclax by using MDS and AML cell line, SKM-1, MDS-L, MOLM-14, THP-1,
MV4;11 and osteoblastic cell line, MC3T3-E1.
Results: We first investigated the relationship between GLS expression and MDS patients
by microarray gene expression data from the online Gene Expression Omnibus (GEO).
In mammalian cells, there are two paralogous GLS genes, GLS1 and GLS2. GLS1 expression
was increased in refractory anemia with excess of blasts (RAEB)-2 cells compared to
normal control cells (GSE19429). In contrast, GLS2 expression was not changed. High
GLS1 expression is associated with poor prognosis in AML patients. Deprivation of
glutamine in culture medium revealed that cellular growth inhibition. We next evaluated
the effect of GLS inhibitor (CB-839 or IPN-60090) or venetoclax on proliferation of
MDS and AML cell lines. 72 h treatment of MDS and AML cells were inhibited by GLS
inhibitor or venetoclax in a dose dependent manner. Cellular cytotoxicity and caspase
3/7 activity was also increased. Glutamine is converted by GLS into glutamate and
related nicotinamide adenine dinucleotide phosphate (NADP) production. Intracellular
NADPH and NADH were reduced by GLS inhibitor. Co-treatment with GLS inhibitor and
venetoclax was superior effect than single drug alone. Adenosine triphosphate (ATP)
is the most important source of energy for intracellular reactions. Intracellular
ATP levels drastically decreased. The bone marrow is a relatively hypoxic microenvironment.
Gene expression of GLS1 is increased under hypoxia condition. The proteasome 20S activity
and phosphorylation of nuclear factor-kappa B (NF-κB) were increased. Efficacy of
venetoclax is reduced under hypoxia condition. Co-treatment with GLS inhibitor and
venetoclax overcome in hypoxia mediated drug resistance. GLS1 shRNA transfected cells
reduced cellular proliferation. Cell cycle analysis showed G1 arrest and increased
sensitivity of venetoclax. Because MDS patients with IOL have reduced overall survival
and poorer outcomes, we next examined IOL by using osteoblastic cell line, MC3T3-E1.
The cellular proliferation was reduced by ferric ammonium citrate (FAC). Intracellular
ROS was increased by FAC. GLS inhibitor treatment protected FAC mediated cell death.
Summary/Conclusion: Targeting of glutaminolysis and BCL2 inhibition combine to enhance
therapeutic efficacy and has been proposed as a novel strategy high-risk MDS and AML.
We also provide the promising clinical relevance as a candidate drug for treatment
of MDS and AML patients.
P744: UNBALANCED TRANSLOCATION DER(1;7)(Q10;P10) AS A DISTINCT SUBTYPE IN MYELODYSPLASTIC
SYNDROMES
R. Okuda1,*, Y. Ochi1, K. Chonabayashi2,3, N. Hiramoto4, M. Sanada1,5, H. Handa6,
S. Kasahara7, S. Sato8, N. Kanemura9, T. Kitano10, M. Watanabe3, W. Kern11, M. Creignou12,
Y. Shiraishi13, M. Watanabe14, K. Usuki15, S. Imashuku16, E. Hellstrom-Lindberg12,
T. Haferlach11, S. Chiba17, N. Sezaki18, L.-Y. Shih19, Y. Miyazaki8, Y. Yoshida2,
T. Ishikawa14, K. Ohyashiki20, Y. Atsuta21, Y. Shiozawa1, S. Miyano13,22, H. Makishima1,
Y. Nannya1, S. Ogawa1,12,23
1Department of Pathology and Tumor Biology; 2Center for iPS Research and Application;
3Department of Hematology and Oncology, Kyoto University, Kyoto City; 4Department
of Hematology, Kobe City Medical Center General Hospital, Kobe City; 5Department of
Advanced Diagnosis, Clinical Research Center, National Hospital Organization Nagoya
Medical Center, Nagoya City; 6Department of Hematology, Gunma University Graduate
School of Medicine, Gunma; 7Department of Hematology, Gifu Municipal Hospital, Gifu;
8Japan Adult Leukemia Study Group, Japan Adult Leukemia Study Group, Japan; 9Department
of Internal Medicine, Gifu University, Gifu; 10Department of Hematology, Kitano Hospital,
Osaka, Japan; 11MLL Munich Leukemia Laboratory, MLL Munich Leukemia Laboratory, Munich,
Germany; 12Department of Medicine, Karolinska Institute, Stockholm, Sweden; 13Laboratory
of Sequence Data Analysis, The University of Tokyo, Tokyo; 14Department of Hematology,
Hyogo Prefectural Amagasaki General Medical Center, Amagasaki; 15Department of Hematology,
NTT Medical Center Tokyo, Tokyo; 16Department of Laboratory Medicine, Uji-Tokushukai
Medical Center, Uji; 17Department of Hematology, University of Tsukuba, Tsukuba; 18Department
of Hematology, Chugoku Central Hospital, Hiroshima, Japan; 19Division of Hematology-Oncology,
Chang Gung University, Taoyuan, Taiwan; 20Department of Hematology, Tokyo Medical
University, Tokyo; 21The Japanese Data Center for Hematopoietic Cell Transplantation,
The Japanese Data Center for Hematopoietic Cell Transplantation, Japan; 22Laboratory
of DNA Information Analysis, The University of Tokyo, Tokyo; 23Institute for the Advanced
Study of Human Biology, Kyoto University, Kyoto City, Japan
Background: der(1;7)(q10;p10) (der(1;7)) is an unbalanced translocation between chromosomes
1 and 7 found in 1.5-6% of patients with myelodysplastic syndromes (MDS), depending
on different ethnicity, where the common consequence is +1q and -7q. Previous studies
have noted a higher incidence of RUNX1 mutation and better prognosis for der(1;7)
cases compared to -7/del(7q) MDS cases. While previous studies have described several
features of der(1;7), the understanding of der(1;7)+ MDS/AML is largely limited due
to their small study size.
Aims: To elucidate the clinical and genetic characteristics of der(1;7) with integrated
omics analysis in a large cohort of myeloid neoplasms.
Methods: We enrolled 148 cases with der(1;7) and analyzed their mutation profiles
and clinical features using whole exome and/or targeted-capture sequencing of major
driver genes of myeloid neoplasms and RNA sequencing. Distinct features of der(1;7)
were investigated by comparing the results with those from an additional 3,238 non-der(1;7)
cases with different myeloid neoplasms comprising -7/del(7q), +1q, and other cases.
Results: 148 der(1;7) cases comprised 72.3% MDS, 23.7% AML, and 3.4% MDS/MPN cases.
Compared to -7/del(7q) AML cases, der(1;7) AML cases were more likely to be AML with
dysplasia or MDS-derived AML and therapy-related AML (84.4% vs 53.2% and 9.1% vs 1.9%,
respectively) (p<0.001). Targeted-capture sequencing of der(1;7) cases revealed that
91.9% of cases harbored at least one copy number change or mutation. The frequency
of genetic mutation and copy number alteration for der(1;7) cases substantially differed
compared to -7/del(7q), +1q, and other cases. der(1;7) cases had frequent mutations
affecting RUNX1 (37.8%), EZH2 (18.9%) and ETNK1 (18.2%), which were less common in
-7/del(7q) cases (OR=5.58, OR=6.52, and OR=6.23, respectively). By contrast, TP53
mutations were less frequent in der(1;7) cases (OR=0.021). Copy number analysis revealed
a frequent co-occurrence of +8 and del(20q) with der(1;7) cases (18.9% and 37.4%),
while del(5q), which frequently co-occurred with del(7q), was not found in der(1;7)
cases. Prognosis of der(1;7) MDS cases tended to be better than -7/del(7q) cases (HR=0.71,
p=0.098), but poorer than +1q (HR=1.36, p=0.11) and others cases (HR=1.8, p<0.001).
The most frequent cause of death in der(1;7) cases was infection (45.5%), followed
by disease progression (36.4%). This high frequency of infection as a cause of death
was unique in der(1;7) as compared to -7/del(7q) (infection 13.9% and disease progression
72.3%) and others (infection 10.8% and disease progression 76.9%). Finally, RNA-sequencing
analysis between der(1;7) MDS cases and non-der(1;7) MDS cases showed unique expression
profiles in der(1;7). 1,463 differentially expressed genes were identified, of which
895 were down-regulated and 573 were up-regulated. GSEA analysis of major pathways
showed down-regulation of those related to cell cycles (including E2F targets), innate
immunity, and TNFα signaling via NFκB. Gene ontology analysis also revealed down-regulation
of cell cycle-related pathways, further supporting the significance of the down-regulated
cell cycles in der(1;7) MDS cases, compared to non-der(1;7) cases.
Summary/Conclusion: der(1;7)-positive MDS and AML represent a distinct subtype of
myeloid neoplasms, characterized by unique patterns of co-mutations/copy-number lesions
and gene expression profiles showing down-regulation of the cell-cycle pathway.
P745: ASSESSMENT OF MIRNA BIOGENESIS GENES VARIANTS IN MDS DEVELOPMENT AND PROGRESSION
D. Bug1,*, A. Tishkov1, I. Moiseev2, Y. Porozov3,4, N. Petukhova1
1Bioinformatics Research Center; 2R.M. Gorbacheva Scientific Research Institute of
Pediatric Hematology and Transplantation, Pavlov University, St Petersburg; 3World-Class
Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First
Moscow State Medical University, Moscow; 4Department of Computational Biology, Sirius
University of Science and Technology, Sochi, Russia
Background: Experimental studies demonstrated that knockout of miRNA processing enzymes
Dicer and Drosha is associated with the development of phenotype similar to myelodysplastic
syndrome (MDS). DICER1 inherited syndrome is associated with cancer predisposition.
There is a limited number of clinical studies evaluating the spectrum of mutations
in the microRNA processing genes in patients with hematological malignancies and the
pathogenic significance of these mutations is unknown. Thus, we conducted a bioinformatics
analysis of sequencing data from our data set of MDS patients and available results
in the public databases.
Aims: We aim to identify DICER1 and DROSHA variants in hematological malignancies
that lead to the protein function alteration based on bioinformatics analysis.
Methods: Data on 35 previously sequenced high-risk MDS patients (Moiseev IS et al.,
PLoS One. 2021 Mar 17;16(3):e0248430) was analyzed, and the list of DICER1 and DROSHA
variants was enriched with disease-specific coding variants retrieved from COSMIC
database, and 3’-UTR-variants - from dbSNP. All collected variants were subjected
to bioinformatics analysis. We used 3D-structures of Drosha and Dicer for molecular
dynamics (MD) simulations for 200 and 300 ns, respectively (Schrödinger Suite 2020-4,
Schrödinger, LLC, New York, NY, USA, 2020). The IntaRNA and MicroSNiPer algorithms
were used to calculate interaction energies of 3’-UTRs hybridizing with the most abundant
miRNAs in MSCs to evaluate noncoding variants.
Results: The prevalence of mutations in DICER1 and DROSHA was 54% and 17%, respectively,
predominantly located in 5’-UTR and 3’-UTR regions. Detected allele frequencies and
prevalence suggested their association with minor clones within the bone marrow niche.
MD simulation demonstrated apparent structural alterations with consequent protein
dysfunction. RMSD results and calculated total energy of studied proteins were higher
for all mutants compared to wild-type Drosha (at least 30% RMSD shift). All mutations
were shown to be located in close proximity of the miRNA binding site, all were destabilizing
for protein structure alone with consequent alterations in bonds and protein-miRNA
binding. The most significant structural shift and protein destabilization was detected
for DICER1 variants F508C and T993R characterized by local transformation of bonds
interplay. The conformational changes in the protein due to Dicer variants were studied
in 300 ns: differences in average RMSD values were within ~ 1Å between WT protein
and mutants. The most significant changes were observed at the local area for I445S,
F508C, and T993R characterized by shift of molecular bonds and alteration of protein
stability parameters.
A list of SNPs that exhibit the highest repression of DICER1 and DROSHA was obtained
(Table 1). The most prominent SNPs (rs1479981622, rs536092006, rs1296755923, rs1207839989)
had less than 0.03% population allele frequency which is in concordance with their
possible pathogenicity.
Image:
Summary/Conclusion: We identified potential coding and non-coding “hot spots” for
future analysis. Also the study shows that 3’-UTR SNPs of DICER1, DROSHA have more
potential to impact MDS progression than coding variants. Simultaneously, we found
low frequency of reports for these mutations in the available databases, indicating
bias and absence of these genes in the targeted sequencing panels. This builds the
foundation for future research of DICER1 and DROSHA clinical role in hematological
malignancies.
P746: COMBINATION OF RAS/MAPK MODULATOR AND AZACITIDINE AFFECTS HISTONE MARKS AND
IMPACTS THE INNATE IMMUNE SIGNALING PATHWAY IN THE MDS-L CELL LINE
R. Rai1,*, F. Patel1, J. Feld1, S. Melana1, S. Navada1, R. Odchimar-Reissig1, E. Demakos1,
E. P. Reddy2, A. Horowitz2, L. Silverman1
1Division of Hematology and Medical Oncology; 2Department of Oncological Sciences,
Icahn School of Medicine at Mount Sinai, New York, United States of America
Background: Myelodysplastic syndrome (MDS) is a stem cell disorder characterized by
ineffective hematopoiesis, and a propensity to transform into acute myeloid leukemia.
MDS patients exhibit chronic activation of the innate immune response and a hyperinflammatory
microenvironment (Barryero L, et al. Blood, 2018). Azacitidine (Aza), as a single
agent has an overall response rate (ORR) of only 50% (Silverman LR, et al., JCO 2002;
Fenaux P et al., Lancet Oncol. 2009). The response to Aza is not durable and all patients
relapse with worsening bone marrow failure. Aza combined with a multi-kinase inhibitor
of RAS and MAPK pathways, rigosertib (RAS/MAPK), reverses the bone marrow failure
state and demonstrates an ORR of 54% in patients who failed a prior HMA based therapy.
(Navada SC, et al. EHA Library 2019). This represents a critical observation in overcoming
the epigenetic clinical resistance phenotype, though the mechanism is still elusive.
Aberrant RAS/MAPK signaling has also contributed to MDS pathophysiology.
Aims: To investigated the impact of RAS modulation combined with Aza on the innate
immune signaling pathway.
Methods: We used the MDS-L cell line as a model to limit the heterogeneity observed
in MDS patients. The cells were treated with, Aza, RAS/MAPK and RAS/MAPK-Aza for 48
hrs and further analyzed by qPCR and western blot. We studied the expression of several
histone proteins and various pattern recognition receptors (TLRs, RIG-I, MDA5, STING),
their intermediate adaptor molecules myeloid differentiation factor 88 (MYD88), mitochondrial
antiviral signaling (MAVS) gene; and interferon regulatory factor (IRF)-3 and -7 by
qPCR.
Results: We found an increase in H3K9ac protein expression with RAS/MAPK and RAS/MAPK-Aza;
Aza and RAS/MAPK alone each had similar effects on H3K4me3, however, its expression
was markedly upregulated with RAS/MAPK-Aza. Effects on H3K36me3 were comparable in
all treated cells. We observed marked effects on the repression marks H3K9me3 and
H3K27me3 by RAS/MAPK and RAS/MAPK-Aza combination. Furthermore, we observed that Aza,
RAS/MAPK and RAS/MAPK-Aza significantly inhibit the expression of TLR1, 2 and 6. However,
TLR-3, 9, RIG-I, MDA5, STING, MAVS, MYD88, and IRF-3, 7 were significantly inhibited
by RAS/MAPK and RAS/MAPK-Aza.
Summary/Conclusion: We find that the RAS/MAPK modulator rigosertib has histone modifying
effects alone and when combined with Aza. The combination has effects on histone modification
of both activator and repressor marks and is associated with down-regulation of the
innate immune signaling pathway. In the clinic RAS/MAPK modulation combined with Aza
reverses bone marrow failure in MDS. Further studies are underway to determine the
correlation of the histone modification and innate immune signaling changes, as well
as the role of RAS/MAPK modulation, to determine how these mechanisms contribute to
the improvement in hematopoiesis in MDS patients.
P747: LR-MDS IS CHARACTERIZED BY DIFFERENTIAL EXPRESSION OF INFLAMMASOME-RELATED GENES
IN SPECIFIC CELL POPULATIONS
C. Rolfs1,*, M. Schneider1, M. Trumpp1, L. Fischer2, S. Winter2, S. Uxa1, V. Menger1,
K. Nenoff1, A. S. Kubasch1, K. Metzeler1, K. Sockel2, C. Thiede2, L. Hofbauer3, A.
Brüderle4, J. Woo5, M. Cross1, U. Platzbecker1
1Department of Hematology and Cell Therapy, University Hospital Leipzig, Germany,
Leipzig; 2Medical Clinic and Policlinic I; 3Department of Medicine III, University
Hospital Carl Gustav Carus, Dresden, Germany; 4Novartis Oncology; 5Translational Clinical
Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
Background: Myelodysplastic Syndromes (MDS) are a group of hematopoietic neoplasms
characterized by clonal expansion of hematopoietic stem and progenitor cells (HSPC)
and peripheral blood cytopenia. MDS can develop from clonal hematopoiesis of indeterminate
potential (CHIP) and progress through low-risk (LR) to high-risk (HR)-MDS and further
to acute myeloid leukemia (AML). Recent findings emphasize the role of sterile inflammation
in the bone marrow (BM) as a driver of neoplastic progression. Our previous analysis
of whole BM mononuclear cells (BM-MNC) has revealed distinct expression patterns of
inflammasome-related genes, including IL1B and IL18, to be associated with LR-MDS
subgroups and clinical features. This raises the prospect of interrupting disease
evolution at an early stage by targeted, personalized anti-inflammatory therapy. The
development and stratification of such therapies will ultimately require more detailed
knowledge of the variety and consequences of inflammation networks in MDS bone marrow.
Aims: Our objective was to assess inflammasome-related gene expression levels in individual
cell types in the BM of CHIP, LR-MDS and HR-MDS individuals as well as non-CHIP controls
in order to determine a) which cell types contribute to the expression of inflammasome-related
genes and b) whether specific disease states are associated with consistent patterns
of cell-specific gene expression.
Methods: Cryopreserved BM-MNC from 3 non-CHIP controls, 3 CHIP individuals, 14 LR-MDS
and 5 HR-MDS patients were obtained from the MDS registry and the BoHemE Study (NCT02867085)
at the University Hospitals in Dresden and Leipzig. Hematopoietic stem and progenitor
cells (HSPC), monocytes, monocytic myeloid-derived suppressor cells (M-MDSC), polymorphonuclear
MDSC (PMN-MDSC), B lymphocytes, T lymphocytes and CD45- cells (control) were sorted
from thawed BM-MNC on a BD FACS Jazz. RNA was isolated and the gene expression of
IL1B, IL18, S100A9, IRAK4, NLRP3, PYCARD, CASP1 and NLRC4 was assessed by RT-qPCR.
Results: The majority of the inflammasome-related genes, including IL1B, NLRP3, NLRC4
and S100A9 were expressed in all disease states predominantly by monocytes and M-MDSC
(p<0.001). However, IL18 was expressed at the highest level in HSPC. Consistent to
our previous analysis of whole BM-MNC, we found an association between cell-specific
gene expression and LR-MDS genetics, with HSPC-derived IL18 mRNA being highest in
SF3B1-mutated LR-MDS, while monocytic IL1B mRNA was highest in del(5q) LR-MDS. Furthermore,
an apparent progression in expression from non-CHIP through to LR-MDS was detected,
with mRNA levels of IL1B and NLRP3 in monocytic cells and PYCARD and CASP1 in HSPC
increasing from non-CHIP to CHIP between 1.5- and 7.9-fold. From CHIP to LR-MDS, mRNA
levels of CASP1 and NLRC4 in monocytic cells and PYCARD both in HPSC and in monocytic
cells increased by 1.8- to 2.1-fold.
Summary/Conclusion: We identify features of inflammasome-related gene expression in
individual cell populations and disease states reflecting the progression from non-CHIP
through CHIP to MDS. Our analysis suggests strongly that high resolution studies of
gene expression in specific populations or single cells will resolve a spectrum of
MDS-related inflammation states relevant to prognosis and personalized therapy. Therefore,
we are currently performing RNA-Seq from sorted cell populations in order to extend
our analysis beyond the core inflammasome reported here.
P748: SINGLE CELL GENOTYPING OF MATCHED BONE MARROW AND PERIPHERAL BLOOD CELLS IN
TREATMENT NAIVE AND AZA-TREATED MDS AND CMML
A. Schnegg-Kaufmann1,2,*, J. Thoms2, G. S. Bhuyan3, H. Henry2, L. Vaughan2, K. Rutherford4,
P. Kakadia5, E. Johansson6, T. Failes7, A. Greg7, J. Koval8, R. Lindeman9, P. Warburton10,
A. Rodriguez-Meira11, A. Mead11, A. Unnikrishnan3, S. Bohlander12, E. Pappaemmanuil13,
O. Faridani2, C. Jolly2, F. Zanini3, J. Pimanda14
1Department of Haematology and Central Haematology Laboratory, Inselspital, Bern,
Switzerland; 2School of Medical Sciences, Lowy Cancer Research Centre; 3Prince of
Wales Clinical School, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia;
4Computational Oncology Service, Department of Epidemiology & Biostatistics, Center
for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, United
States of America; 5University of Auckland, Auckland, New Zealand; 6Flow Cytometry
Facility, Mark Wainwright Analytical Centre; 7Children’s Cancer Institute, Lowy Cancer
Research Centre, UNSW Sydney, Sydney; 8Ramaciotti Centre for Genomics, UNSW Syndey,
UNSW; 9Department of Haematology, Prince of Wales Hospital, Sydney; 10Department of
Haematology, Wollongong Hospital, Wollongong, Australia; 11Haematopoietic Stem Cell
Biology Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford,
Oxford, United Kingdom; 12Leukaemia and Blood Cancer Research Unit, Department of
Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; 13Computational
Oncology Service, Department of Epidemiology & Biostatistics, Center for Computational
Oncology, Memorial Sloan Kettering Cancer Center, New York, United States of America;
14School of Medical Sciences, Lowy Cancer Research Centre, UNSW, Sydney, Sydney, Australia
Background: Somatic mutations in hematopoietic stem cells (HSCs) are a central pathogenic
event in myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML)
where they induce proliferative advantages and impaired differentiation with subsequent
cytopenias in peripheral blood (PB). Patients with high-risk disease who are ineligible
for allogenic stem cell transplantation are treated with hypomethylating agents, including
5-azacytidine (AZA). AZA treatment can improve PB counts and delay progression to
Akute Myeloid Leukemia. However, AZA response does not require eradication of mutated
HSCs and may be related to improved differentiation capacity of mutated hematopoietic
stem and progenitor cells (HSPCs). However, the contribution of mutated HSPCs to steadystate
hematopoiesis in MDS and CMML is unclear.
Aims: In this project, we aimed to determine the distribution of individual subclones
and even wild-type cells within the HSPC-compartment and the contribution of these
subclones and wild-type cells to effective hematopoiesis in patients with MDS or CMML.
Methods: We used a combination of index sorting and single cell genotyping to characterize
the somatic mutations of individual stem- and progenitor cells and matched high-turnover
circulating cells in three patients, one treatment naïve and two AZA-treated. Using
this approach, myeloid driver mutations previously detected in a traditional approach
were tracked in HSC/multipotent progenitors (MPP), MDS stem cells MDS-SC, progenitor
cells (common myeloid progenitor (CMP), granulocyte monocyte progenitor (GMP), megakaryocyte
erythroid progenitor (MEP)), as well as in monocytes, neutrophils and naïve B cells
(nBC).
Results: Patient H198302 and Patient H198303 both had CMML and had been treated with
AZA for ~10 years with complete response. Patient H198304 had MDS-EB1 and has never
been treated with hypomethylating agents. For each patient, the figure shows the proportions
of cells across the haematopoietic hierarchy carrying zero, one, two, three, or four
mutations in the specified alleles. Only very small numbers of HSPCs carrying no mutation
could be detected. In all three patients, most stem and progenitor cells carried a
majority of tracked mutations, with no major differences between stem cells, progenitor
cells and MDS-stem cells. In one patient, H198304, MDS-stem cells had a higher proportion
of cells with 3 or 4 mutations detected. Overall, in all three patients, a similar
frequency of mutations was observed in differentiated monocytes and neutrophils with
a substantial proportion of the circulating cells derived from highly mutated progenitors.
One notable exception were nBCs in Patient H198304, which were mostly wild-type, suggesting
that the small wildtype HSC population is the predominant origin of nBCs in this individual.
Analysis of additional lymphoid populations in PB from this patient revealed that
naïve T, but not NK cells were also predominantly wild-type, suggesting specific impairment
of B- and T-lineage maturation in the mutated cells.
Image:
Summary/Conclusion: In conclusion, attrition of highly mutated cells during myeloid
maturation was not observed in any of the three patients, irrespective of HMA therapy.
This suggests that in vivo, highly mutated stem and progenitor cells retain the capacity
to differentiate to mature myeloid and in some patients mature lymphoid cells and
contribute significantly to circulating blood cells in MDS and CMML, prior to and
following AZA treatment.
P749: PROGNOSTIC SIGNIFICANCE OF SERIAL CIRCULATING TUMOR DNA STATUS POST TREATMENT
IN MYELODYSPLASTIC SYNDROMES AND ACUTE MYELOID LEUKEMIA
H. Tong1,*, X. Zhou1, W. Lang1, C. Mei1, Y. Ren1, L. Ma1, G. Xu1, L. Xu1, Y. Li1
1department of hematology, The first affiliated hospital Zhejiang university school
of medicine, Hangzhou, China
Background: A major advance in our understanding of Myelodysplastic Syndromes (MDS)
and Acute Myeloid Leukemia (AML) biology has been the implementation of next generation
sequencing (NGS) which influenced diagnostic, prognostic, and therapeutic decisions
in myeloid malignancies. Circulating tumor DNA (ctDNA) sequencing, as as a novel and
minimally invasive measure, was reported to exhibit excellent correlations with matched
bone marrow NGS in MDS and AML. However, the clinical relevance of dynamic ctDNA monitor
during active treatment is unknown.
Aims: In this study, we assessed the role of ctDNA as a biomarker to monitor therapeutic
response and clonal evolution in MDS and AML.
Methods: Thirty-one MDS and AML patients who had both bone marrow NGS and ctDNA at
baseline were included for concordance analysis. Twenty-seven patients who had at
least two serial ctDNA assessment with at least one month interval were included for
dynamic ctDNA analysis. Targeted deep sequencing was performed on bone marrow and
ctDNA using a customized panel of 165 genes known to be recurrently mutated in MDS
and acute myeloid leukemia.
Results: Thirty out thirty-one patients were identified with ctDNA mutation at baseline.
Diagnostic ctDNA and matched bone marrow DNA exhibited excellent correlations with
variant allele frequencies (VAFs) (R2=0.721, p<0.001). For 27 patients who had ctDNA
mutation in baseline and treated with HMA or chemotherapy, the average mutation VAFs
at the end of treatment was lower in patients achieving a CR/CRi versus those with
mCR, mLFS or SD (2.7% vs 24.1%, respectively, p<0.001). As expected, mutation VAFs
from all patients with treatment failure did not decrease during treatment. (Figure
1). Similarly, the average decrease in mutation VAFs from pretreatment to end of treatment
was greater in patients achieving a CR versus those who did not (87.7% vs 35.5%, respectively,
p= 0.001). ctDNA negative post treatment was associated with longer PFS (median PFS
not reached (NR) vs. 5.6 (95% CI 4.08-7.13) months; P=0.009)(Fig 2A) and OS (median
OS NR vs. 12.0 (95% CI 9.08-15.0) months; P=0.023) (Fig 2B).
Seven of ten MDS patients who transformed to AML were identified with lately acquired
subclones harboring FLT3 or NF1, mutations involving in signaling pathway. Moreover,
new subclones were detected 0.5 to 4 months prior to AML transformation in 4 cases,
indicating that genetic progression can predate morphological progression.
Image:
Summary/Conclusion: ctDNA status post treatment was relevant to clinical response
and was of prognostic significance for disease progress and relapse. Dynamic ctDNA
changes revealed complex patterns of clonal structure of MDS and AML in response to
treatment. ctDNA sequencing could be an attractive, prospective measure for disease
monitoring.
P750: CLONAL HEMATOPOIESIS AND EPIGENETIC AGE ACCELERATION IN ELDERLY DANISH TWINS
M. Tulstrup1,2,3,4,*, M. Soerensen2,5, J. W. Hansen1,3,4, J. Weischenfeldt3,4,6, K.
Grønbæk1,3,4, K. Christensen2,5,7
1Department of Hematology, Rigshospitalet, Copenhagen; 2The Danish Twin Registry,
University of Southern Denmark, Odense; 3Biotech Research and Innovation Centre; 4The
Danish Stem Cell Centre (DanStem), University of Copenhagen, Copenhagen; 5Department
of Clinical Genetics, Odense University Hospital, Odense; 6Finsen Laboratory, Rigshospitalet,
Copenhagen; 7Department of Clinical Biochemistry and Pharmacology, Odense University
Hospital, Odense, Denmark
Background: Clonal hematopoiesis of indeterminate potential (CHIP) is commonly ocurring
in the elderly and associated with increased morbidity and mortality. CHIP is associated
with accelerated epigenetic age (Robertson et al. Curr Biol 29, R786–R787, 2019; Nachun
et al., Aging Cell, 20, e13366, 2021) and especially with intrinsic measures of epigenetic
age, i.e. measures which reflect cell-type independent effects of aging on DNA methylation,
as opposed to extrinsic measures that reflect age-related changes to the immune-cell
composition of the peripheral blood. Furthermore, Nachun et al. reported that the
combined measure of Hannum and GrimAge acceleration modifies the effect of CHIP mutation
status, resulting in strongly increased risk of all-cause mortality in individuals
with both CHIP and accelerated Hannum age and GrimAge age.
Aims: To replicate and extend these findings in elderly Danish twins, especially investigate
gene-specific effects on extrinsic vs. intrinsic estimators and the effect of shared
environmental and genetic factors in intra-twin pair analyses.
Methods: Blood samples collected from elderly Danish twins (mean age 79 years, range
73–90) in 1997 were analyzed for CHIP mutations using a custom Illumina TruSeq next-generation
sequencing panel targeting 21 og the most commonly mutated genes in CHIP. DNA methylation
profiling was performed on the Illumina HumanMethylation450K BeadChip. Associations
between CHIP and epigenetic age were investigated using linear regression (individual-level
analyses) and conditional logistic regression (intra-pair analyses). The interaction
between CHIP and epigenetic age acceleration was analysed using Cox proportional hazards
regression.
Results: Of 308 individuals (154 twin pairs), 116 carried a CHIP mutation. The strongest
association with CHIP in individual-level analyses was seen for the Intrinsic Epigenetic
Age Acceleration (IEAA) estimator (CHIP carriers 1.40 years older [95% CI: -0.01–2.81]),
and in intra-pair analyses Hannum age showed the strongest association (OR 1.1 [1.01–1.19]).
In mutation-specific analyses, TET2 mutations were associated with the extrinsic Hannum
age estimator in both individual-level (3.12 years [1.07–5.16]) and intra-pair analyses
(OR 1.11 [0.99–1.24]). DNMT3A mutations were associated with IEAA in individual-level
(1.82 years, [0.07–3.57]) but not intra-pair analysis (OR 1.03, 0.92–1.14). Analyses
of logit-transformed VAF were consistent with these results. Individuals with both
CHIP and accelerated Hannum and GrimAge estimates did not have an increased mortality
risk in our cohort (HR 1.02, [0.70–1.46]).
Summary/Conclusion:
TET2 mutations were predominantly associated with extrinsic epigenetic age acceleration
and the effect remained when controlling for shared factors between co-twins. Conversely,
DNMT3A mutations were associated with intrinsic epigenetic age acceleration, and the
intra-pair analysis indicated that this association may be due to a shared genetic
and/or environmental background. The association between TET2 mutations and extrinsic
epigenetic aging indicates that TET2 mutated clones may be drivers of immunosenescence,
while the association between DNMT3A mutations and intrinsic epigenetic aging may
be due to increased stem cell proliferation rates in individuals with DNMT3A mutations.
The CHIP-age acceleration interaction reported by Nachun et al was not replicated,
indicating that this mortality model should be used with caution, especially in this
age group.
P751: CHARACTERIZING CIRCULAR RNA EXPRESSION IN MYELODYSPLASTIC SYNDROME
E. Wedge1,2,3,*, C. R. M. Côme2,3, J. W. Hansen1,2,3, J. S. Jespersen2,3,4, M. Dahl1,2,3,
C. Schöllkopf1, K. Raaschou-Jensen5, B. Porse2,3,4, J. Weischenfeldt2,3,4, L. S. Kristensen6,
K. Grønbæk1,2,3
1Department of Hematology, Rigshospitalet; 2Biotech Research and Innovation Center
(BRIC); 3Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Faculty of
Health and Medical Sciences, University of Copenhagen; 4The Finsen Laboratory, Rigshospitalet,
Copenhagen; 5Department of Hematology, Odense University Hospital, Odense; 6Department
of Biomedicine, Aarhus University, Aarhus, Denmark
Background: Circular RNA (circRNA) research is an expanding field, reflecting a recognition
that circRNAs may have various roles in cancer pathology. CircRNAs are formed when
back-splicing events create covalently closed loop structures from pre-mRNA molecules
(Kristensen LS, Nat Rev Genet, 2019). CircRNA expression is cell-type specific, also
during hematopoietic differentiation (Nicolet BP, Nucleic Acids Res, 2018), but their
biological significance in myeloid cancers is still largely unknown.
Aims: We aimed to profile circRNA expression in myelodysplastic syndrome (MDS), along
with the related diseases chronic myelomonocytic leukemia (CMML) and clonal cytopenia
of undetermined significance (CCUS), in order to identify disease-specific expression
patterns and relevant candidates for biological or biomarker exploration.
Methods: Seventy-one patients and eight healthy age-matched controls were included.
Bone marrow aspirate underwent FACS sorting to obtain CD34+ stem and progenitor cells.
RNA was extracted and total RNA libraries were created using enzymatic ribosomal RNA
removal. The RNA was sequenced on a NovaSeq 6000 (mean 270 million paired-end reads
per sample, read length 150 bp). Bioinformatic pipelines find_circ and CIRI2 were
used to identify and quantify reads originating from the back-splicing junctions of
circRNAs. Detection by both pipelines was required, plus presence in at least 10%
of the samples. DESeq2 was used for normalization and comparison of groups. CircRNAs
with a mean expression of >10 normalized reads were considered highly abundant and
this subset was the focus of analysis.
Results: In total, 8,651 unique circRNAs passed the filtering criteria, and the top
826 were highly abundant. Global circRNA expression showed upregulation in CCUS relative
to healthy controls (p<0.001) and MDS relative to CCUS (p<0.001) (Figure A). We found
that 110 unique circRNAs were significantly upregulated in MDS relative to healthy
controls, whilst none were significantly downregulated (Figure B). In addition, high
or very high IPSS-R score was associated with significantly higher circRNA expression
than lower scores (p<0.001). We used LASSO regression to identify 14 circRNAs which
may be related to clinical outcomes, and used these to calculate a Myeloid Circ Score
(MCS). Patients were designated ‘high MCS’ or ‘low MCS’ with a cut-off at the median.
A high MCS was associated with poorer progression-free survival (PFS) with a hazard
ratio (HR) of 33.5 (95%CI 7.8-143.5, p<0.001) and poorer overall survival (OS) with
a HR of 16.5 (95%CI 3.8-71.5, p<0.001) in the whole cohort. The same could be seen
in analysis of MDS patients only (PFS HR 14.2 (95%CI 3.2-62.7, p<0.001), OS HR 8.5
(95%CI 1.9-38.3, p=0.005)). This a greater hazard ratio than that of the IPSS-R in
this cohort (Very high/high/Intermediate IPSS-R relative to low/very low IPSS-R; HR
for PFS of 4.27 (95%CI 1.7-11.1, p=0.003), and HR for OS of 2.5 (95%CI 0.9-7.3, p=0.09)).
Image:
Summary/Conclusion: We have observed associations between overall circRNA abundance
and disease severity in myeloid cancer, including a global upregulation along the
spectrum of disease from healthy to CCUS to MDS, and upregulation in high risk MDS
compared to lower risk MDS. CircRNAs may be directly implicated in disease biology
or be reflective of other changes at the cellular level. The Myeloid Circ Score has
potential in risk stratification but requires validation in independent cohorts and
further investigation in more readily available tissues such as bulk bone marrow or
peripheral blood.
P752: TARGETING S100A9 IN THE MYELODYSPLASTIC INFLAMMATORY BONE MARROW MICROENVIRONMENT
BY TASQUINIMOD IMPROVES THE SUPPORTIVE FUNCTION OF MESENCHYMAL STROMAL CELLS
M. Wobus1,2,*, E. Balaian1,2, M. Lissner1, R. Towers1, U. Oelschlägel1, R. Wehner2,3,
T. Chavakis4, M. Törngren5, H. Eriksson5, E. Bondesson5, U. Platzbecker6, M. Bornhäuser1,2,
K. Sockel1
1Department of Medicine 1, University Hospital TU Dresden, Dresden; 2German Cancer
Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ),
Heidelberg; 3Institute of Immunology; 4Institute of Clinical Chemistry and Laboratory
Medicine, University Hospital TU Dresden, Dresden, Germany; 5Active Biotech AB, Lund,
Sweden; 6Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, Leipzig
University Hospital, Leipzig, Germany
Background: Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis,
peripheral cytopenia and the risk of transformation into acute myeloid leukemia. An
aberrant innate immune response and a proinflammatory bone marrow (BM) microenvironment
play a critical role in the pathogenesis of MDS. The alarmin S100A9, a key player
for regulation of inflammatory responses, has been shown to be elevated in MDS patients.
It directs an inflammatory cell death (pyroptosis) by increased NF-kB mediated transcription
and secretion of proinflammatory, hematopoiesis-inhibitory cytokines and production
of reactive oxygen species (ROS). Tasquinimod (TASQ, Active Biotech) is a novel, small
molecule inhibitor of S100A9 which has been investigated in several solid tumor entities
and is currently under investigation in a phase Ib/IIa trial of patients with relapsed/refractory
multiple myeloma. So far, little is known about its effects on myeloid malignancies.
Aims: We aimed to investigate the role of S100A9 in cellular models of MDS and the
in vitro potential of TASQ to target S100A9 within the MDS microenvironment.
Methods: Mesenchymal stromal cells (MSCs) from patients with either low-risk MDS,
CMML or age-adjusted healthy donors were exposed to S100A9 (1.5µg/ml) ± TASQ (10µM).
Subsequently, TLR4 downstream and proinflammatory signaling was analyzed by Western
blot and real-time PCR. ROS levels were determined with a CellROX Flow Cytometry Kit
(Thermo Fisher). Moreover, MSC differentiation and colony-forming unit-fibroblast
(CFU-F) capacity were tested. To study the impact on the hematopoietic support, MSCs
were pre-treated for one week with S100A9 ± TASQ before CD34+ hematopoietic stem and
progenitor cells (HSPCs) were seeded on the stromal layer. The colony formation (CAF-C)
was analyzed weekly followed by a CFU-GEMM assay in methylcellulose medium.
Results: Exposure of MDS and healthy MSCs to S100A9 induced TLR4 downstream signaling
as demonstrated by increased expression of IRAK1 and NF-kB-p65 as well as gasdermin,
an inductor of pyroptosis. Addition of TASQ abolished these effects and inhibited
the expression of the mentioned proteins, indicating an alleviation of inflammation.
In line with this, OXPHOS and ROS levels were decreased after addition of TASQ. Furthermore,
we detected a 2-fold increase of mRNA expression of the proinflammatory cytokines
IL-1β and IL-18 as well as a 5-fold increase of their activator caspase 1 in MSCs
after treatment with S100A9, which could be prevented by TASQ. Interestingly, PD-L1
as a potential downstream target was induced by S100A9 by 2.5-fold and could be suppressed
by TASQ to about 50% both at the mRNA and protein level. Moreover, addition of TASQ
to MDS MSC cultures resulted in a significantly higher CFU-F number as well as abolished
blocked the excessed adipogenic differentiation.
In co-cultures with HSPCs, we observed a decreased number of cobblestone area forming
cells (CAF-C) as well as reduced numbers of colonies (CFU) in a subsequent clonogenic
assay, indicating a disturbed hematopoietic support by S100A9 treated MSCs, which
could be restored by TASQ pre-treatment.
Summary/Conclusion: We provide evidence that TASQ mitigates the pathological inflammasome
activation in the myelodysplastic BM niche in vitro by inhibition of the S100A9-mediated
TLR4 signalling as well as its effects on NF-kB-p65 transcription and PD-L1 expression,
resulting in improved hematopoietic support by MSCs, which suggests a beneficial effect
in cytopenic MDS patients.
P753: DYSFUNCTIONAL BONE MARROW ENDOTHELIAL PROGENITOR CELLS ARE INVOLVED IN PATIENTS
WITH MYELODYSPLASTIC SYNDROMES
T. Xing1,2,*, Z.-S. Lyu1,2, C.-W. Duan3, H.-Y. Zhao1, S.-Q. Tang1, Q. Wen1, Y.-Y.
Zhang1, M. Lv1, Y. Wang1, L.-P. Xu1, X.-H. Zhang1, X.-J. Huang1,2, Y. Kong1
1Peking University Institute of Hematology, National Clinical Research Center for
Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation,
Collaborative Innovation Center of Hematology, Peking University People’s Hospital;
2Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary
Studies, Peking University, Beijing; 3Key Laboratory of Pediatric Hematology and Oncology
Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children’s
Medical Center, Shanghai Jiao Tong University School of medicine, Shanghai, China
Background: Myelodysplastic syndromes(MDS) are a group of heterogeneous myeloid clonal
disorders characterized by ineffective haematopoiesis and immune deregulation. Emerging
evidence has shown important roles of the bone marrow(BM) microenvironment in regulating
haematopoiesis and immune balance. As an important component of BM microenvironment,
murine studies and our previous studies(Blood2016, EbioMedicine2020, CEI2021) reported
endothelial progenitor cells(EPCs) modulate the physiology and regeneration of haematopoietic
stem cells(HSCs) and differentiation of effector T cell subsets. Some evidences suggest
that EPCs demonstrate inferior supporting ability to normal HSCs in MDS. However,
the dual supporting abilities of BM EPCs to normal HSCs and malignant cells in patients
with different stages of MDS remain unclear. In addition, the immunomodulatory ability
of BM EPCs in MDS needs to be elucidated.
Aims: To determine the number, the functions and the abilities of haematopoiesis and
immune regulation of BM EPCs in patients with different stages of MDS. Moreover, to
explore the correlation between dysfunctional BM EPCs and different stages of MDS
and underlying mechanisms of dysfunction.
Methods: In this study, patients with lower-risk MDS(N=15), higher-risk MDS(N=15)
and de novo acute myeloid leukaemia(AML)(N=15) and healthy donors(HDs)(N=15) were
enrolled. The number of primary BM EPCs were detected by flow cytometry. The functions
of BM EPCs were evaluated by DiI-Acetylated low-density lipoprotein uptake and FITC-UEA-1
binding assay, tube formation and migration assays. In order to assess the gene expression
profiles of BM EPCs, RNA sequencing and gene set enrichment analysis were performed.
The supporting abilities of BM EPCs from MDS patients to normal HSCs, leukaemia cells
and T cells were assessed by in vitro coculture experiments(coculture with HSCs and
T cells from HDs and HL-60 cells, respectively).
Results: Increased but dysfunctional BM EPCs were found in MDS patients compared with
HDs, especially in patients with higher-risk MDS. RNA-seq indicated the changes of
haematopoiesis- and immune-related pathways in MDS BM EPCs. In vitro coculture experiments
verified that BM EPCs from HD, lower-risk MDS, and higher-risk MDS to AML exhibited
a progressively decreased ability to support normal HSCs, manifested as elevated apoptosis
rates and intracellular reactive oxygen species(ROS) levels and decreased colony-forming
unit plating efficiencies of HSCs. Moreover, BM EPCs from higher-risk MDS patients
demonstrated an increased ability to support leukaemia cells, characterized by increased
proliferation, leukaemia colony-forming unit plating efficiencies, whereas decreased
apoptosis rates and apoptosis-related genes of HL-60 cells. Furthermore, BM EPCs induced
normal T cell differentiation towards more immune-tolerant cells, like regulatory
T cells, in higher-risk MDS patients in vitro. In addition, the levels of intracellular
ROS and the apoptosis ratio were increased in BM EPCs from MDS patients than HDs,
especially in higher-risk MDS patients, which may be underlying mechanisms of dysfunctional
BM EPCs.
Summary/Conclusion: The current study demonstrated that increased but dysfunctional
BM EPCs in MDS patients. Moreover, the dysfunctions of BM EPCs were more severe in
higher-risk than lower-risk MDS patients, manifested as more supportive ability to
leukaemia cells but worse ability to normal HSCs. Our data indicated that repair of
dysfunctional BM EPCs may be a promising therapeutic approach for MDS patients.
P754: INITIAL RESULTS OF PHASE I/II STUDY OF AZACITIDINE IN COMBINATION WITH QUIZARTINIB
FOR PATIENTS WITH MYELODYSPLASTIC SYNDROME AND MYELODYSPLASTIC/MYELOPROLIFERATIVE
NEOPLASM WITH FLT3 OR CBL MUTATION
T. Abuasab1,1,*, E. Jabbour1, N. Short1, M. Konopleva1, K. S. Chien1, S. Fareed Mohamed1,
N. Daver1, R. Kanagal-shamanna2, G. Garcia-Manero1, G. Montalban-Bravo1
1Department of Leukemia; 2Department of Hematopathology, The University of Texas,
MD Anderson Cancer Center, Houston, TX, USA, Houston, United States of America
Background: FMS-like tyrosine kinase 3 (FLT3) mutations occur in about 1% of patients
(pts) with newly diagnosed myelodysplastic syndrome (MDS) and up to 19% at time of
failure of hypomethylating agent (HMA). In addition, Casitas B-lineage Lymphoma mutations
(CBL) are observed in 10% of pts with MDS/MPN and up to 13-15% of pts with CMML (mainly
proliferative type). Preclinical studies suggest that CBL-mutant cells are dependent
on FLT3 signaling.
Aims: The primary objective is to determine the safety, tolerability, and maximum
tolerable dose (MTD) of quizartinib. Secondary objectives include evaluation of overall
response per IWG 2006 criteria, in addition to overall survival (OS), duration of
response, leukemia-free survival (LFS), relapse-free survival (RFS).
Methods: We designed a phase I/II study of azacitidine in combination with quizartinib
for pts with newly diagnosed or previously treated MDS or MDS/MPN with detectable
FLT3 and/or CBL mutations. The study included an initial phase I dose escalation portion
followed by a phase II dose expansion. Dose escalation was planned to evaluate 3 dose
levels of quizartinib (30, 40 and 60mg, respectively) administered on days 1-28 of
every cycle in combination with azacitidine (75 mg/m2/day) on days 1-5 each 28-day
cycle.
Results: As of February 25, 2022, a total of 10 pts have been enrolled (9 in dose
escalation, 1 in dose expansion): 7 pts with CMML, two with MDS with excess blast
and one with atypical chronic myeloid leukemia (aCML). A total of 8 pts (80%) had
normal karyotype, one ptn (10%) had complex karyotype and one (10%) had monosomy 7.
Six pts had FLT3 mutations and 4 pts CBL mutations. Pts characteristics are summarized
in Table 1. Nine out of 10 pts were evaluable at the time of the analysis. Median
number of cycles administered was 5 (range 2-10). All pts have achieved response so
far with 8 pts (89%) having achieved marrow CR (mCR), including hematological improvement
(HI) in 1 patient out of 3 pts with baseline cytopenias evaluable for HI. One patient
had hematologic improvement-erythropoietic (HI-E) with control of leukocytosis and
thrombocytosis to normal levels. During cycle 1, responses were associated with recovery
of platelets to above 50 and 100x109/L by day 28 of therapy in 67% and 56% of pts,
respectively. In addition, absolute neutrophils count (ANC) was recovered to 1 x109/L
by day 28 in all pts except of one patient at day 52.
With a median follow up of 9.87 months (range 2.5-18), the median response duration
is 6.3 months (range 1.6-14.3) with a median OS of 17.34 months (range 2.5 – 18).
A total of 4 pts (44%) have stopped the treatment: 1 patient due to disease relapse
and one due to disease progression after both received 6 cycles of treatment and two
proceeded to allogenic stem cell transplantation after achieving response. No dose-limiting
toxicities have been observed to date up to the maximum dose of 60 mg of quizartinib.
Two pts required dose reductions of Quizartinib, one due to cytopenias and the other
one due to concomitant posaconazole administration. Regarding the grade 3 or more
adverse events (AEs): 3 pts (33%) had severe anemia/thrombocytopenia, 2 pts (22%)
had skin infection, one patient (11%) had cardiac arrythmia with Mobitz II AV block
and one patient (11%).
Image:
Summary/Conclusion: Preliminary data suggest azacitidine in combination with quizartinib
for pts with MDS and MDS/MPN with FLT3 or CBL mutations have an acceptable toxicity
profile and associated with promising responses. Further follow-up with a larger patient
cohort are required to emphasize the safety and efficacy.
P755: HYPERFERRITINEMIA IS A PREDICTIVE BIOMARKER OF POOR CLINICAL OUTCOMES IN CMML
L. E. Aguirre1,*, S. Ball1, A. Jain1, N. Al Ali1, D. Sallman1, A. Kuykendall1, K.
Sweet1, J. Lancet1, E. Padron1, R. Komrokji1
1Malignant Hematology, Moffitt Cancer Center, Tampa, United States of America
Background: CMML is a heterogenous disease exhibiting features innate to MPN and MDS.
Increasing evidence supports a close interplay between systemic inflammation and risk
of myeloid malignancies, notably for those with history of infection or autoimmune
disease. CMML has been associated with inflammation and end-organ damage related to
CKD and CVD. Analysis of gene signatures from CMML-derived monocytes has shown them
to be highly proinflammatory. High ferritin may serve as a practical biomarker of
disease activity to help identify pts at higher risk of poor outcomes.
Aims: We aimed to identify whether hyperferritinemia correlated with worse outcomes
in CMML and to characterize relevant baseline clinical and molecular features associated
with this clinical phenotype.
Methods: Retrospective data was collected from a database of pts with CMML treated
at Moffitt Cancer Center. Pts were stratified in 2 cohorts based on ferritin levels
(<1000 or ≥1000 ng/mL). Hyperferritinemia was defined as ferritin >1000 as seen at
diagnosis or during follow-up. Kaplan–Meier was used to estimate OS. Cox regression
was used for multivariate analysis.
Results: Between August 1995 and October 2020 729 pts with CMML were identified. Median
age at diagnosis was 71 (17-95). Out of 571 pts with available ferritin levels 29%
(n=168) developed hyperferritinemia vs 71% (n=403) who did not. mOS was 32.4 mos (95%CI
30-35 mos).
Pts with higher ferritin tended to present with CMML-2 (p=0.001) and harbor a proliferative
phenotype (p=0.01). They presented with higher marrow cellularity (mean 83%, p =0.08),
PLT (mean 177k, p= 0.038), and lower Hb (mean 9.5, p<0.05). There was no association
with % circulating IMC, monocytes, WBC or ANC at baseline. Hyperferritinemia was associated
with more profound fibrosis (p=0.007), cytopenias (p<0.05), % peripheral blasts (p<0.05),
RBC and PLT transfusion dependence (p<0.05).
Pts with hyperferritinemia had higher risk disease per IPSS-R, CPSS and all CMML models
(p<0.05); and had higher rates of AML transformation (p<0.05). Pts were also more
likely to require treatment earlier (within 3 yrs of diagnosis) (p<0.05).
ASXL1 (p=0.002), EZH2 (p=0.003), and SETBP1 (p=0.019) mutations were more common among
pts who developed hyperferritinemia. Conversely, TET2 (p=0.001), CBL (p=0.028) and
SRSF2 (p=0.003) mutations were less common.
mOS for pts with hyperferritinemia was 23.9 mos (95%CI 19.9-27.9 mos), much lower
than for those with ferritin <1000 (mOS 40.5 mos, 95%CI 35.4-45.5 mos) (p<0.05). In
multivariate analysis, hyperferritinemia was a significant independent covariate for
OS after adjusting for CPSS, transfusion dependence and disease phenotype (dysplastic
vs proliferative) (HR= 0.69; 95%CI 0.53-0.89; p=0.005).
Summary/Conclusion: Almost 1/3 of pts with CMML will develop hyperferritinemia. This
is associated with more aggressive disease and rates of AML transformation leading
to dismal outcomes. ASXL1, EZH2, and SETBP1 MTs confer a higher risk of hyperferritinemia.
Our findings indicate that hyperferritinemia is an independent prognostic biomarker
that may serve as a surrogate representative of disease biology and comorbidities
in CMML.
P756: ALLOGENEIC STEM CELL TRANSPLANTATION IN MYELODYSPLASTIC SYNDROME/MYELOPROLIFERATIVE
NEOPLASM (MDS/MPN) OVERLAP SYNDROMES. A SINGLE CENTER EXPERIENCE AND DISSECTION OF
MUTATIONAL PROFILE
A. Avendaño Pita1,*, M. Martín Izquierdo1, S. Muntión Olave1, T. Jímenez Solas1, M.
García Antúnez1, L. Eva1, M. Del Rey1, S. Toribio Castelló1, A. Yeguas Bermejo1, T.
González1, J. M. Hernández-Rivas1, A. Á. Martín López1, M. Cabrero1, E. Pérez López1,
A. Cabero1, M. Baile1, L. Vázquez1, L. López Corral1, F. Sánchez-Guijo1, D. Caballero
Barrigon1, M. Cortés Rodríguez1, M. Díez Campelo1
1Hematology, University Hospital of Salamanca / IBSAL, Salamanca, Spain
Background: MDS/MPN (myelodysplastic syndrome/myeloproliferative neoplasm) overlap
syndromes are myeloid malignancies for which allogeneic hematopoietic stem cell transplant
(allo-HSCT) is potentially curative in the absence of disease-modifying therapies.
Aims: To describe clinical, biological and molecular characteristics at diagnosis
and transplant outcomes of our series trying to detect witch parameters may have impact
on survival.
Methods: We retrospectively reviewed 35 consecutive patients who underwent to allo-HSCT
from 1999 to 2021 as MDS/MPNS overlap syndrome. Descriptive statistics and survival
analysis were performed trough jamovi (Version 2.2.).
Results: Clinical, biological and molecular baseline characteristics are summarized
in image. Median age at allo-HSCT were 58 (27-70) with a median time from diagnosis
to allo-HSCT of 8 months (1-128). Twenty patients (57%) were referred in complete
remission (CR). Preferred graft source were peripheral blood in 34 patients (97.1%).
Regarding to conditioning regimen, 26 patients (74.3%) received reduced intensity,
7 patients (20%) received myeloablative and 2 patients (5.7%) sequential conditioning.
Eighteen patients (51.4%) had matched sibling donor, 8 patients (22.8%) had matched
unrelated donor, 3 patients (8.6%) mismatched unrelated donor and 6 (17.1%) haploidentical
donor. All patients reached neutrophil graft and 31 patients (88.6%) reach platelet
graft with a median time of 19 (12-29) and 14 (10-36) days respectively. On day +100,
29 patients (82.9%) were in CR, 4 patients (11.4%) did not reach response and 2 (5.7%)
were not evaluable. Twenty three patients (65.7%) developed acute graft versus host
disease (GVHD), in 19 cases (54.3%) grades II-IV, and 15 patients (42.9%) developed
chronic GVHD, in 8 cases (22.9%) moderate-severe grade.
Median follow-up were 13 months (2-153) with 17 months (2-153) for chronic myelomonocitic
leukemia (CMML) and 11.5 months (2-133) for atypical chronic. myeloid leukemia (aCML).
Overall survival (OS) at 1, 3, and 5 years were 69.7%, 54.9% and 42.5% respectively.
Sixteen patients (45.7%) relapsed with a median time of 7.5 months (0-85). Among relapsed
patients, 5 patients (31.3%) were not treated, 3 patients (19.8%) received donor lymphocyte
infusion, in 5 patients (31.3%) we administered hypometilating agents and salvage
chemotherapy in 2 (12.5%) remaining patients reaching second allo-HSCT in two cases.
Non-relapse mortality were 31.4%, in most cases due to infection. Transplant related
mortality (TRM) at day +100 and global TRM were 5.7% and 14.3% respectively. At last
follow-up, 13 patients (37.1%) remain alive, 11 (31.4%) in CR and 2 (5.7%) in relapsed
situation.
On the univariate analysis, harboring signal transduction mutation (p = 0.008), mutational
global burden by variant allelic frequency (VAF) at diagnosis greater than 30% (p
= 0.045) were asociated with worse outcomes. On the other hand, reaching CR at day
+100 (p <0.0001) and developing chronic GVHD (p = 0.0001) seems to improve survival.
If we look at the CMML subgroup, we found that new CMML transplant score significantly
stratifies outcomes (p = 0.006).
Image:
Summary/Conclusion: Once again, poor results are confirmed for this MDS/MPN overlap
syndromes with only a third of the patients being long term survivors. Molecular data
offered by NGS may help to refine prognostic scores and to stablish transplant referral.
Mutations in signaling pathways seems to confer poor prognosis as a late event in
disease course and provide more aggressive clinical behavior.
P757: RESULTS OF A PHASE 1 STUDY OF AZACITIDINE COMBINED WITH VENETOCLAX FOR TREATMENT-NAIVE
AND RELAPSED HIGH-RISK MYELODYSPLASTIC SYNDROME AND CHRONIC MYELOMONOCYTIC LEUKEMIA
A. Bazinet1,*, E. Jabbour1, H. Kantarjian1, K. Chien1, C. DiNardo1, M. Ohanian1, N.
Daver1, R. Kanagal-Shamanna2, T. Kadia1, K. Takahashi1, L. Masarova1, N. Short1, Y.
Alvarado1, P. Thompson1, G. Montalban-Bravo1, M. Yilmaz1, F. Ravandi1, M. Konopleva1,
M. Andreeff1, S. Kornblau1, N. Pemmaraju1, D. Hammond1, H. Schneider1, B. Mirabella1,
G. Garcia-Manero1
1Leukemia; 2Hematopathology, University of Texas MD Anderson Cancer Center, Houston,
United States of America
Background: Patients (pts) with higher-risk myelodysplastic syndromes (HR-MDS) who
are ineligible for allogeneic stem cell transplant (SCT) have limited treatment options
beyond the hypomethylating agents (HMAs) azacitidine (AZA) and decitabine (DAC). Responses
to HMA monotherapy are time-limited and prognosis after HMA-failure is dismal. Venetoclax
(VEN) is an oral small molecule BCL-2 inhibitor with the potential to prolong/deepen
responses when combined with an HMA.
Aims: To evaluate the safety/tolerability, recommended phase 2 dose (RP2D), and preliminary
efficacy of VEN in combination with AZA for the treatment of HR-MDS/CMML.
Methods: This was a single-center dose-escalation phase 1 study with “3 + 3” design.
Key inclusion criteria were a diagnosis of MDS or CMML by WHO 2016, age ≥ 18 yrs,
int-2 or high risk by IPSS, and bone marrow (BM) blasts of 5-19%. Pts could be treatment-naïve
or relapsed/refractory, defined as failure after 4+ cycles of HMA. Prior VEN exposure
was not permitted. Treatment consisted of AZA 75 mg/m2 on D1-5 and escalating doses
of VEN on D1-7 or 1-14 (Table 1) every 28D. VEN dosing was adjusted for pts on concomitant
CYP3A inhibitors. The primary objective was safety/tolerability and determination
of the RP2D. Secondary objectives were response rates, overall survival (OS), and
progression-free survival (PFS). Adverse events (AEs) were graded as per the CTCAE
v5.0 and responses assessed using the modified IWG 2006 criteria. All pts provided
informed consent. The study was registered on ClinicalTrials.gov (NCT04160052).
Results: Data cutoff was February 19th, 2022. 23 pts (13 HMA-naïve MDS, 4 HMA-naïve
CMML, 4 HMA-failure MDS, 2 HMA-failure CMML) were enrolled. The median age was 68
years (range 58-84) and median BM blasts 11% (range 6-19%). IPSS was int-2 in 78%
of pts and high in 22% of pts. 8/23 (35%) had complex cytogenetics, 6/23 (26%) had
TP53 mutations, and 5/23 (22%) had therapy-related MDS/CMML. A single dose-limiting
toxicity (DLT) occurred at dose level +2 (BM aplasia and delayed count recovery).
The maximum tolerated dose (MTD) was not reached. The most common grade 3/4 AEs were
neutropenia (39%), thrombocytopenia (39%), anemia (13%), and infections: pneumonia
(30%), neutropenic fever (17%), diverticulitis (9%), sepsis (9%), cellulitis (4%),
and splenic abscess (4%). The RP2D was established at AZA 75 mg/m2 on D1-5 plus VEN
400 mg on D1-14. 3 deaths occurred on study, all from sepsis. 30 and 60-day mortality
were 4% and 9%, respectively. Other reasons for discontinuing therapy included progression
of MDS/CMML (n=6), transformation to AML (n=4), SCT (n=4), and social reasons (n=1).
The ORR by intention-to-treat (ITT) analysis was 82% (14/17; 3 CR, 5 mCR+HI, 6 mCR)
in the HMA-naïve cohort and 100% (6/6; 6 mCR) in the HMA-failure cohort. A median
of 3 cycles (range 1-11) were given. Responses occurred after a median of 1 cycle
(range 1-2) and lasted a median of 4.5m (range 1.2m to 21.2m). At a median follow-up
of 13.2m, median OS was not reached (1-year OS 68.6%) in the HMA-naïve cohort and
8.3m (1-year OS 22%) in the HMA-failure cohort. Median PFS was 13.1m (1-year PFS 51.5%)
in the HMA-naïve cohort and 6.8m (1-year PFS 0%) in the HMA-failure cohort.
Image:
Summary/Conclusion: AZA combined with VEN is tolerable and yields a high ORR in HR-MDS/CMML,
with or without prior HMA. Rates of myelosuppression and infection are high and careful
monitoring is required. The RP2D is AZA 75 mg/m2 on D1-5 plus VEN 400 mg on D1-14,
every 28D. The phase 2 dose expansion component of this study is ongoing.
P758: INHIBITION OF ATR WITH CERALASERTIB (AZD6738) FOR THE TREATMENT OF PROGRESSIVE
OR RELAPSED MYELODYSPLASTIC SYNDROMES AND CHRONIC MYELOMONOCYTIC LEUKEMIA: A PHASE
IB/II STUDY
A. Brunner1,*, Y. Liu2, L. Mendez3, J. Garcia2, P. Amrein1, D. Neuberg2, E. Dean4,
S. Smith4, R. Stone2, A. Fathi1, M. Walter5, T. Graubert1, M. Jacoby5
1Massachusetts General Hospital; 2Dana-Farber Cancer Institute; 3Beth-Israel Deaconess
Medical Center, Boston, United States of America; 4AstraZeneca, Cambridge, United
Kingdom; 5Washington University School of Medicine, St. Louis, United States of America
Background: Myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML)
are enriched for mutations in the spliceosome complex, including the genes SF3B1,
SRSF2, ZRSR2, and U2AF1. Ceralasertib (AZD6738) is a selective and potent inhibitor
of Ataxia Telangiectasia and Rad3 Related (ATR) kinase. We have previously shown that
cells with splicing factor (SF) mutations have increased R loops, which require ATR
for resolution and preferentially undergo apoptosis after ATR inhibition.
Aims: We hypothesized that inhibition of ATR may be useful in patients with MDS or
CMML, particularly those who harbor SF gene mutations.
Methods: This 2-part study evaluated ceralasertib monotherapy in adult patients with
MDS or CMML progressing on or not responsive to front-line therapy (DNMTI for HR MDS,
ESA for LR MDS). Enrollment cohorts included SF mutant (SF3B1, SRSF2, U2AF1, or ZRSR2)
and SF wildtype (wt). Part 1 enrolled 3-6 patients at de-escalating dose levels: 160mg
BID days 1-14 of 28 d cycles, 120mg BID d1-14, and 80mg BID d1-14. DLTs were assessed
the first 30d and de-escalation would occur for DLTs in >1/6 patients. Higher-risk
MDS (HR-MDS, IPSS-R > 3.5) and CMML patients were enrolled first; after a dose was
deemed safe, lower-risk MDS (LR-MDS, IPSS-R 3.5, transfusion-dependent post/ineligible
for ESA or severe neutropenia/thrombocytopenia) was evaluated. Part 2 enrolled up
to 20 SF mut patients and 20 SF wt patients for efficacy. A Simon 2-stage design mandated
that at least 1/10 patients in each group needed to respond (by IWG 2006 criteria)
to enroll the final 10 patients.
Results: At the data cut-off (12/15/21) 32 patients have been enrolled with 30 evaluable:
22 with HR-MDS or CMML, and 8 with LR-MDS. The median age was 73 (range 43-88) and
predominantly male (26/30). 21 patients harbored SF mut disease while 9 had SF wt
MDS/CMML. The most frequently seen ≥grade 3 adverse events (AEs) for higher-risk patients
(n=20) included anemia (n=6), febrile neutropenia (n=4), neutropenia (n=5), thrombocytopenia
(n=6). In LR-MDS (n=8), ≥grade 3 AEs included thrombocytopenia (n=2), and neutropenia
(n=2). 5 deaths occurred within 30d of study treatment; 1 patient with active CMML
had an intracranial bleed with thrombocytopenia, possibly related to study treatment,
while 4 deaths were attributed to underlying disease (3 with progression to AML, 1
MDS patient with sepsis). No DLTs were observed in the HR or LR groups and 160mg BID
days 1-14 of 28 days was deemed the RP2D.
Responses were assessed across the cohort as well as according to SF mutant status
(Table). A total of 8 patients had an IWG response (BOR: CR, n=1; mCR, n=3; HI, n=4),
including 6/21 SF mut patients (ORR 29%), and 2/9 SF wt patients (ORR 22%). The responses
in SF wt patients included one mCR (5% to 1% marrow blasts in a patient with DNMT3A
and ETV6 mut and del(chr7)), and one HI-P (82 BL to 234 max in a patient with DNMT3A,
IDH2, ETV6 mut and del(chr7)). Responses in SF mut included CR (n=1), mCR (n=2, one
with associated HI-N), SD with HI (n=3). An additional 3 SF mut patients had improvements
in blood counts but not lasting 8 weeks (ANC n=2, platelets n=1) and one with platelet
improvement but a baseline not meeting IWG criteria (plt avg=108k prior to C1D1).
Image:
Summary/Conclusion: Ceralasertib is an oral ATR inhibitor with preliminary activity
in patients with progressive/refractory MDS and CMML at doses of 160mg BID on days
1-14 of a 28 day cycle. The study is ongoing and will evaluate additional dosing schedules.
P759: CORRELATION ANALYSIS OF DNA METHYLATION LEVEL AND CLINICAL CHARACTERISTICS IN
JMML PATIENTS
Y. Cai1,*, J. Zhang1, M. Yi1, X. Liu1, X. Zhang1, Y. Wan1, L. Chang1, L. Zhang1, X.
Chen1, Y. Guo1, Y. Zou1, Y. Chen1, Y. Zhang1, W. Yang1, X. Zhu1
1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences
& Peking Union Medical College, Tianjin, China
Background: As a rare, aggressive pediatric myeloproliferative disease, juvenile myelomonocytic
leukemia (JMML) encompassed both biological features of myelodysplastic syndrome and
myeloproliferative neoplasm. Studies have shown that the methylation level in JMML
patients is closely related to prognosis, and patients with high methylation level
have poor prognosis.
Aims: This study aimed to find clinical indicators that were associated with different
methylation levels and prognosis.
Methods: The clinical information of 24 JMML patients with DNA samples admitted to
our center from December 2013 to May 2020 was retrospectively analyzed, and the DNA
methylation level of their whole genome was detected.
Results: The median age of onset was 14.5 months (0.1-153 months) among the 24 cases,
including 17 males and 7 females. At diagnosis, the median WBC count was 27.1×109/L
(6.2-98.1×109/L), and the median platelet count was 38×109/L (10-277×109/L). Chromosome
karyotype abnormalities were found in 12.5% (3/24) of patients. Next-generation sequencing
results showed that 79.2% (19/24) patients had at least one Ras pathway-related classical
gene mutation, and 41.7% (10/24) patients had two or more somatic mutations. Genomic
DNA methylation levels were divided into three groups: 11 cases in the hypomethylation
group, 2 cases in the moderate methylation group, and 11 cases in the hypermethylation
group. The concordance rate of our methylation subgroup compared with the international
consensus classification was 91.7%. There were significant differences in age, platelets,
PTPN11 gene mutation and the number of somatic mutations ≥2 in different methylation
groups (P<0.05). Correlation analysis showed that hypermethylation level was significantly
correlated with PTPN11 gene mutation and ≥2 somatic mutations (P<0.001).
Image:
Summary/Conclusion: JMML patients with high methylation level in the DNA genome at
diagnosis were older and with lower platelet levels, and hypermethylation were significantly
correlated with high-risk prognostic factors such as PTPN11 gene mutation and ≥2 somatic
mutations.
P760: PATTERNS OF HYPOMETHYLATING AGENT FAILURE IN PATIENTS WITH MYELODYSPLASTIC SYNDROMES
K. Chien1,*, K. Kim1, Z. Li2, R. Kanagal Shamanna3, F. Ong1, G. Montalban Bravo1,
T. Kadia1, E. Jabbour1, N. Pemmaraju1, D. Hammond1, N. Short1, F. Ravandi1, Y. Alvarado1,
S. Pierce1, X. Q. Dong1, H. Kantarjian1, G. Garcia-Manero1
1Leukemia; 2Biostatistics; 3Hematopathology, The University of Texas MD Anderson Cancer
Center, Houston, United States of America
Background: Hypomethylating agents (HMA), such as azacitidine and decitabine, are
the current standard-of-care for patients (pts) with myelodysplastic syndrome (MDS).
Though HMA improve cytopenias and delay progression of disease, most pts eventually
response to these agents with poor survival outcomes afterward. This poses significant
treatment challenges after HMA failure (HMA-F) and an increased risk of transformation
to acute myeloid leukemia (AML).
Aims: Here, we assess the clinicopathologic characteristics and outcomes of pts with
HMA-F MDS.
Methods: We retrospectively evaluated all pts with MDS seen at a single tertiary cancer
center from July 2017 to July 2021 and identified those who were previously untreated
and later developed HMA-F. Pt characteristics, laboratory values, and bone marrow
(BM) data, including cytogenetics and next generation sequencing (NGS), were assessed
at both diagnosis and the time of HMA-F. Genomic DNA was extracted from whole BM aspirate
samples and subject to 28- (n=12) or 81-gene (n=131) target PCR-based sequencing using
a NGS platform at diagnosis and an 81-gene NGS panel (n=85) at the time of HMA-F.
Survival data was updated in January 2022.
Results: Out of 799 untreated MDS pts, 147 pts (32%) developed HMA-F with a median
follow-up time of 44.0 months (mo) from diagnosis. Baseline pt characteristics and
subsequent treatment are summarized in Table 1. The median age was 71 with therapy-related
MDS (t-MDS) in 37%. At diagnosis, most pts had higher-risk MDS with IPSS-R high (29%)
or very high (39%) disease, and complex karyotype was seen in 44%. The median number
of mutations identified was 2 with TP53 (44%) and ASXL1 (21%) the most frequently
discovered mutations. Regarding HMA therapy, 67% received HMA monotherapy and 33%
received HMA in combination with various agents. The median number of cycles of HMAs
received was 7 (range: 1-37).
At the time of HMA-F, pts presented with higher BM blasts (6% vs 10%, p<0.001) and
lower platelet counts (63 K/µL vs 36 K/µL, p<0.001) than at diagnosis. The median
number of cytogenetic abnormalities increased from 1 to 2 with no change in the median
number of mutations from diagnosis to HMA-F, but both had significant increases by
paired t-test (p<0.001). The median time from diagnosis to HMA-F was 10.8 mo, and
the median overall survival (mOS) from HMA-F was 6.8 mo (95% CI: 5.4, 9.0). Survival
was well-stratified by both IPSS-R at diagnosis (14.6 mo in lower-risk MDS vs 5.4
mo in higher-risk MDS, p=0.001) and at HMA-F (11.4 mo in lower-risk MDS vs 3.5 mo
in higher-risk MDS, p<0.001). TP53 mutations at diagnosis (HR 3.02, p<0.001) and t-MDS
(HR 2.50, p<0.001) were associated with shorter mOS. In pts with higher-risk MDS,
initial response to HMA therapy (7.2 mo in responders vs 2.7 mo in non-responders,
p=0.011) and stem cell transplantation at HMA-F (4.7 mo in non-transplanted vs not
reached in transplanted, p<0.001) were associated with improved survival. Progression
to acute myeloid leukemia (AML) was seen in 72 pts (49%), and the mOS from the time
of AML diagnosis was 3.4 mo.
Image:
Summary/Conclusion: Pts with HMA-F MDS have high-risk features, including poor cytogenetics
and adverse mutations, and are often therapy-related. At the time of HMA-F, patients
present with higher BM blasts, worsening thrombocytopenia, and more cytogenetic abnormalities
and mutations. Unfortunately, after failure of HMA, pts have frequent transformation
to AML and dismal overall survival. Further understanding of the underlying biology
of HMA-F MDS is warranted with an urgent need for therapeutic interventions after
failure of HMA therapy.
P761: MONOCYTOSIS IN PRIMARY CARE AND HEMATOLOGICAL MALIGNANCIES
M. Christensen1,2,*, V. Siersma2, M. Kriegbaum2, B. Lind3, J. Samuelsson4, K. Grønbæk1,
L. Granfeldt5, C. L. Andersen1
1Department of Hematology, Rigshospitalet; 2Institute for Public Health, University
of Copenhagen, Copenhagen; 3Department of Clinical Biochemistry, Copenhagen University
Hospital Hvidovre, Hvidovre, Denmark; 4Department of Hematology, Universitetssjükhuset,
Lindköping, Sweden; 5Department of Hematology, Odense University Hospital, Odense,
Denmark
Background: Monocytosis (blood monocyte count > 0.8 x109/L) is relatively often observed
in routine blood work from primary care, however, the significance is not always apparent
as monocytosis may be present in a spectrum of diseases spanning from mild infections
to chronic myelomonocytic leukemia.
Aims: We aimed to examine the predictive value of monocytosis in primary care by relating
monocyte count to subsequent 3-year incidence of hematological malignancy.
Methods: We included 663.184 adult patients from primary care in the greater Copenhagen
Area who had a complete blood cell count (CBC) performed at any time between 2000-2016.
For patients with multiple samples registered, a random sample was selected and samples
predating the index CBC by 3 months were assessed to account for sustained monocytosis.
Using the extensive Danish health data registers, we collected data on incident hematological
malignancy for 3 years following the CBC. We modelled the association between monocyte
count and hematological malignancies as well as all-cause mortality using multiple
logistic regression analysis.
Results: Monocytosis was observed in 4,6 % of CBCs. We saw an increased risk of myeloid
malignancies with the most noticeable being an odds ratio (OR) for CMML > 100 with
monocytosis > 1.0 x 109/L. Sustained monocytosis increased this risk to OR >140. The
incidence was, however, still low with only 0.1% of patients with monocytosis developing
CMML within 3 years. Risk of lymphoproliferative and M-component related diseases
were also increased, however, other relevant cell line were affected as well in these
cases.
Summary/Conclusion: Monocytosis is a common finding in primary care, but even with
monocytosis > 1 x 109/L hematological malignancies are rare. Optimized referral to
secondary care remains a challenge and if the clinical presentation leaves doubt of
the diagnosis, repeated measurements help demask if malignancy is the cause of the
monocytosis.
P762: NONLINIEAR IMPACT OF CLINICAL FEATURES ON THE SURVIVAL OUTCOME OF MYELODYSPLASTIC
SYNDROMES PATIENTS
F. Darbaniyan1,*, G. Montalban Bravo2, K. S. Chien2, R. Kanagal Shamanna3, K. sasaki2,
H. Yang2, K. A. Soltysiak2, Z. Li1, K.-A. Do1, G. Garcia-Manero2
1Biostatistics; 2Leukemia; 3hematopathology, MD Anderson Cancer Center, Houston, United
States of America
Background: Myelodysplastic syndromes (MDS) are a group of hematopoietic stem-cell
disorders with heterogenous prognosis. The revised international prognostic scoring
system (IPSS-R) (Greenberg et al., Blood, 2012) is the most widely used score for
prognostication. However, the allocation of IPSS-R risk categories remains imprecise
for some patients leading to unsatisfactory therapy decisions. Newer molecular prognostic
models (Nazha et al., JCO, 2021 and Bernard et al., ASH, 2021) have added mutation
data to improve survival predictions for MDS patients.
Aims: Our aims were to investigate the nonlinear association of normalized clinical
data in combination with molecular and cytogenetic data and to improve the prognostication
potential of IPSS-R for patients with MDS.
Methods: A multivariable fractional polynomials (MFP) algorithm was applied on clinical
data of 943 MDS patients and nonlinear associations between continuous covariates
and survival outcomes were efficiently modeled within Cox regression. Selected transformed
variables were picked and the potential overfit was avoided by a closed test procedure
for function selection (Royston et al., John Wiley & Sons, 2008). The performance
of the model was evaluated in an independent set of 436 MDS patients and the accuracy
was assessed using a concordance (c)index and the area under the Receiver Operating
Characteristic curve (AUC) at each time point. Mutation data was obtained in a subset
of 695 patients using next-generation sequencing, evaluating a panel of 81 genes at
the time of diagnosis.
Results: An MFP-model consisting of cytogenetics per IPSS-R and WHO subtypes, continuous
forms of age and hemoglobin, and non-linear transformations of platelets and bone
marrow blast percentage yielded a c-index performance of 0.82 vs 0.78 for IPSS-R and
a significant AUC upgrade at all time points in the training set (Figure a). In comparison
with the IPSS-R, absolute neutrophil count was not identified as an independent prognostic
covariate and common features were treated in a non-linear fashion. Validation of
the model in an independent set of 438 patients yielded a higher c-index (0.72 vs
0.68) with an improved AUC over time compared to the IPSS-R (Figure b). Based on MFP
risk score, patients were then clustered into 5 different risk categories and compared
with IPSS-R classifications. Our data showed no significant difference in time to
death between IPSS-R groups for very low risk and low risk or between low risk and
intermediate risk (Figure c); however, MFP classification showed 5 significantly different
subgroups in predicting time to death for MDS patients (Figure d).
We further investigated the effect of molecular data on disease prognostics in a subset
of 695 patients sequenced using an 81 gene panel. In addition to the selected clinical
covariates, occurrence of TP53, SRSF2, and ZRSR2 mutations were independently associated
with lower survival, while the presence of TET2 mutation showed a depression effect
on TP53 mutation towards a better outcome. As SF3B1 was not selected in the final
model, we further evaluated its potential interactions and identified its correlation
with cytogenetics and platelets, which predominated in the MFP model. This might be
due to heterogeneity within the data set (e.g., 25% co-occurrence of SF3B1 with TP53
mutation) and needs to be investigated in larger cohorts.
Image:
Summary/Conclusion: We have established a nonlinear model that yields improved risk
assessment compared to IPSS-R criteria.
P763: VENETOCLAX WITH AZACITIDINE IN RELAPSE/REFRACTORY HIGHER RISK MYELODYSPLASTIC
SYNDROME: UPDATED PHASE 1 RESULTS
S. P. Desikan1,*, G. Montalban-Bravo1, M. Ohanian1, N. Daver1, T. Kadia1, S. Venugopal1,
K. Chien1, H. Kantarjian1, G. Garcia-Manero1
1Leukemia, MD Anderson, houston, United States of America
Background: Patients with higher risk (HR) MDS status post HMA failure have poor survival,
4-6 months. [Lancet Oncology Apr 2016] The combination of azacitidine(Aza) and venetoclax(Ven)
has produced encouraging results in the frontline settings for patients with HR disease.(Garcia
et.al ASH 2019)
Aims: The primary objective is to establish the safety of azacitidine and venetoclax
in HR MDS after HMA failure. The secondary objectives include rates of CR, marrow
response, transfusion independence, duration of response, and overall survival.
Methods: In this phase 1 study, patients 18 years and older with HMA failure (relapse
or progression after 4 cycles), adequate organ function and performance status are
being enrolled. Patients who had received prior Ven were excluded. All patients received
Aza 75mg/m2 on D1-5. A 3 + 3 design was utilized with Ven. A 3 + 3 design was utilized
with Ven with dose levels 0, 1, 2 receiving 100mg, 200mg, and 400mg respectively.
Patients are currently being enrolled in the expansion cohort(400mg). Response was
determined based on IWG06 criteria.
Results: Baseline characteristics are in Table 1. Fifteen patients have been enrolled
with a median age of 78[67 – 81]. The median blast% on enrollment was 9%[6 – 17] with
a median ANC, Hgb, and PLT of 1.67 K/µL, 7.7mg/dL, and 39 K/µL. When stratified based
on cytogenetics, 1 (7%) had good risk, 7 (47%) patients had intermediate risk, 2 (13%)
had poor risk, and 5(33%) had very poor risk. When stratified based on IPSS-R criteria:
2(13%) had intermediate risk, 5(33%) had high risk, and 8(54%) had very high risk
disease. ASXL1 and TP53 mutations were enriched, occurring in 46% and 53% of patients.
Four(33%) patients had Grade ¾ cytopenias (either thrombocytopenia or neutropenia)
relating to this combination. Three patients required dose reductions. No tumor lysis
syndrome has occurred to date.
Two patients recently started treatment and could not be included for response assessment.
Among the 13 patients evaluable, the ORR is 62%(n=8). The median number of cycles
to response was 1[1-4]. Responses consisted of 1 CR, 4 marrow responses with either
platelet or neutrophil recovery, and 3 marrow responses without count recovery. Three
patients achieved transfusion independence. Five patients had NR with 4 having TP53
mutations.
The median OS was 8.5 months with a 30 day and 60 day mortality of 8% and 23% respectively.
Early mortality was related to infection in 2 patients and progression in another.
Image:
Summary/Conclusion: With a response rate of 62% and an OS of 8.5 months, this combination
may have benefit in HR RR MDS. TP53 mutations and higher cytogenetic risk confer poor
prognosis.
P764: RESPONSE AND SURVIVAL OUTCOMES WITH HYPOMETHYLATING AGENTS IN AN ARGENTINEAN
COHORT OF 113 PATIENTS WITH CHRONIC MYELOMONOCITIC LEUKEMIA
J. Gonzalez1,*, A. Perusini2, F. Russo3, L. Fuentes4, T. Deluca5, A. L. Basquiera6,
A. Navickas7, L. Kornblihtt8, N. Pintos9, E. Nucifora2, M. Iastrebner10, A. Enrico5,
J. Arbelbide2, C. Belli11
1Hematology, Hospital Agudos Carlos G Durand; 2Hematology, Hospital Italiano de Buenos
Aires; 3Hematology, Hospital Paroissien; 4Hematology, Instituto Alexander Femming,
CABA; 5Hematology, Hospital Italiano de La Plata, La Plata; 6Hematology, Hospital
Privado de Cordoba, Cordoba; 7Hematology, Hospital El Cruce, Florencio Varela; 8Hematology,
Hospital de Clinicas Jose de San Martin; 9Hematology, Sanatorio Mendez; 10Hematology,
Sanatorio Sagrado Corazon; 11Genetic, IMEX-CONICET/ANM, CABA, Argentina
Background: Hypomethylating agents (HMA) are the first line option for high-risk patient
and lower-risk patient with transfusion dependence, or according to worsening clinical
features. Data regarding HMA efficacy in CMML has been largely retrospective, or from
MDS studies that included CMML patients, with overall response rates (ORR) ranging
from 40% to 50% and true CR rates being <20%. Local data is scarce.
Aims: To examine the influence of prognostic factors at diagnosis and during the follow-up
in the outcome and response to HMA therapy in a CMML cohort from Argentine.
Methods: We performed a retrospective analysis of 113 CMML patients from the Argentine
Registry of MDS promoted by the Argentine Society of Hematology who were treated between
January-07/February-22. Statistical analysis included Kaplan-Meier survival analysis,
Cox proportional hazard and Chi2/Fisher’s exact test.
Results: The median age at diagnosis was 67 years (IQR 58-74) being 71.7% >60 years
old, 18.8% a Charlson’s Index (CCI)>2, 81 (72%) were males, and 59 (52%) CMML-0, 25
(22%) CMML-1 and 29 (26%) CMML-2. At treatment initiation, 70.5% showed hemoglobin
<10g/dL, 22.1% platelet counts <30,000/µL, 13.1% poor karyotypes and 91.2% at high
risk according to the CPSS, IPSS, IPSS-R or Bournemouth scoring systems. During the
follow-up, median 20 months (IQR 8.6-42.7), 45.1% evolve to AML and 75.2% died. Regarding
HMA therapy, the median time to treat was 2.4 months, 78.8% patients received AZA
and 22.2% DAC, the median number of cycles of 7 (IQR 4-12) during a median period
of 8.1 months (m). The median overall survival (OS) of the cohort was 25.5m (95%CI
18.9-32.1), since treatment initiation 17.1m (95%CI 13.3-20.8), and after cessation
5.3m (95%CI 4.3-6.3).
Most of parameters and scoring systems analyzed were useful to predict outcome from
diagnosis or from treatment initiation to last follow up. Cox regression analysis
revealed that CCI>2 (HR 2.7, 95%CI 1.5-5.0, p=0.001), WHO classification (ref. CMML-0,
CMML-1, HR 2.1, 95%CI 1.1-4.1, p=0.026; CMML-2, HR 3.4, 95%CI 1.6-6.8, p=0.001), lower
hemoglobin level (<10g/dL, HR 1.9, 95%CI 1.1-3.5, p=0.023), platelet count <30.000/µL
(HR 3.5, 95%CI 2.0-6.0, p<0.001), time to treat <6m (HR 4.7, 95%CI 2.4-9.1, p<0.001)
and the presence of blast in PB (HR 1.7, 95%CI 1.0-2.8, p=0.05) and were independently
associated with a reduced OS. Almost all parameters, with the exception of lower Hb
levels, sustained their independency since treatment initiation.
A total of 99 patients were evaluated for response to treatment with an overall response
rate (at 4-6 cycles) of 59.6% (CR/mCR/PR: 36.4%, HI: 23.2%, SD: 14.1%). The median
overall survival of responders was 44.8m, similar to those with SD (33.2m, p=0.246),
afterward grouped, with 8.3m in non-responders (p<0.001). We also evaluated whether
prognostic factors were useful to predict response. Only the WHO 2016 proposal (CMML-0
61.6% vs 34.6%, CMML-1 21.9% vs 30.8%, CMML-2 16.4 vs 34.6%, p=0.046) and the absence
of peripheral blasts (61.1% vs 36.0%, p=0.037) were associated with statistically
different rates of response, with a tendency to treat later ≥6m (37.0% vs 15.4%, p=0.05).
Summary/Conclusion: The present series represents the first experience in Latin America
evaluating HMA agents in CMML patients. Our results highlight the adverse impact of
several parameters, including the severity of the thrombocytopenia <30,000/µL on the
outcome of CMML patients under HMA. However, clinical parameters are limited to predict
the response rate.
P765: UPDATED ENROLLMENT AND RESULTS FROM THE PHASE 1 SUBSTUDY OF IVOSIDENIB IN PATIENTS
WITH IDH1-MUTANT RELAPSED/REFRACTORY MYELODYSPLASTIC SYNDROME (R/R MDS)
C. D. DiNardo1,*, J. M. Foran2, J. M. Watts3, E. M. Stein4, S. de Botton5, A. T. Fathi6,
G. T. Prince7, R. M. Stone8, P. A. Patel9, G. J. Roboz10, M. L. Arellano11, H. P.
Erba12, A. Pigneux13, P. Baratam14, R. K. Stuart14, X. Thomas15, I. R. Lemieux16,
X. Bai16, S. M. Kapsalis16, G. Garcia-Manero1, D. A. Sallman17
1MD Anderson Cancer Center, University of Texas, Houston; 2Mayo Clinic, Jacksonville;
3Sylvester Comprehensive Cancer Center, University of Miami, Miami; 4Memorial Sloan
Kettering Cancer Center, New York, United States of America; 5Institut Gustave Roussy,
Villejuif, France; 6Massachusetts General Hospital, Harvard Medical School, Boston;
7Johns Hopkins Hospital, Baltimore; 8Dana-Farber Cancer Institute, Boston; 9University
of Texas Southwestern Medical Center, Dallas; 10Weill Cornell Medicine and The New
York Presbyterian Hospital, New York; 11Winship Cancer Institute, Emory University
School of Medicine, Atlanta; 12Duke University, Durham, United States of America;
13Centre Hospitalier Universitaire de Bordeaux Haut-Lévêque, Pessac, France; 14Medical
University of South Carolina, Charleston, United States of America; 15Centre Hospitalier
Universitaire de Lyon-Sud, Lyon, France; 16Servier Pharmaceuticals LLC, Boston; 17Moffitt
Cancer Center, Tampa, United States of America
Background: Isocitrate dehydrogenase 1 (IDH1) is mutated in ~3% of patients with MDS,
increasing the risk of transformation to acute myeloid leukemia (AML). Ivosidenib
(IVO), an oral, potent, targeted inhibitor of the mutant IDH1 (mIDH1) enzyme, is FDA
approved for mIDH1 R/R AML and mIDH1 newly diagnosed AML in patients ≥75 years old
or with comorbidities precluding the use of intensive induction chemotherapy. Promising
safety and efficacy findings in the first-in-human, open-label, dose escalation and
expansion study of IVO in patients with mIDH1 advanced hematologic malignancies (NCT02074839)
led to the FDA granting Breakthrough Therapy designation to IVO in mIDH1 MDS. 12 patients
with R/R MDS received IVO 500 mg once daily (QD). Median age was 72.5 (range 52–78)
years and all had received prior MDS treatment. The investigator-assessed overall
response rate (ORR) was 75%, with median response duration of 21.4 months. This substudy
enrolled additional patients with mIDH1 R/R MDS; therefore, we report updated results.
Aims: To further evaluate safety, tolerability, clinical activity, and pharmacokinetics/pharmacodynamics
of IVO in patients with mIDH1 R/R MDS.
Methods: This substudy of the single-arm study of IVO evaluated patients with mIDH1
R/R MDS after documented failure or relapse following prior standard therapy including
intensive chemotherapy and hypomethylating agents. Patients must have given informed
consent, have high disease burden based on IPSS or IPSS-R risk at baseline, and an
Eastern Cooperative Oncology Group performance status score of 0–2. IVO 500 mg QD
was given orally on days 1–28 of 28-day cycles.
Results: As of 08May2021, 16 patients with R/R MDS were enrolled: 3 of the 16 who
received 500 mg IVO QD were women; median age was 73.5 (range 52–78) years and 44%
were ≥75 years of age. As of the data cut, 5 (31%) patients remained on treatment
and free from leukemic transformation and 11 (69%) had discontinued including 6 for
disease progression, 1 for allogeneic stem cell transplantation, and 1 owing to an
adverse event (AE) of sepsis (the only fatal AE; reported by investigator as not related
to IVO). Differentiation syndrome was observed in 2 patients (grade 2). Electrocardiogram
QT prolonged was reported in 2 patients (grade 1 and 2). The Table outlines AEs. Complete
response (CR) was achieved by 7 of 16 patients (44%; 95% CI, 20%, 70%), partial response
(PR) by 1 (6%), and marrow CR by 5 (31%), giving an ORR of 81% (95% CI, 54%, 96%).
At 12 months, the Kaplan-Meier estimate of duration of CR+PR was 60.0%. 3 patients
experienced CRs lasting 24.0, 63.7, and 65.4 months, which remain ongoing. 11 of 16
(69%) patients achieved hematologic improvement in ≥1 lineage. Among 7 patients who
were transfusion dependent at baseline, 5 (71%) became independent of red blood cell
or platelet transfusions for 56 or more consecutive days on treatment. Further translational
data are being processed.
Image:
Summary/Conclusion: IVO monotherapy induced durable remissions and transfusion independence
in patients with mIDH1 R/R MDS, with a manageable safety profile. These results contribute
to IVO’s potential as an effective, oral, targeted treatment for patients with mIDH1
R/R MDS.
P766: CLINICAL OUTCOMES IN PATIENTS WITH HIGHER-RISK MYELODYSPLASTIC SYNDROMES RECEIVING
HYPOMETHYLATING AGENTS: A LARGE POPULATION-BASED ANALYSIS
A. M. Zeidan1,*, E. S. Mearns2, C. Ng2, A. Shah2, N. Lamarre3, A. Yellow-Duke2, N.
Alrawashdh2, B. Yang4, W. Cheng5, C. N. Bui5, A. Svensson5
1Yale Cancer Center, Yale University School of Medicine, New Haven; 2Genentech, Inc.,
South San Francisco; 3Real World Data Analytics, Genesis Research, Hoboken; 4Roche
Diagnostics, Santa Clara; 5AbbVie, North Chicago, United States of America
Background: Myelodysplastic syndromes (MDS) are a spectrum of hematopoietic neoplasms
characterized by variable degrees of cytopenias and risk of progression to acute myeloid
leukemia (AML). Although the hypomethylating agents (HMA) azacitidine (AZA) and decitabine
(DEC) are widely used to treat higher-risk (HR)-MDS in the United States (US), questions
remain about their longer-term clinical benefits.
Aims: To understand the survival outcomes and progression-related events of patients
(pts) treated with AZA or DEC for HR-MDS in the US.
Methods: Older adults (≥66 years old) diagnosed with refractory anemia with excess
blasts (RAEB), an established proxy for HR-MDS, between January 2009 and December
2017, were included from the Surveillance, Epidemiology, and End Results (SEER)-Medicare
linked database. The index date was date of HMA initiation. Pts who received AZA or
DEC, had ≥12 months (mo) of continuous enrollment prior to the index date, and who
did not receive a stem cell transplant (SCT) prior to the index date were included.
Overall survival (OS) from the index date to death was estimated using Kaplan–Meier
methodology. Baseline platelet and red blood cell (RBC) transfusion dependence (TD)
were defined as ≥2 transfusion episodes in the 8 weeks prior to the index date; baseline
transfusion use (TU) was defined as one transfusion episode in the 8 weeks prior to
the index date. A competing risk analysis was used to assess the incidence of progression
to AML, with death as the competing event.
Results: Of 973 pts with HR-MDS treated with HMA, 75.8% received AZA and 24.2% received
DEC. Median time from diagnosis to HMA initiation was 1.2 mo (interquartile range
[IQR]: 0.6–2.6). Median treatment duration was 5.7 mo (IQR: 2.3–11.5) overall (5.8
mo [IQR: 2.6–11.8] for AZA and 5.0 mo [IQR: 1.9–11.0] for DEC). At baseline, 13.8%
of pts had platelet TD/TU, 29.9% had RBC-TD and 27.0% had RBC-TU. Only 6.4% of pts
received SCT during follow-up.
Median follow-up from the index date until death or censoring was 13.8 mo (IQR: 6.2–25.3).
Median OS (mOS) was 13.9 mo (95% confidence interval: 12.9–15.0). There was no significant
difference in mOS for pts treated with AZA vs DEC (13.4 vs 15.2 mo, respectively;
p=0.22). Pts with RBC-TD/TU had shorter mOS vs those with RBC-transfusion independence
(TI) (10.7 vs 18.6 mo, respectively; p<0.0001). Similarly, pts with platelet TD/TU
had shorter mOS vs those with platelet TI (8.4 vs 14.9 mo, respectively; p<0.0001).
There were no statistically significant differences in OS by the route of AZA administration,
insurance category, county metropolitan status, or county poverty level. Overall,
38.0% of HMA-treated pts progressed to AML; median time from HMA initiation to AML
progression was 8.2 mo (IQR: 3.5–15.1). Cumulative incidences of AML and death were
25.7% and 30.7%, respectively, at Year 1 and 34.3% and 45.8%, respectively, at Year
2, with the rates of progression slowing over time.
Summary/Conclusion: Compared with previously published literature from the SEER-Medicare
database (Zeidan Br J Haematol 2016; mOS: 11 mo [AZA] and 12 mo [DEC] in pts diagnosed
with HR-MDS between 2004 and 2011), pts with HR-MDS treated with HMA have improved
mOS, but OS remains substantially worse vs the landmark AZA-001 trial (Fenaux Lancet
Oncol 2009; mOS: 24.5 mo). The incidence of death and AML progression was highest
in the first year after initiation of therapy. SCT rates remain very low among pts
with HR-MDS, further emphasizing the urgent need for novel treatment options for pts
with HR-MDS.
P767: CLINICAL SIGNIFICANCE OF ROUTINE HIGH-RESOLUTION STRUCURAL VARIANT PROFILING
IN MYELODYSPLASTIC SYNDROMES
H. Yang1, G. Garcia-Manero1, K. Sasaki1, G. Montalban-Bravo1, Z. Tang1, Y. Wei1, T.
Kadia1, K. Chien1, D. Rush1, H. Nguyen1, A. Kalia1, M. Nimmakayalu1, C. Bueso-Ramos1,
H. Kantarjian1, L. J. Medeiros1, R. Luthra1, R. Kanagal-Shamanna1,*
1THE UNIVERSITY OF TEXAS M.D. ANDERSON CANCER CENTER, Houston, United States of America
Background: Structural Variant Profiling (SVP) using high-resolution technologies,
in combination with NGS-based mutation analysis, can inform critical cytogenomic data
needed for improving the prognostication and outcomes in patients (pts) with MDS.
Optical Genome Mapping (OGM) is a novel non-sequencing-based technique for genome-wide
high-resolution SVP for all types of SVs: copy number alterations, structural rearrangements,
inversions and partial tandem duplications (PTD).
Aims: The goal was to evaluate the clinical implications of SVP on prognosis and risk-stratification
in a large series of consecutive treatment naïve MDS pts.
Methods: All patients (pts) underwent conventional chromosomal banding analysis (CBA),
OGM and targeted 81-gene panel NGS. We compared the cytogenetic [based on comprehensive
cytogenetic scoring system (CCSS)] and R-IPSS risk groups between data generated by
CBA and OGM respectively and correlated with outcomes. For OGM, CCSS was calculated
from the predicted karyotype generated by consolidatied alignment of multiple individual
SVs.
Results: There were 101 newly diagnosed MDS pts (71 men; 29 women). The median age
was 72 (25-92). The distribution of CCSS groups [Very Good (3%), Good (43%), Intermediate
(22%), Poor (10%), Very Poor (21%)] and R-IPSS risk categories [Very low (5%), low
(30%), Intermediate (27%), High (17%), Very high (20%)]. Two patients had incomplete
karyotype.
By generating predicted karyotypes from OGM data, there were 224 clinically significant
unique aberrations. Of these, 94 (40%) aberrations (seen across 34 patients) were
not detected by CBA. These included SVs affecting KMT2A, MECOM, TP53, NUP98 among
others. Comparison of CCSS cytogenetic groups between OGM and CBA showed that OGM
data upgraded the CCSS cytogenetic-risk in 12 pts (resulting in R-IPSS upgrade in
9 pts) and downgraded in 7 pts (R-IPSS downgraded in 6 pts). Together with the results
for 2 patients with non-informative karyotype, OGM modified the CCSS for 21% pts,
and R-IPSS category for 17 (17%) pts. Since CCSS is influenced by the number of alterations,
there were patients where OGM data, though clinically important, did not change the
CCSS/R-IPSS category. To capture this, we defined an “actionable SV” as (1) prognostic,
not recognized in the CCSS criteria (KMT2A-PTD, TP53 allele status) or (2) potential
eligibility for a clinical trial, such as rearrangements in KMT2A, MECOM, or NUP98.
Well-established SVs defined in CCSS were ignored. Based on this, 13 (13%) pts had
an “actionable alteration” (9 with a positive finding). When both the CCSS change
and actionable alterations were combined, OGM results provided informative data in
28 (28%) patients.
Next, we evaluated the impact of CCSS-CBA and CCSS-OGM groups using outcome data.
Over a median follow-up duration of 34 months, the median OS was 86 months. Using
Harrell’s concordance index, OGM had a slightly improved prediction of prognosis for
both CCSS [0.7 vs. 0.665] and R-IPSS categories [0.705 vs. 0.69] compared to CBA.
By UVA, the following parameters associated with outcome: SF3B1 mutation [p=0.030;
HR 0.343; CI 0.130-0.901], TP53 mutation [p=0.001; HR 3.232; CI 1.582-6.601], CCSS
score by CBA [p=0.001; HR 1.690; CI 1.250-2.286] and CCSS by OGM [p<0.001; HR 2.051;
CI 1.488-2.827]. By MVA analysis, CCSS by OGM [p<0.001; HR 1.956; CI, 1.367-2.798],
but not CCSS by CBA), BM blast percentage [p=0.026; HR 1.079; CI, 1.009-1.153] and
TP53 mutation [p=0.040; HR 2.335; CI, 1.038-5.250] associated with prognosis.
Summary/Conclusion: High-resolution SVP by OGM improved the prognostic risk stratification
in MDS.
P768: GUADECITABINE (SGI-110) VS. TREATMENT CHOICE (TC) IN RELAPSED/REFRACTORY(R/R)
MYELODYSPLASTIC SYNDROME (MDS), RESULTS OF A GLOBAL, RANDOMIZED, PHASE 3 STUDY.
G. Garcia-Manero1,*, S. Bart2, J. K. McCloskey3, P. Fenaux4, D. Selleslag5, G. Reda6,
D. Valcárcel7, V. Santini8, J. Mayer9, B. Xicoy10, H. Yamaguchi11, M. Lübbert12, Y.
Miyazaki13, H. Keer14, Y. Hao14, M. Azab14, H. Döhner15
1The University of Texas MD Anderson Cancer Center, Houston; 2Fred Hutchinson Cancer
Research Center, Seattle; 3John Theurer Cancer Center at Hackensack University Medical
Center, Hackensack, United States of America; 4Hôpital Saint Louis, Paris, France;
5Algemeen Ziekenhuis Sint-Jan Brugge-Oostende, Brugge, Belgium; 6Fondazione IRCCS
Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy; 7Hospital Universitari Vall
d’Hebrón, Barcelona, Spain; 8MDS Unit AOU Careggi, DMSC University of Florence, Florence,
Italy; 9Fakultní Nemocnice Brno, Brno, Czechia; 10Institut Català d’Oncologia-Hospital
Universitari Germans Trias i Pujol, Badalona, Spain; 11Nippon Medical School Hospital,
Tokyo, Japan; 12Universitaetsklinikum Freiburg, Freiburg, Germany; 13Nagasaki University
Hospital, Nagasaki, Japan; 14Astex Pharmaceuticals, Inc., Pleasanton, United States
of America; 15Ulm University Hospital, Ulm, Germany
Background: Guadecitabine (G) is a next-generation hypomethylating agent (HMA), administered
as a small-volume subcutaneous (SC) injection, designed with the potential to overcome
pharmacokinetic resistance to first-generation HMAs (decitabine and azacitidine).
Preliminary guadecitabine phase 2 data in r/r MDS showed an overall survival of almost
12 months (Garcia-Manero,G, et al; Lancet Haematol http://dx.doi.org/10.1016/S2352-3026
[19]30029-8) leading to the ASTRAL-3 study.
Aims: Compare overall survival of guadecitabine to that of TC consisting of low-dose
cytarabine (LDAC), intensive chemotherapy (IC), or best supportive care (BSC). Secondary
objectives included multiple standard assessments of response and safety.
Methods: Subjects with r/r MDS or Chronic Myelomonocytic Leukemia (CMML) who had progressed
or failed to respond after 6 full cycles of standard HMA therapy were randomized 2:1
between guadecitabine (60 mg/m2) SC Days 1-5 every 28 days vs. a pre-selected TC (BSC,
IC, or LDAC). Stratification was by disease, disease burden (>10% bone marrow blasts
or not, geography, and pre-selected TC option). Primary endpoint was overall survival
(OS) and the study was designed with ~90% power to detect a hazard ratio of 0.68 (approximately
2.8 month difference in median survival).
Results: 417 MDS/CMML subjects were randomized to G or TC (G:277, TC: 140). Demographics
and disease status were reasonably balanced across the groups (see Table 1 below)
as was percentage of commonly associated genetic mutations (TET2, DNMT3A, SF3Baa,
RUNX1, and TP53). Median guadecitabine exposure was 4 cycles and 3 cycles for TC.
Neither overall survival (G: median 9.1 months, TC: median 8.3 months, p=0.61, HR:0.94
with 95% CI: [0.74-1.19]) nor leukemia-free survival (LFS) (G: median 5.7 months,
TC: median 5.9 months; p=0.38) demonstrated a significant difference between the two
groups and subgroup analyses did not suggest a difference between guadecitabine and
any of the different TC options or different genetic mutations. Transfusion dependence
for 8 weeks was similar with 32.1% and 22.4% of the G group and 37.9% and 20.0% of
the TC group being platelet transfusion or red blood cell transfusion independent,
respectively. Safety was consistent with known profiles of the respective agents with
the G group having a slightly higher overall incidence of adverse events (AE) (99.3%
vs 92.6% for TC) and grade 3 or higher AE (92.2% vs 70.5% for TC). The AEs with the
highest incidence in subjects who received guadecitabine were febrile neutropenia
(38.5 vs 18.9% for TC), pneumonia (34.4% vs 18.9% for TC), neutropenia (34.1% vs 15.6%
for TC), and thrombocytopenia (32.2% vs 21.3% for TC).
Image:
Summary/Conclusion: This large, global, randomized phase 3 study did not demonstrate
superiority of guadecitabine over Standard TC in MDS/CMML patients who were refractory
or relapsed following full course of prior HMA treatment.
P769: MUTATION PROFILES AND RISK STRATIFICATION IN HYPOCELLULAR MYELODYSPLASTIC SYNDROME
K. Kim1,*, K. Chien1, O. Faustine1, G. Montalban-Bravo1, R. Kanagal-Shamanna2, T.
Kadia1, E. Jabbour1, Y. Alvarado1, K. Sasaki1, X. Q. Dong1, S. Pierce1, C. Bueso-Ramos2,
H. Kantarjian1, G. Garcia-Manero1
1Department of Leukemia; 2Department of Hematopathology, The University of Texas MD
Anderson Cancer Center, Houston, United States of America
Background: Hypocellular myelodysplastic syndrome (hMDS) is a subset of MDS, defined
by marrow cellularity less than 30% in age under 70 and less than 20% in age 70 or
more. The clinicopathologic features of hMDS have been previously studied, but genetic
data and clinical impact of hMDS remain unclear.
Aims: We conducted a comparative study of hMDS to normo-/hypercellular MDS (non-hMDS)
with clinical characteristics and mutations and further stratified the risk by mutation
profile.
Methods: We conducted a retrospective review using a single tertiary cancer center
registry of untreated MDS patients from 2020 to 2021. Mutation data was obtained by
28- or 81-gene next-generation sequencing (NGS) panels. We analyzed clinicopathological
and mutation data comparing hMDS to non-hMDS pts and conducted risk stratification
in hMDS using mutation profiles.
Results: In a total of 1899 patients, 174 (9%) patients had hMDS. 1352 (71%) pts had
mutation profiles that were further analyzed; 56% and 44% patients had 81-gene and
28-gene NGS panels performed, respectively. The median follow-up time was 19.5 months.
Thrombocytopenia (p=0.016) and neutropenia (p<0.0001) were more common in hMDS than
in non-hMDS. By IPSS-R cytogenetic scores, hMDS had scores of 3 than non-hMDS (20%
vs. 9%, p<0.001), but non-hMDS had more cytogenetic scores of 4 (27% vs. 19%, p=0.018).
The distribution of other IPSS-R cytogenetic scores or IPSS-R risk classifications
was similar between the 2 groups. hMDS patients were more likely to be age<65 (68%
vs. 47%, p<0.001) and have therapy-related MDS (t-MDS) (44% vs. 32%, p=.001). hMDS
patients also had a median overall survival (mOS) of 29.2 months, which was similar
to that observed in non-hMDS pts (HR 1.04, p=0.702). This finding was maintained after
multivariate adjustment including age, IPSS-R, t-MDS, and certain high-risk mutations
(TP53, ASXL1, RUNX1, RAS pathway). Hypocellularity did not affect survival in either
de novo or t-MDS. AML transformation rates (20% in both groups) were similar. Interestingly,
pts with hMDS had lower rates of high-risk mutations (38% vs. 51%, p=0.009) with less
RUNX1 (p=0.079) and ASXL1 (p=0.010) mutations while the frequency of TP53 and RAS
pathway mutations were comparable. In t-MDS, hMDS pts had less frequent TP53 (p=0.005)
or U2AF1 (p=0.087) mutations. In de novo MDS, hMDS patients had less RUNX1 (p=0.036),
SRSF2 (p=0.014), and TET2 (p=0.019) mutations. In hMDS, mutations in the RAS pathway
(HR 3.64, p=0.007) or in TP53 (HR 2.47, p=0.002) were associated inferior mOS, while
TET2 mutations (HR 0.29, p=0.036) correlated with superior mOS. hMDS also had worse
mOS in RAS pathway-mutated MDS (HR 2.62, p=0.043).
Based on mutation data and multivariate selection models, patients were stratified
into 3 groups: (1) pts with TET2 or SF3B1 (only available 81-gene panels) mutations,
(2) pts with TP53 mutations or RAS pathway mutations, and (3) pts without TET2/SF3B1/TP53/RAS
mutations. The TET2/SF3B1 group showed better mOS (HR 0.34, p=0.041) compared to the
TP53/RAS group showing inferior mOS (HR 4.64, p<0.001) (shown in figure). The mOS
difference observed with the TET2/SF3B1 group did not carry over into non-hMDS pts
(p=0.889).
Image:
Summary/Conclusion: Our study showed that hMDS patients have similar outcomes with
non-hMDS pts with unique molecular profiles. hMDS pts have less frequent high-risk
mutations with distinct risk stratification. Improving risk stratification tools in
hMDS by incorporating genomic data, such as TP53/RAS pathway and TET2/SF3B1 mutations,
would be a study of interest.
P770: A COMPARATIVE STUDY OF LEUKEMIC TRANSFORMATION IN THERAPY-RELATED AND DE NOVO
MYELODYSPLASTIC SYNDROME AFTER HYPOMETHYLATING AGENT FAILURE
K. Kim1,*, F. Ong1, Z. Li2, R. Kanagal-Shamanna3, G. Montalban-Bravo1, T. Kadia1,
E. Jabbour1, N. Pemmaraju1, D. Hammond1, N. Short1, F. Ravandi1, Y. Alvarado1, S.
Pierce1, X. Q. Dong1, H. Kantarjian1, G. Garcia-Manero1, K. Chien1
1Department of Leukemia; 2Department of Biostatistics; 3Department of Hematopathology,
The University of Texas MD Anderson Cancer Center, Houston, United States of America
Background: Therapy-related myelodysplastic syndrome (t-MDS) is subset of myelodysplastic
syndrome (MDS), known for having dismal survival outcomes. The majority of patients
(pts) with t-MDS have similar clinical courses with de novo MDS patients after hypomethylating
agent failure (HMA-F) that often results in transformation to acute myeloid leukemia
(AML).
Aims: We conducted a study to identify unique clinicopathologic features of t-AML
from t-MDS compared to secondary AML (sAML) from de novo MDS after HMA-F.
Methods: We identified sAML patients after HMA-F from all untreated MDS pts in a tertiary
cancer center registry from 2017-2021. Clinical characteristics and pathologic data
including cytogenetic (CG) and next generation sequencing (NGS) information were collected
at the time of MDS diagnosis (dx), HMA-F, and AML dx.
Results: 71 (48%) MDS pts with AML transformation after HMA-F were included in the
study with a median follow-up time of 15.4 months (mo). At the time of MDS dx, 31
(43%) pts had t-MDS: 97%, 58% and 29% had a history of chemotherapy, radiation, and
prior stem cell transplant (SCT), respectively. t-MDS patients were more likely to
present with age<65 (48% vs. 28%, p=0.070) and thrombocytopenia (33,000 vs. 75,500,
p=0.004) when compared to de novo MDS pts. The majority of t-MDS pts had very high
risk disease by IPSS-R (67% vs. 39%, p=0.031), while de novo MDS pts frequently had
high risk disease (50% vs. 20%, p=0.013). 90% of t-MDS pts had adverse CG (IPSS-R
CG score 3-4) at MDS dx, compared to 46% in de novo MDS pts (p=0.004). NGS at MDS
dx showed more frequent TP53 mutations in t-MDS than in de novo MDS (73% vs 36%, p=0.003).
Interestingly, ASXL1 (36% vs. 1%, p=0.001), RUNX1 (9% vs. 0%, p=0.004), STAG2 (21%
vs. 0%, p=0.013), TET2 (13% vs. 0%, p<0.001), and SRSF2 (18% vs. 0%, p=0.027) were
more frequently found in de novo MDS (Table). t-MDS had worse overall survival (OS)
from MDS dx (HR 1.82, p=0.026). The median OS from MDS dx was 19.4 mo and 14.3 mo
in de novo MDS and t-MDS, respectively.
The number of HMA cycles received before HMA-F was 6 (range: 1-13) in t-MDS and 5
(range: 1-23) in de novo MDS. At the time of HMA-F, t-MDS pts presented with more
anemia (p=0.005) and thrombocytopenia (p=0.064). The number of CG clones were similar,
but the number of CG abnormalities were higher in t-MDS pts (p=0.025). Among pts with
subsequent AML transformation after HMA-F, the median time from HMA-F to AML was 1.5
mo in t-MDS to AML, compared to 4.6 mo in de novo MDS to AML.
At time of AML dx, 48 (68%) pts had mutation panels available. All TP53 mutation status
stayed the same in t-MDS pts at the time of MDS and AML dx. Other changes in mutations
were shown in Table. Though the median OS from AML transformation was short in all
pts, t-MDS pts had inferior outcomes (2.4 mo vs. 5.0 mo, HR 1.70, p=0.043). The initial
response rate to salvage therapy was comparable between t-MDS and de novo MDS after
AML transformation (50% and 56%, p=0.761), but more pts underwent SCT with de novo
MDS compared to t-MDS (10% vs. 3%, p=0.383).
Image:
Summary/Conclusion: Transformation to AML after HMA-F results in poor outcomes, but
pts with t-MDS have worse survival with distinct CG and mutation profiles. Further
understanding of the pathogenesis of AML transformation in both t-MDS and de novo
MDS after HMA-F is warranted with improved therapy options an area of unmet need.
P771: PROGNOSTIC CHARACTERISTICS OF PATIENTS WITH MDS WITH ABERRATIONS OF CHROMOSOME
5. DATA FROM THE DÜSSELDORF MDS REGISTRY
F. Schulz1, J. Kostova1,*, B. Hildebrandt2, K. Nachtkamp1, C. Strupp1, N. Gattermann1,
M. Lübbert3, S. Blum4, S. Parmentier5, K. Götze6, J. Lipke7, F. Beier8, W.-K. Hofmann9,
U. Germing1
1Department of Hematology, Oncology and Clinical Immunology; 2Institute of Human Genetics,
University Hospital Düsseldorf, Düsseldorf; 3Department of Medicine I: Hematology,
Oncology, and Stem-Cell Transplantation, University Medical Center Freiburg, Freiburg,
Germany; 4Department of Hematology and Oncology, Lausanne University Hospital, Lausanne;
5Oncology and Hematology, Tumor Center, St. Claraspital, Basel, Switzerland; 6Department
of Internal Medicine III, University Hospital Klinikum Rechts der Isar, Munich; 7Gefos
Dortmund, Dortmund; 8Department of Hematology, Oncology, Hemostaseology and Stem Cell
Transplantation, University Hospital Aachen, Aachen; 9Department of Hematology and
Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
Background: Myelodysplastic syndromes (MDS) are hematopoietic stem cell disorders
with a wide spectrum of biological and clinical features. Up to 20% of MDS patients
present with aberrations of chromosome 5, either as isolated del(5q) or combined with
other aberrations. Patients with a medullary blast count < 5% and isolated del(5q),
or with one additional aberration except anomalies of chromosome 7, are diagnosed
as MDS del(5q), a defined entity according to the WHO 2016 classification. They have
a favorable prognosis compared to other types of MDS, although profound anemia, chronic
transfusion dependency and the risk of disease progression into AML present clinical
challenges.
Aims: We analyzed the prognostic impact of karyotype complexity in MDS del(5q).
Methods: From the Düsseldorf MDS registry we retrieved 499 patients with MDS or CMML
according to WHO 2016, who showed any kind of del(5q). Biological and clinical parameters
such as karyotype, IPSS-R and type of treatment were evaluated. Survival and progression
to AML was calculated using Kaplan-Meier-plots. Cox regression analysis was performed
to identify disease-related prognostic factors. Patients were followed up until Dec
31th 2021.
Results: 58% of the patients were female, median age was 67 years. Median medullary
blast count was 4% (0-19%), and median survival in the entire group was 28 months.
For assessing the natural course of disease, we screened for patients with non-intensive
treatment only (best supportive care, lenalidomide, low-dose Ara-C, hydroxyurea).
Separation by medullary blast count (< 5%, 5-9% or ≥ 10%) was associated with significantly
different median survival times, ranging from 63 months in the first group to 3 months
in patients with ≥ 10% medullary blasts (p < 0.001). Survival was also affected by
the cytogenetic risk group according to IPSS-R. Patients in the low-risk group (median
survival 72 months) differed significantly from patients in the very high risk group
(median survival 5 months) (p < 0.001). A similar trend was observed according to
karyotype complexity represented by number of aberrations apart from del(5q). Median
survival of patients with an isolated del(5q) was 86 months, while patients with a
complex karyotype (del(5q) and ≥3 additional aberrations) had a median survival of
5 months. Biological risk factors were assessed using Cox regression analyses. Considering
overall survival as endpoint, medullary blast percentage, cytogenetic risk group,
and complexity of the karyotype had a significant impact on survival (p < 0.001).
In the entire patient cohort, as well as among patients receiving non-intensive treatment,
the rate of AML progression was significantly influenced by karyotype complexity (p
< 0.001). While the median time to AML progression in both isolated del(5q) and del(5q)
combined with one aberration had not been reached at the end of the observation period,
the median time to AML progression for patients with del(5q) as part of a complex
karyotype was 31 months in the entire cohort and 14 months in the low-intensity treatment
group.
Image:
Summary/Conclusion: Both the IPSS-R cytogenetic risk group and the complexity of the
5q- karyotype showed prognostic impact in patients with del(5q). This was true for
overall survival as well as risk of AML progression. Regardless of therapy or medullary
blast count in patients with del(5q), the risk of AML progression increased dramatically
with increasing complexity of the 5q- karyotype.
P772: PROGNOSTIC IMPACT OF DISEASE-RELATED RISK FACTORS AND TREATMENT APPROACHES IN
PATIENTS WITH ADVANCED MYELODYSPLASTIC SYNDROMES (MDS)
C. Lesch1,*, A. Kündgen1, K. Nachtkamp1, A. Kasprzak1, C. Strupp1, M. Rudelius2, N.
Gattermann1, G. Kobbe1, B. Hildebrandt3, U. Germing1
1Department of Haematology, Oncology and Clinical Immunology, Heinrich-Heine University
Hospital, Düsseldorf, Duesseldorf; 2Institute of Pathology, Ludwig-Maximilians-University
of Munich, Munich; 3Institute of Human Genetics, Heinrich- Heine University Hospital,
Düsseldorf, Duesseldorf, Germany
Background: Myelodysplastic syndromes (MDS) are heterogeneous hematopoietic stem cell
disorders. Treatment approaches depend on the type of MDS according to the WHO classification
and on risk assessment according to the international prognostic scoring system (IPSS-R).
For patients with advanced MDS, as defined by an excess of blasts, therapeutic options
include hypomethylating agents and allogeneic stem cell transplantation.
Aims: To determine the prognostic impact of disease-related risk factors and treatment
approaches in 432 patients with MDS EB1 and EB2.
Methods: Patients included in this analysis were classified as having MDS EB1 (5-9%
marrow blasts) or MDS EB2 (10-19% marrow blasts) at diagnosis. All therapeutic procedures
prior to the development of acute myeloid leukemia (AML) were documented. Patients
were followed up until 31st December 2021, or until death. Therapeutic approaches
were categorized as best supportive care (BSC) only, low-dose chemotherapy, immunomodulatory
treatment, hypomethylating agents (HMA), intensive chemotherapy (IC) and allogeneic
stem cell transplantation (alloSCT). The mentioned therapy regimes were weighted in
different categories according to their intensity, with BSC defined as the therapy
with the lowest intensity and alloSCT as the one with the highest (BSC < low-dose-chemotherapy
< immunomodulatory treatment < HMA < IC < allo SCT).
Results: MDS EB1 was diagnosed in 182 patients (42,1%), and MDS EB2 in 250 patients
(57,9%). Distribution among IPSS-R risk groups was as follows: 2% low risk, 26.3%
intermediate risk, 38% high risk, and 33,7% very high risk. Median life expectancy
in the entire cohort including patients with all types of treatment, was 22 months.
Progression to AML occurred in 186 (43%) of the patients.
Patients receiving supportive care only (42.6%) had a median survival of 13 months.
Patients treated with a hypomethylating agent (18%) showed an overall survival of
26 months, which was significantly better compared with best supportive care (p=0.002).
Patients undergoing induction chemotherapy (6%) had a median survival of 21 months,
comparable with HMA treatment. Patients receiving low-dose chemotherapy (4.6%) had
a median survival of 9 months. The longest median survival (131 months) was seen in
patients undergoing alloSCT.
Multivariate Cox regression analysis showed that treatment category (p<0.00005), together
with cytogenetic risk category according to IPSS-R (very low, low and intermediate
versus high versus very high) (p<0.0005), and hemoglobin (<≥10g/dl) (p=0.003) were
the only parameters influencing survival independently, whereas WHO type, gender,
ANC, and platelets did not show independent influence on survival.
Summary/Conclusion: Stratification according to cytogenetic risk groups is very important
for the prognostic assessment of MDS patients. Assignment to the cytogenetic group
high risk and very high risk has greater prognostic impact than the medullary blast
count. Even though hypomethylating agents can improve overall survival compared to
best supportive care, long-term survival can only be achieved through allogeneic stem
cell transplantation. Therefore, all patients eligible for intensive treatment should
be evaluated for this treatment option at the time of diagnosis. Furthermore, in recognition
of the fact that clonal evolution may occur in MDS patients, consideration should
be given to repeating the chromosome analysis as the most important prognostic factor
at regular intervals.
P773: THE ELEVATION OF RED BLOOD CELL DISTRIBUTION IS AN INDEPENDENT PROGNOSTIC FACTOR
FOR JUVENILE MYELOMONOCYTIC LEUKEMIA
W. Liang1,*, C. Liu2, J. Zhang2, M. Yi2, Y. Cai2, A. Zhang2, L. Liu2, L. Zhang2, X.
Chen2, Y. Zou2, Y. Chen2, Y. Guo2, Y. Zhang2, X. Zhu2, W. Yang2
1Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China;
2Department of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical
College, Tianjin, China
Background: Juvenile myelomonocytic leukemia (JMML) is an overlapping myeloproliferative
and myelodysplastic disorder of infants and early children. The existing clinical
and laboratory prognostic features are inadequate to predict a worse outcome alone.
New prognostic indicators are urgently needed since the great clinical heterogeneity.
Red cell distribution width (RDW) is a parameter of the complete blood count (CBC)
measured automatically by haematology analyzers to reflect the heterogeneity of erythrocyte
size. In recent years, RDW has been suggested to have a high negative value for outcomes
in various cancers. However, the role of RDW at diagnosis in patients with JMML is
still unclear.
Aims: Our aim is to investigate the prognostic role of RDW in children with JMML.
Methods: We conducted a single center retrospective study to investigate the prognostic
role of RDW in children with JMML. Seventy-seven patients were enrolled from January
1, 2008 to November 31, 2019. Survival rates were calculated and compared using Kaplan-Meier
methods and log-rank tests. Univariate and Multivariate analysis was carried out according
to the Cox proportional hazard regression model.
Results: The mean red cell distribution width standard deviation (RDW-SD) for patients
is 53.3(39.3-73.7) fl. Cox proportional hazard models showed that patients with RDW-SD
>50.50fl at diagnosis were significantly associated with poor overall survival (OS)
(HR = 5.22, CI = 1.50-18.21, P=0.010). The multivariate analysis demonstrated that
the RDW-SD count was the independent risk factor for OS. Patients with PTPN11 mutation
and RDW elevation displayed the worst survival rate, in comparison with other subgroups
(P=0.00013).
Summary/Conclusion: The results suggested that the elevated level of RDW is an independent
prognostic factor for JMML. Furthermore, the PTPN11 mutation and a high RDW level
may involve a synergy during the natural course of JMML.
P774: CLINICAL OUTCOMES ASSOCIATED WITH AZACITIDINE MONOTHERAPY FOR TREATMENT-NAIVE
PATIENTS WITH HIGHER-RISK MYELODYSPLASTIC SYNDROME (HR MDS): A SYSTEMATIC LITERATURE
REVIEW AND META-ANALYSIS
K. Hasegawa1,*, A. H. Wei2, G. Garcia-Manero3, N. G. Daver3, N. Rajakumaraswamy1,
S. Iqbal1, R. J. Chan1, H. Hu1, P. Tse4, J. Yan4, M. J. Zoratti4, F. Xie4, D. A. Sallman5
1Gilead Sciences, Inc., Foster City, United States of America; 2Department of Clinical
Haematology, The Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne,
Australia; 3The University of Texas MD Anderson Cancer Center, Houston, United States
of America; 4McMaster University, Hamilton, Canada; 5Moffitt Cancer Center, Tampa,
United States of America
Background: Azacitidine (AZA) is a standard of care for patients with HR MDS; however,
the number of large data sets describing outcomes of AZA monotherapy in HR MDS is
limited.
Aims: To aggregate clinical outcomes data associated with AZA monotherapy for treatment-naive
patients with HR MDS.
Methods: CENTRAL, EMBASE, and MEDLINE were searched to identify interventional, prospective,
and retrospective observational studies using AZA monotherapy in treatment-naive HR
MDS (defined as intermediate-2 or high risk by International Prognostic Scoring System
[IPSS] or intermediate to very high risk by Revised IPSS). Inclusion of observational
studies was limited to those with >20 patients in the AZA monotherapy arm. Responses
according to International Working Group (IWG) 2000 or IWG 2006 criteria—including
complete remission (CR) rate, overall response rate (ORR; defined as CR, marrow CR
[mCR], partial remission, and hematologic improvement), overall survival (OS), duration
of response (DOR), and time to response (TTR)—were extracted. All articles were reviewed
by 2 independent abstractors. Response rates were synthesized using random-effects
models; median of medians was used for OS, DOR, and TTR.
Results: Of 3250 abstracts identified, 34 publications describing 16 studies met inclusion
criteria: 5 randomized controlled trials (RCTs), 3 prospective studies, and 8 retrospective
studies. None of the response endpoints were reported in all studies (Table). The
pooled CR rates, reported in 2 RCTs (N=55), 1 prospective study (N=27), and 3 retrospective
studies (N=509), were 14%, 11%, and 16%, respectively. The pooled CR rate across all
studies (N=591) was 16% (95% CI, 13%-19%). The mCR rate from 1 RCT was 19% (95% CI,
10%-34%; N=42), and the pooled mCR rate from 2 retrospective studies was 18% (95%
CI, 13%-26%; N=126). The pooled ORRs ranged from 44% to 55% across RCTs, prospective
studies, and retrospective studies.
The pooled median OS was 16.7 months in 3 RCTs (N=161), 16.5 months in a single prospective
study (N=34), and 14.4 months in 5 retrospective studies (N=1472). The pooled median
OS across all studies (N=1667) was 16.4 months (95% CI, 12.0-17.3 months).
The pooled median DOR was 10.1 months (95% CI, 9.1-11.0 months), and the median TTR
was 4.6 months (95% CI, 3.0-9.0 months).
Table:
Outcomes in treatment-naive patients with HR MDS treated with AZA monotherapy
Endpoint
RCTs
Total N
Outcome(95% CI)
Prospectivestudies
Total N
Outcome(95% CI)
Retrospective studies
Total N
Outcome(95% CI)
All studies
Total N
Outcome(95% CI)
CR rate, %
2
55
14(6-31)
1
27
11(4-29)
3
509
16(13-20)
6
591
16(13-19)
mCR rate, %
1
42
19(10-34)
0
-
-
2
126
18(13-26)
3
168
19(13-25)
ORR, %
3
159
50(42-57)
1
27
44(27-63)
4
967
55(42-67)
8
1153
52(44-60)
Median OS,
months
3
161
16.7(9.5-26.3)
1
34
16.5(10.4-21.9)
5
1472
14.4(11.6-17.3)
9
1667
16.4(12.0-17.3)
Median DOR,
months
1
20
9.1(6.0-NR)
0
-
-
1
405
11.0 (NA)
2
425
10.1(9.1-11.0)
Median TTR,
months
1
42
4.6(2.0-6.5)
0
-
-
2
493
6.0 (3.0-9.0)
3
535
4.6(3.0-9.0)
NA, not available; NR, not reached.
Summary/Conclusion: These findings provide evidence that benefit with AZA monotherapy
in treatment-naïve patients with HR MDS is limited. Opportunity exists for novel therapies
to increase CR rates and prolong survival.
P775: CLINICAL OUTCOMES AND SOCIOECONOMIC FACTORS IN CHRONIC MYELOMONOCYTIC LEUKEMIA
IN THE UNITED STATES USING THE NATIONAL CANCER DATABASE: 2004-2017
N. Mclaughlin1,*, G. Ruan2, K. Wudhikarn3, A. Al-Kali2, A. Tefferi2, N. Gangat2, R.
Go2, M. Patnaik2, M. Shah2
1Division of Internal Medicine; 2Division of Hematology; 3Mayo Clinic, Rochester,
United States of America
Background: Chronic myelomonocytic leukemia (CMML) is a rare myeloid malignancy characterized
by proliferation of dysplastic monocytic cells and carries a risk for leukemic transformation.
Previously, we described the incidence and outcomes of CMML at the population level
(2004-2015) using the Surveillance, Epidemiology, and End Results Program (SEER) and
National Cancer Database (NCDB) (Ruan et al., ASH 2020). We studied population-based
outcomes in CMML using the NCDB from 2004-2017.
Aims: Our aim was to understand the impact of social determinants of health on outcomes
in CMML. Secondly, we studied if the outcomes of CMML have improved.
Methods: The NCDB is a nationwide oncology outcomes database for >1500 cancer programs
in the US and Puerto Rico, capturing 70% of all newly diagnosed cases of cancer. We
queried the NCDB using the ICD-O-3 diagnosis code 9945/3 from 2004-2017. Only patients
(pts) with diagnostic confirmation via histopathologic examination were included.
Percent without high school degree (HSD) reflects the education level within the pt’s
zip code, which was stratified into 4 quartiles. Median household income was estimated
by matching the pt’s zip code at time of diagnosis against US Census 2000 data, which
was stratified into 4 quartiles. Hazard ratios (HR) with confidence intervals (CI)
were calculated using a Cox proportional hazards model. Overall survival (OS) was
estimated using the Kaplan-Meier method. Variables significant in univariable analysis
were included in a multivariable analysis.
Results: We identified 7143 CMML pts in the NCDB. The median age at diagnosis was
72 years (interquartile range [IQR] 65-81) and 4386 (61%) were males. With a median
follow up of 6.1 years (IQR 3.6-9.4), the median OS was 1.4 years (IQR 0.4-3.8). 1600
(23%) had private insurance, 4862 (70%) had Medicare, 298 (4%) had Medicaid, and 129
(2%) were uninsured. In univariate analysis, private insurance had an improved OS
compared to patients with Medicaid (HR 1.2, 95% CI 1.06-1.4; p value 0.007), Medicare
(HR 1.5, 95% CI 1.4-1.6; p value < 0.001), and other government insurance (HR 1.5,
95% CI 1.1-2.0; p value 0.004). Pts living in a zip code with percent of adults without
an HSD of < 14% had an improved OS compared to zip codes with > 29% (HR 1.2, 95% CI
1.1-1.3; p value < 0.001). Median household income by zip code of ≥ $46,000 had an
improved OS compared to < $30,000 (HR 1.2, 95% CI 1.1-1.4; p value < 0.001). Receiving
treatment at an academic program had an improved OS compared to community cancer programs
(HR 1.2, 95% CI 1.1-1.3; p value < 0.001), comprehensive community cancer programs
(HR 1.3, 95% CI 1.3-1.4; p value < 0.001), and integrated network cancer programs
(HR 1.3, 95% CI 1.2-1.4; p value < 0.001).
Finally, pts diagnosed in 2012-2017 had an improved OS compared to pts diagnosed in
2004-2011 (HR 0.8, 95% CI 0.8-0.9; p value < 0.001). Variables significant in univariate
analysis were included in multivariate analysis (Table 1). When education level and
median household income per zip code were included in multivariate analysis, education
level remained statistically significant and income did not.
Image:
Summary/Conclusion: CMML continues to have a poor prognosis, but OS has improved in
recent years. Socioeconomic factors including insurance status, level of education
and median household income per zip code, and facility type appear to be associated
with outcomes. On multivariable analysis, age < 65, private insurance (vs government
insurance), living in a zip code with increased percent HSD, and treatment at an academic
program had improved outcomes.
P776: A PHASE 2, OPEN-LABEL, ASCENDING DOSE STUDY OF KER-050 FOR THE TREATMENT OF
ANEMIA IN PATIENTS WITH VERY LOW, LOW, OR INTERMEDIATE RISK MYELODYSPLASTIC SYNDROMES
S. Tan1,*, A. Arbelaez2, L. Chee3, C. Y. Fong4, D. Hiwase5, G. Kannourakis6, J. Kwan7,
J. Liang8, A. Puliyayil9, H. Rose10, D. Ross11, T.-C. Teh12, D. Westerman13, J. Wight14,
W. Feng15, J. Lachey15, A. McGinty15, H. Natarajan15, C. Rovaldi15, S. Cooper15
1St Vincent’s Hospital, Melbourne; 2Tweed Hospital, Tweed Heads; 3Royal Melbourne
Hospital and Peter MacCallum Cancer Centre, Melbourne; 4Austin Health, Hiedleberg;
5Royal Adelaide Hospital, Adelaide; 6Ballarat Oncology, Ballarat; 7Westmead Hospital,
Westmead, Australia; 8Middlemore Hospital, Auckland, New Zealand; 9Albury Wodonga
Health, Albury-Wodonga Regional Cancer Centre, Auckland; 10Barwon Health, Geelong;
11Flinders Medical Centre, Adelaide; 12Box Hill Hospital, Eastern Health, Victoria;
131. Peter MacCallum Cancer Centre. 2. University of Melbourne, Melbourne; 14Townsville
University Hospital, James Cook University, Douglas, Australia; 15Keros Therapeutics,
Lexington, United States of America
Background: Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis;
many patients develop high transfusion burden (HTB) requiring ≥4 units RBCs every
8 weeks and have high unmet medical need. KER-050, an investigational modified activin
receptor type IIA inhibitor, is designed to target TGF-β ligands to promote differentiation
of erythroid and megakaryocyte lineages to improve anemia and thrombocytopenia.
Aims: Evaluate safety, tolerability, pharmacodynamics (PD), efficacy of KER-050 in
MDS in an open-label Phase 2 study.
Methods: IPSS-R lower-risk anemic MDS patients are enrolled, including non-transfused,
low-transfusion burden (LTB) and HTB patients. In Part 1 base study, ascending dose
cohorts receive KER-050 subcutaneously q4w for 4 doses until a recommended Part 2
dose is determined. Safety endpoints include incidence of adverse events (AEs); erythroid
efficacy endpoints (≥8 weeks duration) include rates of transfusion independence (TI)
and reduction in RBC transfusions by ≥4 units (IWG 2006 HI-E) in HTB patients. Safety-set
includes patients receiving ≥1 dose. PD/efficacy results are reported in efficacy
evaluable (EE) patients who contributed ≥8 weeks of HGB and transfusion data. New
analyses presented here describe response and markers of erythropoiesis and thrombopoiesis
in HTB patients.
Results: At data cut-off (25-Oct-2021), median follow-up was 124 days (range 10-217).
24 patients received ≥1 dose of KER-050 across 4 dose levels (0.75mg/kg to 3.75mg/kg
q4W). Of these, 16 were EE (8 not EE due to: duration of treatment (n=6), death (n=1),
study discontinuation (n=1)). Median time from diagnosis was 2.2 years (range 0.2
– 8.6), 14 (58.3%) were HTB. Of HTB patients, 7 were RS+, 6 were on iron chelators.
No DLTs were reported in the safety-set. 5 (20.8%) reported grade ≥3 treatment-emergent
AEs (TEAEs), 5 (20.8%) reported SAEs (none related), 4 (16.7%) developed grade 1 or
2 treatment-related AEs. Most frequent TEAEs (≥10%) were diarrhea, dyspnea, fatigue,
nausea, anemia, headache. 2 discontinued study drug: 1 participant decision, 1 death.
1 required dose modification due to unrelated TEAE; none required dose modification
for increased HGB or thrombocytosis.
Results from 16 EE cohort 1-3 patients were presented at ASH 2021 (Poster #3675),
8 (50%) achieved HI-E or TI. New analyses focused on HTB patients found that 6 of
9 HTB, EE patients achieved HI-E and 4 achieved TI for ≥8 weeks (Panel A). Increases
in reticulocytes (RETs) (Panel B) and sTfR and decreases in serum ferritin (Panel
C) in these HTB patients were observed. Mean maximum RETs increase was 56.87 x109/L,
range 15.7-142.5x109/L with mean maximum ferritin reduction of 29.7%, range -17 to
65% and mean maximum sTfR increases of 51%, range 29-90%. Increases in platelets were
observed in HTB patients achieving HI-E or TI (Panel D). Maximum increase from baseline
was 98 x109/L (mean), range 33-179 x109/L.
Image:
Summary/Conclusion: KER-050 was generally well-tolerated as of data cut-off date.
HI-E and TI ≥8 weeks have been observed in HTB patients treated with KER-050. Observed
PD effects in RETs, sTfR and ferritin support the proposed mechanism of action of
increasing erythropoiesis. Increases in platelets have been observed in HTB patients
achieving HI-E or TI which supports the potential of KER-050 as a treatment for multilineage
cytopenias in difficult-to-treat HTB patients. Dose escalation is ongoing in this
Phase 2 study of anemic patients with MDS; updated data from Part 1 with safety, PD
and efficacy data from cohorts 4 and 5 will be presented for the first time at the
meeting.
P777: RUNX1 VARIANTS WITH HIGH VARIANT ALLELE FREQUENCY IN MYELOID NEOPLASMS. GERMLINE
OR NOT?
N. J. Nitschke1,2,*, M. Krogh3, K. Raaschou-Jensen4, M. T. Severinsen5, A. S. Roug6,
J. S. Jespersen2, J. W. Hansen1,2, J. Weischenfeldt2, M. K. Andersen7, K. Grønbæk1,2
1Department of Hematology, Rigshospitalet; 2Biotech Research and Innovation Center
(BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen;
3Department of Hematology, Sjælland University Hospital, Roskilde; 4Department of
Hematology, Odense University Hospital, Odense; 5Department of Hematology and Clinical
Cancer Research, Aalborg University Hospita, Aalborg; 6Department of Clinical Medicine,
Aarhus University Hospital, Aarhus; 7Department of Clinical Genetics, Rigshospitalet,
Copenhagen, Denmark
Background:
RUNX1 is a transcription factor frequently mutated in myeloid neoplasms e.g. 10% of
acute myeloid leukemia (AML) patients, but RUNX1 variants detected in tumor tissue
at a variant allele frequency (VAF) close to 50% can potentially be germline. As pathogenic
germline RUNX1 variants are causative for RUNX1 Familial Platelet Disorder with Associated
Myeloid Malignancies it is crucial to distinguish between somatic and germline variants.
Previous studies in AML patients estimate the frequency of germline RUNX1 variants
to be approximately 3%. The frequency of RUNX1 germline variants in lower risk myeloid
neoplasms (MN) such as myelodysplastic syndrome (MDS), chronic myelomonocytic leukemia
(CMML), clonal cytopenia of undetermined significance (CCUS) and idiopathic cytopenia
of undetermined significance (ICUS) remains unknown.
Aims: To estimate the frequency of RUNX1 germline variants in MN.
Methods: Patients referred to a hematological department in Denmark with suspicion
of MN (excluding myeloproliferative disorders) were included in the study. At first
visit, a skin biopsy was drawn and frozen dry at -80 °C. RUNX1 variants were identified
in either blood or bone marrow using 4 different amplicon based next generation sequencing
panels. Variants detected with a VAF > 40 % were further classified as somatic or
germline by bi-directional sanger sequencing of germline DNA purified from skin biopsies.
Results: Among 595 patients, 85 (14%) had variants in RUNX1 and 28 patients (5%) had
variants with a VAF above 40%. High VAF RUNX1 variants were either missense, frameshift,
nonsense, or splice-site variants. A majority of the variants were located in the
runt homology domain (RHD). We observed a higher proportion of patients with AML and
a lower proportion of patients with CCUS in patients with a RUNX1 variant compared
to patients without a RUNX1 variant, and this tendency was enhanced if the RUNX1 variants
were present at VAF > 40%.
Among 22 patients with a high VAF RUNX1 variant and available germline DNA two variants
(9%) were classified as germline (NM_001754.5; c.668A>G, c.649G>A, minimal allel frequency
reported in gnomAD; 0.012%, 0.003%). The c.668A>G variant was detected in a patient
with a low platelet count and CCUS diagnosed at age 24. The c.649G>A variant was detected
in a patient with a normal platelet count and MDS diagnosed at age 64. Both germline
variants were missense variants located in the region between the RHD and the transactivating
domain (TAD) and both were predicted to be pathogenic in-silico by several prediction
tools. Unfortunately, family history wasn’t available for any of the two patients
with germline variants.
Pathogenic variants co-occurring at similar VAF as RUNX1 were detected in 17 out of
20 patients with a somatic RUNX1 variant, but not in any of the patients with germline
variants (Figure 1). Pathogenic variants in SRSF2, TET2 and ASXL1 were frequently
observed to co-occur with high VAF RUNX1 variants.
Image:
Summary/Conclusion:
RUNX1 variants with a VAF above 40% were classified as germline in two out of 22 MN
patients (9%). The variants were detected in patients with MDS and CCUS indicating
that germline testing should be considered not only in AML, but also in other MN with
high VAF of a RUNX1 variant. Furthermore, a high VAF RUNX1 variant without co-occurring
variants with similar VAF in other genes should increase the consideration for germline
testing.
P778: LONG-TERM UTILIZATION AND BENEFIT OF LUSPATERCEPT IN PATIENTS (PTS) WITH LOWER-RISK
MYELODYSPLASTIC SYNDROMES (LR-MDS) FROM THE MEDALIST TRIAL
P. Fenaux1,*, V. Santini2, R. S. Komrokji3, A. Zeidan4, G. Garcia-Manero5, R. Buckstein6,
D. Miteva7, K. Keeperman8, N. Holot8, J. Zhang8, J. A. Nadal7, B. Rosettani7, A. Yucel8,
U. Platzbecker9
1Service d’Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux
de Paris and Université Paris 7, Paris, France; 2MDS Unit, AOU Careggi, University
of Florence, Florence, Italy; 3Moffitt Cancer Center, Tampa; 4Department of Internal
Medicine, Yale School of Medicine and Yale Cancer Center, New Haven; 5Department of
Leukemia, The University of Texas MD Anderson Cancer Center, Houston, United States
of America; 6Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Canada;
7Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland;
8Bristol Myers Squibb, Princeton, United States of America; 9Medical Clinic and Policlinic
1, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
Background: Luspatercept has been shown to improve anemia in the phase 3 MEDALIST
trial of pts with LR-MDS ineligible/intolerant or refractory to erythropoiesis-stimulating
agents (ESAs).
Aims: To report the long-term clinical value of luspatercept treatment (Tx) in pts
from the MEDALIST study including dosing, duration of Tx (DOT) and response, baseline
characteristics of pts receiving luspatercept in a long-term follow-up (LTFU) study,
and rates of progression to acute myeloid leukemia (AML) and high-risk MDS (HR-MDS).
Methods: Eligible pts were ≥18 y of age, had LR-MDS requiring regular red blood cell
(RBC) transfusions, and were ineligible/intolerant or refractory to ESAs. Pts were
randomized 2:1 to subcutaneous luspatercept or placebo every 3 wk for 24 wk. The primary
endpoint was RBC transfusion independence (RBC-TI) ≥8 wk during wk 1–24. MEDALIST
pts who continued to receive luspatercept due to ongoing clinical benefit were eligible
for enrollment into the LTFU study, a phase 3b rollover study to evaluate the long-term
safety of luspatercept in pts who participated in other luspatercept studies. DOT
was calculated as (Tx end date − date of first dose) + 1) / 7. Duration of response
was determined by Kaplan–Meier analysis. Total person-years for pts at risk of HR-MDS/AML
progression was calculated from LR-MDS diagnosis to HR-MDS/AML diagnosis, or to last
HR-MDS/AML follow-up date for pts who did not progress.
Results: As of Jan 15, 2021, the median (95% confidence interval [CI]) DOT was 11.70
(8.97–16.33) mo for luspatercept pts and 5.52 (5.52–5.59) mo for placebo pts. Median
(interquartile range [IQR]) DOT for placebo pts who transitioned to LTFU was 24.0
(24.0–25.3) wk and 24.0 (24.0–39.0) wk for placebo pts who discontinued prior to LTFU.
The median (IQR) DOT for luspatercept pts who transitioned to LTFU was longer than
for those pts who discontinued (190.0 [86.0–211.0] wk vs 31.0 [24.0–64.1] wk). Baseline
characteristics more frequently observed in luspatercept pts who transitioned to LTFU
than in pts who discontinued include younger age, female sex, and lower Eastern Cooperative
Oncology Group performance status score, transfusion burden, erythropoietin, and serum
ferritin levels (Table A). In MEDALIST, 106/153 (69.3%) pts receiving luspatercept
and 64/76 (84.2%) receiving placebo escalated to the maximum dose of 1.75 mg/kg. During
the entire Tx phase, RBC-TI ≥8 wk was observed in 74/153 (48.4%) and 12/76 (15.8%)
luspatercept and placebo pts, respectively, with a median (95% CI) cumulative duration
of response of 80.7 (53.71–154.14) wk and 21.0 (10.86–NE) wk, respectively. During
the entire Tx period, RBC-TI ≥16 wk was observed in 48/153 (31.4%) and 6/76 (7.9%)
luspatercept and placebo pts, respectively (Table B). Among luspatercept pts, 13/153
(8.5%) progressed to HR-MDS/AML during the entire Tx period, compared with 5/76 (6.6%)
placebo pts. The total person-years for pts randomized to luspatercept at risk of
progressing to HR-MDS/AML was 401.7 y vs 190.9 y for placebo.
Image:
Summary/Conclusion: Pts receiving luspatercept had an extended period of clinical
benefit and >50% of pts continued to receive luspatercept for >1 y, the majority of
whom underwent dose escalations to achieve an optimal response. Pts experienced durable
responses with luspatercept, with a median cumulative duration of RBC-TI response
of approximately 20 mo. Pts receiving luspatercept also remained on Tx longer than
placebo regardless of participation in the LTFU study, suggestive of luspatercept’s
clinical benefit with significant durability.
P779: OPTIMIZING TREATMENT OUTCOMES FOR CHILDREN WITH HIGHER-RISK MYELODYSPLASTIC
SYNDROME UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION
Y. Ren1,*, F. Liu1, X. Chen1, X. Li1, L. Liu1, W. Yang1, Y. Guo1, X. Zhu1
1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences
& Peking Union Medical College, Tianjin, China
Background: Myelodysplastic syndrome (MDS) are rare clonal hematopoietic disorders
in children. Risk stratification system for adults is unfit for children. Refractory
anemia with excess blasts(RAEB), acute myeloid leukemia with myelodysplasia-related
changes(AML-MRC), monosomy 7 and complex karyotype always indicate poor prognosis.
Hypomethylating agent (HMA) improved survivals of high-risk MDS in adults, and decitabine
as part of conditioning regimen for allogenetic hematopoietic stem cell transplantation(allo-HSCT)
was associated with favorable outcomes. However, these findings have not been tested
in pediatric cohorts broadly.
Aims: To evalute the efficacy and safety of hypomethylating agents as a part of pre-transplant
treatments and contidioning regimens in children with higher-risk MDS.
Methods: Patients from 1 to 14 years of age who were diagnosed with higher-risk MDS
and underwent allo-HSCT consecutively between February 2019 and August 2020 in Pediatric
Blood Diseases Center were enrolled. All identified patients were retrospectively
reviewed with a collection of clinical data, laboratory test results, and outcome.
Results: We reviewed 18 pediatric patients with higher-risk MDS, including 7 with
RAEB, 8 with AML-MRC, and 3 with refractory cytopenia of childhood with monosomy 7.
Regardless of initial diagnosis, monosomy 7 is the most common cytogenetic aberration
(8/18, 44.4%). Next-generation sequencing (NGS) was performed on 15 specimens and
the most frequent detection of mutation gene was SETBP1, followed by ETV6. Before
transplantation, ten patients received monotherapies or combined chemotherapies with
HMA, two patients received AML-like chemotherapies, and nine of those achieved bone
marrow complete remission. The stem cell sources came from matched sibling donor (n=1),
haploidentical donor (n=11), and unrelated cord blood (n=6), respectively. Low-dose
decitabine-containing myeloablative conditioning regimens were administered in all
cases. About 50% of patients (n=9) experienced Grade II- IV acute graft versus host
diseases post-transplant. The cumulative incidence of transplantation-related mortality
(TRM) was 11.1%. During a median follow-up of 10.5 (range, 2–36) months after transplantation,
15 of 18 patients were alive. At time of last follow-up, all survivors had resolution
of hematologic disorder without relapse.
Summary/Conclusion: In summary, administration of hypomethylating agents to allogeneic
SCT resulted in excellent outcomes in children with higher-risk MDS.
P780: COMBINATION THERAPY OF ELTROMBOPAG AND CYCLOSPORINE A CAN IMPROVE HEMATOPOIESIS
IN PATIENTS WITH LOWER RISK MYELODYSPLASTIC SYNDROME
L. Song1,*, C. Chang1
1Hematology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai,
China
Background: Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal myeloid
disorders characterized by ineffective hematopoiesis and cytopenias, with variable
risks of progression to acute myelogenous leukemia (AML). Peripheral blood cytopenias
(thrombocytopenia, leucopenia, and anemia) are the major sources of morbidity and
mortality for lower-risk MDS (LR-MDS) patients. Among the cytopenias, thrombocytopenia
is a severe complication of ineffective hematopoiesis and is associated with poor
overall survival.Eltrombopag (EPAG) is an orally administered small-molecule, nonpeptide
thrombopoietin (TPO) receptor agonist, C-mpl is a TPO receptor expressed on megakaryocytes,
HSCs, and progenitor cells. The interaction between EPAG and megakaryocytes regulates
platelet production, maturation, and release through the binding of C-mpl on megakaryocytes.
Aims: The above studies provided a rationale for the use of the combination of EPAG
and CysA in the treatment of LR-MDS. In this study, we investigated the efficacy and
safety of the combination of EPAG and CysA in LR-MDS in a non-randomized phase II
investigator-initiated trial (IIT).
Methods: Herein, a non-randomized phase II investigator-initiated trial was conducted
to investigate the efficacy and safety of the combination of EPAG and cyclosporine
A (CysA) in LR-MDS. EPAG dose was escalated from 50 mg/day to a maximum of 100 mg/day.
The dose of CysA is 3–5 mg/kg/d to maintain serum trough concentration at 200 mg/mL.
Results: All the 31 patients were evaluated for response at the primary endpoint.
Among them, 21 (67.7%) patients achieved treatment response in at least one eligible
lineage with an objective response rate (ORR), 4 (12.9%) patients achieved a CR, 5
(16.1%) patients achieved bi-lineage responses (HI-PE R), 11 (24.3%) patients achieved
platelet lineage responses (HI-P R), and 1 (3.2%) patient achieved erythroid lineage
response (HI-E R). The median time to response was 8 (range, 2–24) weeks. Of the 21
patients eligible for an erythroid lineage response (RBC transfusion-dependent or
Hb <90 g/L),10 patients (47.6%) had responded by the endpoint and the platelet response
rate was 64.5% (20/31). The median increase in Hb levels and platelets was from 60.4±18.2 g/L
to 68.0±21.2 g/L (P=0.015) and from 12.3±11.2×109/L to 32.5±37.3×109/L (P=0.003).
The median DORs of HI-E and HI-P were not achieved, but the response rates of erythroid
and platelet lineages in 20 months were 80.0% and 70.6%, respectively. The RBC and
platelet transfusion-dependent rate decreased from 42.9% (9/21) to 28.6% (6/21) (X2=0.933,P=0.334)
and from 58.0% (18/31) to 25.8% (8/31) (X2=6.624, P=0.010). respectively. The median
time to response is 8 weeks.
All 31 patients were evaluated for toxicity, and the treatment was found to be well-tolerated.
The most commonly reported adverse events (AEs) of any grade were nausea (6/31,19.4%),
elevated liver transaminases (4/31,12.9%), and headache (3/31,9.7%); no severe AEs
had been reported until the cutoff date.
Summary/Conclusion: In conclusion, our results indicated that the addition of Epag
to CysA is well-tolerated and is an effective treatment for patients with low to intermediate-1
risk MDS. These response rates are better than CysA monotherapy, especially HI-P.
Also, the hematological responses are durable, although continued Epag administration
might be necessary. Further large prospective and controlled studies are warranted
to define the role of this combination treatment in LR-MDS in comparison to CysA
P781: CLINICAL OUTCOMES AND HEALTHCARE RESOURCE UTILIZATION (HCRU) IN PATIENTS WITH
LR-MDS REINITIATING ESAS FOLLOWING PREVIOUS ESA TREATMENT
G. Garcia-Manero1,*, R. K. Matsuno2, A. McBride3, T. D. Brown2, D. Idryo2, R. Broome2,
A. Herriman2, T. Johnson2, K. Wilkinson2, S. Walters2, A. Schrag2, C. Johanson2, M.
Izano2, S. Mukherjee4
1MD Anderson Cancer Center, Houston; 2Syapse, San Francisco; 3Bristol Myers Squibb,
Princeton; 4Cleveland Clinic, Cleveland, United States of America
Background: The median survival for patients with lower-risk myelodysplastic syndromes
(LR-MDS) is estimated to be 5–10 years. Erythropoiesis-stimulating agents (ESAs) are
utilized as first-line treatment for anemia in LR-MDS; however, real-world treatment
patterns, clinical outcomes, and healthcare resource utilization (HCRU) for patients
with LR-MDS who reinitiated ESAs following previous discontinuation are not well-established.
Aims: To examine treatment patterns, clinical outcomes, and HCRU in patients with
LR-MDS who reinitiated treatment with ESAs.
Methods: This was a retrospective study of patients with LR-MDS in a network of US
community health systems who initiated ESAs as first-line therapy between January
1, 2016 and June 30, 2019 and were followed through June 30, 2021. Patients were required
to have discontinued (had a ≥ 3-week gap between treatment with epoetin alfa, or a
≥ 6-week gap between treatment with darbepoetin alfa) and subsequently reinitiated
ESA therapy at least once during follow-up. Outcomes included failure to achieve transfusion
independence (TI), progression to acute myeloid leukemia (AML), overall survival (OS),
number and type of health system visits, and medication use. TI was defined per IWG
2006 criteria in the subset of patients who had received ≥ 1 transfusion in the 8
weeks prior to ESA initiation.
Results: There were 108 patients with confirmed LR-MDS who initiated ESA-based therapy.
Patients had a median age of 79 years at diagnosis (interquartile range [IQR] 73–85),
were predominantly male (58%), White (97%), and overweight or obese (52%). The median
follow-up period was 17.0 months (IQR 7.2–34.7). The most used ESA was darbepoetin
alfa (60%), followed by epoetin alfa (38%) and epoetin alfa-epbx (2%). Of the 33 patients
who received a baseline transfusion, 52% did not achieve TI; 53% and 43% failed to
achieve TI at 6 and 12 months, respectively. The majority of patients were repeatedly
treated with ESAs (n = 61, 56%). Among these patients, 2% progressed to AML and 48%
died during follow-up; median OS was 45 months. The median number of health system
visits per patient-month was 3.2 (IQR 0.6–4.9); 89%, 62%, and 72% of pts had ≥ 1 outpatient,
emergency department, or inpatient visit respectively, with median length of hospitalization
of 12 days (IQR 4.8–27.8). Most patients (54%) received an antibiotic at least once
during follow-up; overall, use of immunosuppressive therapy (2%) or iron chelation
(3%) was rare.
Summary/Conclusion: In this real-world study of community practice in the USA, reinitiation
of ESA treatment after prior discontinuation in LR-MDS was prevalent. In these patients,
poor clinical outcomes and frequent health system visits were observed. This study
highlights the need to consider alternative treatment options for patients with LR-MDS.
P782: IMMUNOPHENOTYPIC AND MOLECULAR GENETIC MARKERS OF UNFAVORABLE PROGNOSIS IN PATIENTS
WITH MYELODYSPLASTIC SYNDROME
H. Trubkina1,*, I. Iskrou2, V. Smolnikova3, T. Lebedeva3, I. Lendzina1
1hematology department №3, State Institution “Minsk Scientific and Practical Center
for Surgery, Transplantology and Hematology”; 2Department of Clinical Hematology and
Transfusiology, Belarusian Medical Academy of Postgraduate Education; 3clinical laboratory
STCM, State Institution “Minsk Scientific and Practical Center for Surgery, Transplantology
and Hematology”, Minsk, Belarus
Background: Therapy is selected based on risk, need for transfusion of blood components,
percentage of bone marrow blasts, and cytogenetic profile. In low-risk groups, the
goal of treatment is to reduce the need for replacement therapy and prevent transformation
to higher-risk or AML. In higher risk groups, the goal is to prolong survival.
Currently, there are no approved criteria for selecting therapy for patients with
MDS based on the classifications and prognostic scales used, treatment regimens for
progression of MDS or its refractory forms.
Aims: The aim of the study was to identify unfavorable prognostic immunophenotic and
molecular genetic markers of blast cells in patients with high-risk myelodysplastic
syndrome
Methods: The prospective cohort study included 68 patients with newly diagnosed MDS
in the period from January 2017 to December 2021. The patients underwent a primary
diagnostic complex of MDS, including immunophenotyping and molecular genetic research
of bone marrow aspirate. The age of patients is 25-82 years, the median age is 64
years.
According to the IPSS classification included: high risk (10 pats), intermediate-2
(26 pats), intermediate-1 (26 pats), low risk (6 pats). According to the IPSS-R classification
included: very high risk (23 pats), high risk (17 pats), intermediate (14 pats), low
risk (12 pats), very low risk (2 pats). The following molecular genetic markers were
found: complex aberrations >3 (10 pats), trisomy 8 chromosome (8 pats), absence of
molecular genetic disorders (27 pats), isolated 5q chromosome (13 pats), any changes
of the 7 chromosome (8 pats), deletion of the 20 chromosome (3 pats), deletion of
the 11 chromosome (1 pats), TP53 (2 pats).
To explore immunophenotic and molecular genetic markers we use three state “illness-death”
model. We consider the following states: diagnosis, transformation into leukemia;
death.
Results: We identified high-risk immunophenotypic and molecular genetic markers
- · for transition “diagnosis-death”:
- CD38 <50% HR 3.7 (1.2-11.5 CI; p = 0.022),
- CD13> 50% HR 8.7 (1.1-67.8 CI; p = 0.04),
- complex aberrations >3 HR 5.8 (1.4-24.6 CI; p=0,015);
- · for transition “diagnosis-transformation”:
- CD71 ≥65% HR 4.1 (1.35-12.4 CI; p = 0.013);
- CD13> 75% HR 2.8 (1.1-7.1 CI; p = 0.034),
- complex aberrations >3 HR 2.8 (0,86-9,0 CI; p=0,087);
- · for transition “transformation-death“:
- CD25 <5% HR 6.3 (1.4-28 / 4 CI; p = 0.017),
- CD 33 <50% HR 6.6 (1.3-34.7 CI; p = 0.026)),
- complex aberrations >3 HR 0,22 (0,06-0,84 CI; p=0,027),
- trisomy 8 chromosome HR 7.9 (0.71-88 CI; р=0,093).
Figure 1 – The probability of various conditions depending on the time since the diagnosis.
State 1 - estimation of the probability of being in the state «diagnosis-transformation»
State 2 - estimation of the probability of being in the state «transformation-death»
State 3 - estimation of the probability of being in the state «diagnosis-death»
Image:
Summary/Conclusion: The selection of groups of patients with identified immunophenotypic
and molecular genetic markers of a high risk of transformation into acute leukemia
and a high risk of death (“diagnosis-death”: CD38<50%, CD13>50%, complex aberrations>3;
“diagnosis-transformation”: CD71≥65%, CD13>75%, complex aberrations>3; “transformation-death“:
CD25<5%, CD33<50%, complex aberrations>3, trisomy 8 chromosomes) requires consideration
of a new approach to therapy.
P783: SYSTEMIC THERAPY UTILIZATION AND HEMATOLOGIC OUTCOMES IN LOWER-RISK MYELODYSPLASTIC
SYNDROMES (LR-MDS): FINDINGS FROM A REAL-WORLD MEDICAL RECORD REVIEW STUDY IN THE
US, UK, AND EUROPE (EU)
M. Diez-Campelo1,*, A. Yucel2, R. Goyal3, R. C. Parikh3, S. Dhuliawala3, M. Jimenez3,
M. Sluga-O’Callaghan4, C. Hughes5, D. Tang2, U. Germing6
1Hematology Department, Institute of Biomedical Research of Salamanca, University
Hospital of Salamanca, Salamanca, Spain; 2Bristol Myers Squibb, Princeton, NJ; 3RTI
Health Solutions, Research Triangle Park, NC, United States of America; 4RTI Health
Solutions, Lyon, France; 5Bristol Myers Squibb, Summit, NJ, United States of America;
6University Hospital of Dusseldorf, Dusseldorf, Germany
Background: Patients (pts) with LR-MDS experience a mean life-year loss of 6 years,
and the treatment focus is improvement of quality of life and prolongation of life
expectancy.
Aims: This retrospective medical record review assessed real-world data to determine
prevailing LR-MDS treatment patterns and hematologic outcomes.
Methods: Eligible pts (≥18 years of age) with LR-MDS (Jul 1, 2013–Sep 30, 2018) were
identified by participating hematologist-oncologists in the US, Canada (CAN), UK,
and EU (France, Germany, Spain). Study measures and hematologic outcomes were descriptively
assessed (data abstraction Jun–Nov 2021). Aggregated analyses used pooled US/CAN and
UK/EU data sets for the overall population and pt subgroups treated with clinician-defined
first-line (1L) and second-line (2L) erythropoiesis-stimulating agents (ESAs).
Results: Medical record data were abstracted by 114 US/CAN (n=174 pts; median age
60.6 years; 68.4% male) and 166 UK/EU clinicians (n=319 pts; median age 67.1 years;
72.4% male). Median follow-up duration from MDS diagnosis was 60.6 months (US/CAN)
and 67.6 months (UK/EU). Approximately half of the pts had “low” risk status (49.4%
US/CAN; 53.9% UK/EU) categorized by the Revised International Prognostic Scoring System
at initial diagnosis. The most common karyotype abnormality was del(5q) (27.6% US/CAN;
20.4% UK/EU). SF3B1 mutation was recorded for 7.5% US/CAN and 8.2% UK/EU pts. Almost
all pts had received ≥1 line of systemic treatment for managing MDS-associated anemia
(US/CAN: 165 [94.8%] 1L, 77 [44.3%] 2L; UK/EU: 294 [92.2%] 1L, 133 [41.7%] 2L). In
1L, ESA-based regimens were the most common treatment with median duration of therapy
of ≥2 years (US/CAN: 142 [86.1%], of which the majority (80.3%) was ESA monotherapy;
UK/EU: 264 [89.8%], comprising 82.2% ESA monotherapy). Treatment characteristics and
outcomes are shown in the Table. Among pts who were transfusion dependent (TD) before
1L ESA, a very small proportion achieved transfusion independence (TI) or red blood
cell transfusion reduction ≥50%, which was maintained for a median of <1.5 years.
Among the 1L ESA-treated pts who were TD, TI was achieved by 5.7% (US/CAN) and 12.9%
(UK/EU). In 2L ESA-treated pts (n=23 US/CAN; n=46 UK/EU), almost all were previously
treated with a 1L ESA-containing regimen (95.7% US/CAN; 84.8% UK/EU). The most common
reasons for reinitiating ESA in 2L (n=22, 15.5% US/CAN; n=39, 14.8% UK/EU) were overall
health status (40.9% US/CAN; 33.3% UK/EU), treatment efficacy (40.9% US/CAN; 48.7%
UK/EU), and compliance with national guidelines (13.6% US/CAN; 46.2% UK/EU). Among
the pts who reinitiated ESA in 2L, the most common reasons for discontinuation of
1L ESA were completion of planned course of therapy (45.5% US/CAN; 28.2% UK/EU), pt
decision (40.9% US/CAN; 20.5% UK/EU), progressive disease (36.4% US/CAN; 51.3% UK/EU),
and adverse events (13.4% US/CAN; 15.4% UK/EU). Lenalidomide was used in 1L therapy
among 8.5% pts in US/CAN and 5.8% in UK/EU. Among those with del(5q) mutation, ESA
was used in 1L therapy among 83.3% pts in US/CAN and 87.7% in UK/EU.
Image:
Summary/Conclusion: ESA-based regimens were the most common treatment for anemia management
in pts with LR-MDS; >1/3 of pts were TD before initiating 1L ESA therapy. A considerable
proportion of 1L ESA-treated pts reinitiated ESA in 2L despite the majority having
discontinued 1L treatment due to planned therapy completion, pt decision, or progressive
disease. This real-world study suggests that there is an unmet need in addressing
anemia burden with ESAs, and novel and effective treatments are needed.
P784: A PHASE I/II STUDY OF VENETOCLAX IN COMBINATION WITH ASTX727 (DECITABINE/CEDAZURIDINE)
IN TREATMENT‐NAÏVE HIGH‐RISK MYELODYSPLASTIC SYNDROME (MDS) OR CHRONIC MYELOMONOCYTIC
LEUKEMIA (CMML)
S. Venugopal1,*, H. kantarjian1, A. Maiti1, N. Short1, G. Montalban-Bravo1, Y. Alvarado1,
K. S. Chien1, R. Kanagal-Shamanna2, N. Pemmaraju1, N. Daver1, T. Kadia1, G. Borthakur1,
E. Jabbour1, G. Garcia-Manero1
1Leukemia; 2Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston,
United States of America
Background: In MDS, hypomethylating agents (HMA) remain the standard of care. ASTX727,
an oral fixed dose combination of HMA decitabine (35mg) and cytidine deaminase inhibitor
cedazuridine (100mg), was recently approved in the US for the treatment of MDS and
CMML. Venetoclax (Ven), an orally bioavailable BCL-2 inhibitor in combination with
azacitidine has shown preliminary clinical activity in treatment-naïve, higher risk
MDS
Aims: Based on this data, we designed a study to evaluate a total-oral regimen of
Ven+ASTX727 combination in pts with higher risk MDS or CMML
Methods: This single arm Phase I/II study of orally administered ASTX727 in combination
with Ven (NCT04655755) is enrolling patients ≥18 years with treatment‐naïve, higher
risk MDS (intermediate-2- or high-risk categories) per IPSS or CMML with excess blasts
≥5%. The primary objective is to determine the safety and tolerability (phase 1) and
overall response rate (ORR, defined as CR+mCR) (phase 2) of Ven+ASTX727 combination.
ASTX727 is administered orally daily on D1‐5 and Ven is administered orally daily
on D1‐14 of 28-d cycles. 3 dose levels of venetoclax in combination with ASTX727 will
be tested. To mitigate tumor lysis syndrome, pretreatment white blood cell count should
be less than 10 × 109/L. Cytoreduction is allowed. The safety population includes
all patients who received any dose of Ven+ ASTX727, and the efficacy population includes
patients who have a valid baseline and post-baseline disease assessment and had received
at least one dose of the study drug
Results: 15 pts have been enrolled to date (Figure). The median age is 72 years (range
54-94) with 12 pts aged ≥65 years. These patients had a median bone marrow blast count
of 12% (range 6-15%) and harbored a median number of 4 (range 1-9) mutations. Most
pts had adverse risk mutations such as ASXL1 (67%) and RUNX1 (47%).
No DLTs were observed in the initial 6 patient safety lead-in. There were no deaths
during the 30-day and 60-day window. No tumor lysis syndrome was observed. The ORR
was 87% with 3 pts achieving CR (20%) and 10 pts achieving marrow CR (67%). All pts
achieved a response within 1 cycle among which 4 pts, including one with TP53
mut,proceeded to hematopoietic stem cell transplant. 2 pts had stable disease at the
end of 1 cycle. At a median follow up of 8.8 months, the median duration of response
was not reached (range 0.9-11.1 months), and the median overall survival was not reached
(range 1.0-12.1 months).
Image:
Summary/Conclusion: Ven+ASTX727 combination appears safe and demonstrates preliminary
efficacy in pts with higher risk MDS or CMML with excess blasts. Total-oral regimen
of Ven+ASTX727 combination appears to be a promising strategy for high-risk MDS or
CMML pts and may alleviate the burden of chronic, long-term parenteral HMA treatment
P785: CURRENT CARDIOVASCULAR DISEASE RISK PREDICTION MODELS ARE NOT APPLICABLE IN
MDS PATIENTS: PRELIMINARY RESULTS OF A PROSPECTIVE OBSERVATIONAL SINGLE-CENTRE COHORT
STUDY
C. Misidou1, E. Gkolia2, C. Kymparidou1, V. Papadopoulos1, M. Papoutselis1, K. Liapis1,
I. Mitroulis1, G. Vrachiolias1, S. P. Deftereos2, D. Stakos3, I. Kotsianidis1,*
1Department of Hematology, Democritus University of Thrace Medical School; 2Department
of Radiology, University Hospital Of Alexandroupolis; 3Cardiology Department, Democritus
University of Thrace Medical School, Alexandroupolis, Greece
Background: Cardiovascular disease (CVD) is a leading cause of death in lower-risk
Myelodysplastic syndrome (LR-MDS) patients and a shared pathobiology between MDS and
CVD has been postulated. However, current evidence emanates only from retrospective
cohorts suffering from crucial limitations.
Aims: Prospective annotation can overcome these limitations and delineate the role
of MDS as an independent CVD risk factor. For this reason we conducted a prospective
observational single-centre cohort study in LR-MDS patients.
Methods: Patients underwent thorough evaluation for CVD every 6 months. Cerebrovascular,
peripheral and coronary vascular beds were assessed for atherosclerosis by ultrasound
and coronary artery calcium (CAC), respectively. Cardiac structure and function was
assessed by echocardiography. Multi-Ethnic Study of Atherosclerosis (MESA), Framingham
(FRS) risk scores and serum markers of myocardial injury (Serum high-sensitivity troponin
T, hsTnT), stress (N-terminal pro–B-type natriuretic peptide, NT-proBNP), and systemic
inflammation (high-sensitivity C-reactive protein, hsCRP) were also estimated at each
visit. Chi-square, Wilcoxon signed-ranks tests and Pearson’s correlation coefficient
were used as appropriate. Multiple linear regression was applied for modeling the
relationship between a dependent and one or more independent variables. The level
of statistical significance was set to P=0.05.
Results: Table 1 presents baseline characteristics of 24 patients included so far.
There were no correlations of CAC, MESA and FRS with MDS subtype and IPSS-R risk category
when adjusting for MDS-CI, preexisting CVD, age, sex, and transfusion dependence.
In sharp contrast to the general population (Okwuosa, TM et al. JACC 2011), CAC score
was not associated with FRS and was disproportionately increased in low and very low
FRS risk patients (Figure 1). Likewise, the number of affected vascular beds (AVB)
did not correlate with FRS (Figure 2), further suggesting that the current CVD risk
prediction tools are inaccurate in MDS and arguing for an intrinsic tendency of MDS
towards CVD development.
In 9 out of 24 patients a second evaluation at 6 months was completed. All 9 patients
increased significantly CAC (p=0.004) and MESA (p=0.003) scores at 6 months, while
5/9 patients also increased the number of AVB. The average increase of CAC score (189.77
+/-212.89) was markedly higher than expected in the age-matched general population
(Budoff, MJ et al. JACC 2013) and was not associated with FRS at baseline, while it
was also independent of MDS subtype and IPSS-R risk category after adjusting for MDS-CI,
preexisting CVD, age, sex, and transfusion dependence. Finally, baseline NT-proBNP
levels correlated strongly with CAC score (Figure 3) in line with numerous evidence
showing that NT-proBNP can serve as a predictor of CVD risk in both MDS and non-MDS
individuals.
Image:
Summary/Conclusion: To our knowledge this is the first study in LR-MDS patients assessing
longitudinally all critical parameters and providing objective measurements of subclinical
atherosclerosis in all vascular beds. Our preliminary results indicate that the established
CVD prediction models do not operate in LR-MDS patients and reinforce the notion that
MDS represents an independent factor for CVD development. Accurate assessment of CVD
risk and prompt detection of subclinical cardiovascular disease in MDS patients will
assist clinical decisions on the introduction of intensive monitoring and preventive
use of CVD-specific interventions in these individuals.
P786: ANTI-PD-1 ANTIBODY (SINTILIMAB) PLUS DECITABINE AS FIRST LINE TREATMENT FOR
PATIENTS WITH HIGHER-RISK MYELODYSPLASTIC SYNDROME(MDS): PRELIMINARY RESULTS FROM
A SINGLE-ARM, OPEN-LABEL, PHASE II STUDY
J. wang1,*, S. li1, H. Jiang1, Y. Chang1, X. Zhao1, J. Jia1, X. Zhu1, L. Gong1, X.
Liu1, W. Yu1, X. Huang1,2
1Peking University People’s Hospital, Peking University Institute of Hematology. National
Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic
Stem Cell Transplantation; 2XiaoJun Huang, Correspondence.Peking University, People’s
Hospital, Peking University Institute of Hematology, 11 Xizhimen South Street, Beijing,
China
Background: Hypomethylating agents (HMAs) are the preferred treatment for untreated
patients (pts) with higher-risk MDS, but the survival of pts after HMAs treatment
is poor. It has been demonstrated that the PD-1/PD-L1 expression was upregulated by
HMAs in MDS pts, providing a strong rationale for combining HMAs with PD-1 antibody
for MDS treatment.
Aims: To evaluate safety and efficacy of Sintilimab plus decitabine for pts with higher-risk
MDS in a single-arm, open-label,phase 2 study(ChiCTR2100044393).
Methods: Adult pts with higher-risk MDS by the IPSS-R were enrolled. Patients received
decitabine 20mg/m2 intravenously daily for 5 days and sintilimab 200mg IV starting
on the first and 22nd day of a cycle every 42 days until unacceptable toxicity, relapse,
or progression, for a maximum of 8 cycles. The primary endpoint was overall response
rate (CR+PR+mCR). Simon’s optimal two-stage design was employed.If five or more pts
in stage I (13 pts) achieved ORR, the study would enroll 34 additional pts in stage
II (47 pts).Secondary endpoints included safety, and survival outcomes. The relationship
between expression levels of immune-checkpoint and efficacy of the combination therapy,
as well as other potential biomarkers were also explored via genomic profiling.
Results: At data cut-off (February 22, 2022), 21 pts were enrolled with a median follow-up
time of 6.5 months. The median age of pts was 64 years (range 30-83), and the other
characteristics are summarised in Table 1.The ORR was 62%, with six pts reaching complete
response (CR),four pts reaching marrow CR and three pts achieved marrow CR+ hematologic
improvement (HI). In addition,four pts reaching HI,so the overall improvement rate
was 81%. The most common grade 3 treatment-emergent adverse events(TEAEs)(>10%) were
febrile neutropenia (76.2%) and pulmonary infection (38.1%). No grade 4 TEAEs and
treatment-related deaths occurred. A total of 12 pts (57.1%)experienced immune-related
adverse event, including rash (28.6%), pneumonia (9.5%), hypothyroidism (9.5%), elevated
serum bilirubin (4.8%) and transpeptidase (4.8%), which were all resolved by glucocorticoid.
In these 21 pts, the most frequently mutated genes are ASXL1(28.6%), RUNX1 (14.3%)
and TET2(14.3%). Updated biomarker data will be presented during the EHA meeting.
Image:
Summary/Conclusion: To the best of our knowledge, this is the first study to evaluate
the efficacy and safety of sintilimab plus decitabine in pts with untreated higher-risk
MDS. The preliminary results demonstrate that the combination therapy is relatively
safe with anti-tumor activity.
P787: SABATOLIMAB (MBG453) COMBINATION TREATMENT REGIMENS FOR PATIENTS WITH HIGHER-RISK
MYELODYSPLASTIC SYNDROMES: THE MYELODYSPLASTIC SYNDROMES STUDIES IN THE STIMULUS IMMUNO-MYELOID
CLINICAL TRIAL PROGRAM
A. M. Zeidan1,*, A. Al-Kali2, U. Borate3, T. Cluzeau4, A. E. DeZern5, J. Esteve6,
A. Giagounidis7, K. Kobata8, R. Lyons9, U. Platzbecker10, D. A. Sallman11, V. Santini12,
G. F. Sanz13, M. A. Sekeres14, A. H. Wei15, Z. Xiao16, M. Van Hoef17, C. Nourry-Boulot17,
I. Sadek18, F. Ma18, A. Iordan17, J. Sabo18, G. Garcia-Manero19
1Yale University and Yale Cancer Center, New Haven; 2Mayo Clinic, Rochester; 3Oregon
Health & Science University, Portland, United States of America; 4Centre Hospitalier
Universitaire de Nice, Nice, France; 5Sidney Kimmel Comprehensive Cancer Center, Johns
Hopkins, Baltimore, United States of America; 6Hospital Clinic, Barcelona, Spain;
7Marien Hospital, Düsseldorf, Germany; 8None, Los Angeles; 9The US Oncology Network,
San Antonio, United States of America; 10University Hospital Leipzig, Leipzig, Germany;
11H. Lee Moffitt Cancer Center & Research Institute, Tampa, United States of America;
12University of Florence, Florence, Italy; 13Hospital Universitari i Politécnic La
Fe; and CIBERONC, Instituto de Salud Carlos III, Valencia; Madrid, Spain; 14Sylvester
Comprehensive Cancer Center, University of Miami, Miami, United States of America;
15Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne
Hospital, Melbourne, Australia; 16Blood Diseases Hospital, Chinese Academy of Medical
Sciences, Tianjin, China; 17Novartis Pharma AG, Basel, Switzerland; 18Novartis Pharmaceuticals
Corporation, East Hanover; 19MD Anderson Cancer Center, Houston, United States of
America
Background: Patients (pts) with higher-risk myelodysplastic syndromes (HR-MDS) unfit
for hematopoietic stem cell transplant (HSCT) have poor outcomes with single-agent
hypomethylating agent (HMA) therapy. Novel treatments with durable responses and survival
benefit are needed. TIM-3 regulates innate and adaptive immunity and is expressed
on leukemic stem cells (LSCs) and blasts but not normal hematopoietic stem cells.
Sabatolimab is a novel immuno-myeloid therapy that binds to TIM-3 on immune cells,
facilitating immune activation and phagocytosis of leukemic cells. Sabatolimab also
binds to TIM-3 on leukemic cells, potentially impeding self-renewal of LSCs. Sabatolimab
+ HMAs had promising efficacy and response durability in HR-MDS in early-phase (ph)
trials, with few significant immunologic adverse events (AEs; Brunner AM. ASH 2021.
Oral 244).
Aims: The STIMULUS immuno-myeloid clinical trial program investigates the safety,
efficacy, and durability of sabatolimab-based combination therapies in patients with
myeloid malignancies. Exploration of sabatolimab’s mechanism of action and identification
of potential biomarkers predictive of response are planned. This abstract summarizes
the design of 4 ongoing STIMULUS trials in previously untreated adult pts with HR-MDS
ineligible for intensive chemotherapy or HSCT.
Methods: STIMULUS-MDS1 (NCT03946670) is an international, randomized, double-blind,
placebo-controlled, ph II trial evaluating sabatolimab + HMA in pts with very-high,
high-, or intermediate-risk (vH/H/IR)-MDS who have completed enrollment (N=127). Primary
endpoints are complete remission (CR) and progression-free survival (PFS).
STIMULUS-MDS2 (NCT04266301) is an international, randomized, double-blind, placebo-controlled,
ph III trial of sabatolimab + azacitidine (AZA) in pts with vH/H/IR-MDS or chronic
myelomonocytic leukemia-2; the study has completed recruiting (N=530). Primary endpoint
is overall survival (OS). Key secondary endpoints are time to definitive deterioration
of fatigue; red blood cell transfusion-free interval; and improvement of fatigue,
physical, and emotional functioning.
STIMULUS MDS-US (NCT04878432) is a US-based, open-label, single-arm, ph II trial of
sabatolimab + HMA of investigator’s choice (AZA intravenous [IV] or subcutaneous,
decitabine IV, or oral decitabine/cedazuridine) in pts with vH/H/IR-MDS. Target enrollment
is 90 pts. Primary endpoint is safety. Secondary endpoints include CR, PFS, leukemia-free
survival (LFS), and OS.
STIMULUS-MDS3 (NCT04812548) is an international, open-label, single-arm, ph II trial
that explores triplet therapy of sabatolimab + AZA + BCL-2 inhibitor venetoclax (VEN)
in pts with vH/HR-MDS. Part 1 is a safety run-in comprising 2 safety cohorts with
~6 pts receiving sabatolimab 400 mg + AZA + VEN and ~12 pts receiving sabatolimab
800 mg + AZA + VEN. Primary endpoint of part 1 is safety. If (both 400- and 800-mg
doses) sabatolimab + AZA + VEN are safe, the trial will move to an expansion cohort
(~58 pts) of sabatolimab 800 mg Q4W + AZA + VEN. Primary endpoint is CR. Secondary
endpoints include CR + mCR rate, overall response rate (CR + mCR + PR + hematologic
improvement), OS, PFS, LFS, and event-free survival.
The STIMULUS immuno-myeloid clinical trial program is investigating the efficacy and
safety profiles and the quality-of-life improvements for sabatolimab-based combination
therapies in pts with myeloid malignancies. The STIMULUS-MDS trials aim to gain insight
into the durable benefit of sabatolimab combination therapies in pts with HR-MDS.
Results: None.
Image:
Summary/Conclusion: None.
P788: FACTORS DRIVING TREATMENT DECISION IN PATIENTS WITH INTERMEDIATE-RISK MYELODYSPLASTIC
SYNDROME (MDS): A RETROSPECTIVE ANALYSIS FROM THE GRUPO ESPAÑOL DE SÍNDROMES MIELODISPLÁSICOS
SPANISH MDS REGISTRY
M. Díez-Campelo1,2,*, L. Hernandez-Donoso3, E. Sasse3, S. Colicino3, J. Curto4, A.
Molero Yordi5, M. Tormo Díaz2,6,7, M. Arnan8, G. Sanz9,10, M. Díaz-Beyá11, M. T. Cedena
Romero12, A. Jerez13, D. Valcárcel Ferreiras2,5
1Hospital Universitario de Salamanca, Salamanca; 2Grupo Español de Síndromes Mielodisplásicos
(GESMD), Valencia, Spain; 3Novartis Pharma AG, Basel, Switzerland; 4MFAR Clinical
Research SL, Madrid; 5Vall d’Hebron Institute of Oncology (VHIO), University Hospital
Vall d’Hebron, Barcelona; 6Hospital Clínico Universitario Valencia; 7INCLIVA Biomedical
Research Institute, Valencia; 8Institut Català d’Oncologia, Hospital Duran i Reynals,
Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona,
L’Hospitalet de Llobregat, Barcelona; 9Hospital Universitario y Politécnico La Fe,
Valencia; 10CIBERONC, Instituto de Salud Carlos III, Madrid; 11Hospital Clinic Barcelona,
Barcelona; 12Hospital Universitario 12 de Octubre, Madrid; 13Hospital Universitario
Morales Meseguer, Murcia, Spain
Background: Current treatment (tx) options for patients (pts) with MDS are limited,
although multiple potential txs are in development. The use of the Revised International
Prognostic Scoring System (IPSS-R) for diagnosis of MDS supports tx decisions and
prognostic assessment. Pts with very low/low-risk (vL/LR) MDS usually receive supportive
care (SC) to manage symptoms, whereas those with very high/high-risk (vHR/HR) MDS
commonly receive active txs such as hypomethylating agents (HMAs), allogeneic hematopoietic
stem cell transplantation (alloSCT), or chemotherapy (CT). There is limited agreement
on whether active tx or standard SC should be used for pts with intermediate-risk
(IR) MDS.
Aims: We aimed to identify factors possibly associated with the real-world use of
tx in pts with IR-MDS in the Spanish MDS registry.
Methods: This was a retrospective, observational study of data from pts registered
in the Grupo Español de Síndromes Mielodisplásicos (GESMD) from January 2008-June
2020. Adult pts with MDS (WHO 2008 classification) who signed an informed consent
and had a minimum follow-up of 6 months in the registry were included. Pts with unavailable
data on IPSS-R risk or HMA tx were excluded. We collected demographic, clinical, and
laboratory data, and tx used, including azacitidine (AZA), CT, alloSCT, SC (eg, transfusions,
growth factors) and other tx (eg, immunosuppressors, androgens). Univariate analysis
was performed to evaluate the association of potential risk factors, using categorical
and continuous variables, with the tx use. We focused our analysis on AZA and SC in
pts with IR-MDS ineligible for alloSCT. Odds ratio, 95% confidence interval, and exploratory
P value were reported for comparison between tx use.
Results: Overall, 4604 pts (median age: 76 y; male: 61%; IPSS-R score vHR: 432, 9.4%;
HR: 487, 10.6%; IR: 761, 16.5%; LR: 1795, 39%; vLR: 1129, 24.5%) were included. Pts
with IR-MDS were 63% male and similar in number by score and median age at diagnosis
compared with the total study population: 3.5 (n=268, 73 y), 4.0 (n=270, 75 y) and
4.5 (n=223, 75 y). Most common treatments received were AZA (n= 241, 31.7%) and SC
(n=250, 32.9%); a minority received CT and/or alloSCT (n=84, 11%), while 186 pts (24.4%)
did not receive any. AZA was the only HMA used in this population. The median time
from diagnosis to start of AZA in this group was 3 months. When comparing AZA with
SC use in pts with IR-MDS, univariate analysis (Table 1) showed that gender, age at
diagnosis, year of diagnosis, bone marrow and peripheral blood blasts, transfusion
dependency, and hematologic parameters at diagnosis (eg, hemoglobin, platelet, and
monocyte counts) were associated with tx selection. A multivariate analysis is ongoing
for deeper understanding of these data.
Image:
Summary/Conclusion: These preliminary data show several factors significantly associated
with tx use, suggesting that in some pts with IR-MDS, AZA is more likely to be used
versus SC when these factors are considered. It is possible that selection bias, misclassification
and confounding may have occurred as the registry only collects routine data according
to local practices, with variability in the quality and completeness of data across
participating centers. Other factors not captured, such as pt characteristics, comorbidities,
access to health care, and social/familial support, could have also influenced the
tx decision. Further analyses are ongoing to identify the relationship of multiple
factors at once and assert the strength of these associations while removing the effect
of confounders.
P789: ANALYSIS OF THE CLINICAL FEATURES, PROGNOSIS AND GENE MUTATIONS IN PRIMARY MYELODYSPLASTIC
SYNDROME PATIENTS WITH MYELOFIBROSIS
G. Xu1,*, S. Zhu1, H. Tong1
1Hematology, Hospital, Hangzhou, China
Background: Bone marrow(BM) fibrosis in myelodysplastic syndrome (MDS-MF) is observed
approximately in 10–50% of primary MDS patients in the previous reports. BM biopsy
is essential for the diagnosis and evaluation of MDS-MF patients and they differ from
MDS without MF in terms of clinical performance, prognosis and survival. Although
MDS-MF is associated with an adverse prognosis in the view of most hematologists,
MDS-MF is not independently listed as a subtype of MDS according to the 2016 World
Health Organization classification, indicating the need for further investigation.
In this study, we retrospectively analyzed 417 newly diagnosed primary MDS cases in
our hospital and to compare the clinical characteristics, prognosis and gene mutations
of the two group patients with or without myelofibrosis.
Aims: This study aims to analyze the clinical features, prognosis and characteristics
of the gene mutations of the primary myelodysplastic syndrome with myelofibrosis (MDS-MF)
patients, ultimately to improve the cognition of MDS-MF.
Methods: From January 1, 2013, to February 1, 2020, 417 newly diagnosed primary MDS
patients in the first affiliated hospital of Zhejiang University with bone marrow
(BM) biopsy examination were included. They were divided into two groups according
to whether BM associated with fibrosis (2005 European Myelofibrosis Network criteria),
the MDS-MF group(MF grades 1–3)and the MDS without MF group (MF grade 0).The clinical
features, prognosis and gene alterations were analyzed between the two groups retrospectively.
Results: ① MF was confirmed in 46.3% cases of all the MDS patients(193/417), of which
66.3%(128/193)were MF-1, 25.9%(50/193)were MF-2 and 7.8% were MF-3 respectively. There
is no statistically significant difference in the proportion of subtypes patients
according to the 2016 who classification (P=0.342).
②Compared with those without MF, MDS-MF were significantly associated with worse OS(P=0.035)
and worse PFS(p<0.001). Subgroup analysis showed that both OS and PFS of either the
MDS-MF1-2 group or the MDS-MF 3 group were also significantly worse than those of
the MDS without MF group(P=0.013 and <0.001 respectively).
③The clinical features of the two groups were statistically significant differences
in the higher number of BM blast cell(P=0.006), higher prevalence of death (P=0.031),
higher rate of Leukemic transformation (P<0.001), higher level of serum lactate dehydrogenase
(LDH) (p<0.001), higher level of β2-microgram (p=0.003) and higher level of CRP in
the peripheral blood (P=0.004) in the MDS-MF group than in the MF0 group.
④Among all the patients,337 were finished Next-generation-sequencing (NGS) test. Using
the same panel analysis, U2AF1 gene mutation was more frequently in the MDS-MF compared
with those patients without MF (P=0.027). There was totally 14.2% (48/417) patients
with the mutation of U2AF1, of which 19.7%(29/147) in MDS-MF group and 10.0%(19/190)
in MDS without MF group. There was significant difference concomitant with U2AF1 mutation
between the two groups(P=0.038).
Image:
Summary/Conclusion: MDS-MF has unique laboratory and clinical characteristics. MF
is an independent risk factor for shorter overall survival (OS) and worse progression-free
survival (PFS) in primary MDS. Evaluation of MF is very significant for prognosis
judgment in primary MDS. The U2AF1 mutation may be a biomarker related to the poor
prognosis of MDS-MF. However, the mechanism of U2AF1 in MDS-MF patients needs to be
further explored.
P790: THE EXPRESSION LEVEL OF WT1 MRNA IN PATIENTS WITH IDIOPATHIC CYTOPENIA OF UNDETERMINED
SIGNIFICANCE (ICUS) PROGRESSING TO MYELODYSPLASTIC SYNDROME (MDS)
X. Ye1,*, J. Huang1
1Hematology, The First Affiliated Hospital of Zhejiang University School of Medicine,
Hangzhou, China
Background: Idiopathic cytopenia of undetermined significance (ICUS) is defined as
patients with one or more unexplained cytopenia who do not meet diagnostic criteria
for myelodysplastic syndrome (MDS) or another hematologic disorder. In the clinical
course of ICUS, some patients eventually have typical MDS changes. Until now, there
is no effective method to predict the development of the disease. Recent studies have
shown that the expression level of WT1 mRNA in peripheral blood has important clinical
significance in the diagnosis, occurrence, development and prognosis of MDS. At the
same time, some studies have proved that there is a significant difference in the
expression level of WT1 mRNA between MDS and hemocytopenia caused by other reasons.
Aims: In order to clarify the clinical significance of WT1 mRNA expression in the
progression of ICUS to MDS.
Methods: Between April 2017 and January 2020, untreated adult patients with ICUS or
lower-risk MDS were enrolled in the study. All patients detected the WT1 mRNA expression
of bone marrow and peripheral blood at diagnosis. Then patients with ICUS were followed
up. Every three months, the patients with ICUS detected the WT1 mRNA expression of
peripheral blood, until the patient progressed to MDS. The results of WT1 transcript
levels are depicted as WT1 (copies/mL)/c-ABL (copies/mL) as a percentage.
Results: Total of 80 patients with ICUS and 40 patients with lower-risk MDS were enrolled.
At diagnosis, the median expression level of WT1 mRNA in MDS group was 5.99 (0.08-79.27),
and that in ICUS group was 0.50 (0.16-22.48). There was significant difference between
them (P = 0.000). Then 80 patients with ICUS were followed up to January 2022. The
median follow-up time was 30 months (11-54 months). 8 patients developed MDS, the
progression rate was 10%, and the median progression time was 6.5 months (2-19 months).
The median expression level of WT1 mRNA at the initial diagnosis in ICUS group without
progressing to MDS was 0.44 (0.16-7.58), and that in progressing to MDS group was
9.44 (0.26-22.48). There was significant difference between the two groups (P = 0.031).
The median expression level of WT1 mRNA was 19.185 (0.73-42.30) when MDS was diagnosed.
The median maximum expression level of WT1 mRNA in peripheral blood of ICUS group
without MDS was 0.98 (0.22-7.58). There was significant difference between the two
groups (P = 0.007).
Summary/Conclusion: The study clarified the WT1 mRNA expression status of ICUS patients.
There were significant differences in WT1 mRNA expression level between patients with
ICUS and lower-risk MDS, and the WT1 mRNA expression level of patients with lower-risk
MDS was relatively high. The study confirmed that patients with high WT1 mRNA expression
level in the patients with ICUS (WT1 mRNA expression level > 9.44) are very likely
to progressing to MDS; Dynamic monitoring of WT1 mRNA expression in peripheral blood
is an effective method for following-up after ICUS diagnosis. The progressive increase
of WT1 mRNA expression is one of the indicators for researchers to judge the progress
of ICUS to MDS.
P791: COMMON VARIANTS IN COMPLEMENT PROTEINS, C3 AND CR1, ENHANCE COMPLEMENT ATTACK
ON PNH ERYTHROCYTES AND INCREASE RISK FOR EXTRAVASCULAR HAEMOLYSIS
A. J. Baral1,*, C. Mckinley2, R. Fellows1, T. Hallam1, T. Cox1, D. Payne3, S. Richards2,
A. Pike4, R. Kelly4, M. Griffin4, P. Hillmen2, K. Marchbank1, D. Kavanagh5, D. Newton6,
C. Harris1
1Translational and Clinical Research Institute, Newcastle University, Newcastle upon
Tyne; 2Division of Haematology and Immunology, Leeds Institute of Medical Research,
University of Leeds; 3Haematological Malignancy Diagnostic Service, St. James’s University
Hospital; 4Department of Haematology, St James University Hospital, Leeds; 5Translational
and Clinical Research Institute, Newcastle University, Newcastle upon Tyne; 6Division
of Haematology and Immunology, Leeds Institute of Medical Research, University of
Leeds, Leeds, United Kingdom
Background: In paroxysmal nocturnal haemoglobinuria (PNH), absence of glycosylphosphatidylinositol
(GPI) anchors leads to loss of the complement inhibitor, CD59, on PNH erythrocytes
(E) causing complement-mediated intravascular haemolysis. Treatment with anti-C5 anitbody
(eculizumab or ravulizumab) rescues PNH-E but loss of another GPI-anchored regulator,
CD55, leads to accumulation of C3 fragments on E and extravascular haemolysis (EVH)
due to C3-driven erythrophagocytosis. Therefore, approximately 30% of patients still
require blood transfusion. Deposition of C3b and generation of downstream inactivation
fragments such as iC3b and C3dg is controlled by binding of complement receptor type
1 (CR1) to C3b with factor I (FI), forming a trimolecular complex (TMC). We hypothesise
that polymorphisms in C3 and CR1 influence the efficacy of this inactivation process
and dictate propensity for EVH.
Aims: To determine the combined effect of C3-S/F and CR1 density polymorphisms in
susceptibility to EVH by the inactivation of C3b/iC3b on PNH-E.
Methods: Forty-two eculizumab-treated PNH patients were genotyped for their single
nucleotide polymorphism in C3 (rs2230199,S/F) and CR1 (rs11118133,H/L). C3 loading
on PNH-E of patients were measured by flow cytometry as part of the routine analysis
at the Leeds Haematological Malignancy Diagnostic Service. Lactate dehydrogenase,
haemoglobin levels and reticulocyte count were also recorded. Soluble CR1 (sCR1) constructs
were generated using recombinant technology and C3 variants were purified from human
plasma using classical chromatography techniques. Interactions between C3b and CR1
and formation of the alternative pathway regulatory TMC (C3b:CR1:FI) were analysed
using surface plasmon resonance (SPR).
Healthy donors were genotyped for their CR1 density polymorphism and relative CR1
expression on E was quantitated by flow cytometry. Impact of CR1 density polymorphism
on C3b inactivation was studied using C3b-coated streptavidin beads and solubilised
E from CR1 H/H or H/L donors and C3b breakdown to iC3b/C3dg was measured using flow
cytometry.
Results: There was a trend for higher mean percentage of C3 loading in patients who
have the C3-S/S allele (n=28, 30%) compared to patients with the C3-S/F allele (n=12,
18%) and a patient who had the C3-F/F allele (7%). Patients with high (H/H) CR1 expression
(n=25, 20%) showed a trend for lower mean percentage of C3 loading on PNH-E than patients
with intermediate (H/L) CR1 expression (n=17, 32%). In patients with the C3-S/S polymorphism,
haemoglobin levels were significantly lower (p=0.0132) and higher numbers of patients
(20 of 29) had at least one transfusion event.
Using SPR, we demonstrated that CR1 bound C3b-F with a higher affinity than C3b-S.
This led to higher levels of TMC formation with FI and more effective decay of C3-F
convertase (C3b-F:Bb) by CR1. Solubilised E from a CR1-H/H donor converted iC3b more
effectively to C3dg compared to E from CR1-H/L donors.
Summary/Conclusion: These data indicate that both C3-S/F and CR1 density polymorphisms
may influence C3b loading on PNH-E, with C3 fragments influencing EVH risk. Weaker
binding of C3b-S to CR1 led to a decreased regulatory TMC formation with FI and slower
decay of C3-S convertase (C3b-S:Bb). These data also indicate that E from individuals
with lower CR1 expression convert the iC3b to C3dg more slowly. Overall, these data
demonstrate mechanisms that enhance erythrophagocytosis in susceptible PNH patients
by increased C3 loading on PNH-E and a decreased in removal of phagocytic opsonin,
iC3b.
P792: CRISPR/CAS9-BASED MODEL OF HETEROZYGOUS CXCR4 WT/R334X MUTATION TO STUDY CELLULAR
PHENOTYPES IN WHIM SYNDROME
K. Zmajkovicova1,*, S. Pawar1, S. Maier-Munsa1, A. Badarau2, A. G. Taveras3
1X4 Pharmaceuticals (Austria) GmbH; 2Former Employee at X4 Pharmaceuticals (Austria)
GmbH, Vienna, Austria; 3X4 Pharmaceuticals, Boston, United States of America
Background: WHIM syndrome is a phenotypically heterogenous primary immunodeficiency
characterized by Warts, Hypogammaglobulinemia, Infections, Myelokathexis, neutropenia
and lymphopenia. WHIM syndrome pathogenesis is causally linked to heterozygous gain-of-function
(GOF) mutations in the C-terminus of the chemokine receptor CXCR4, a master regulator
of immune cell trafficking and homeostasis, causing desensitization defects and hyperactivation
of downstream signaling. The most frequently reported mutation in patients with WHIM
syndrome is c.1000C>T, which results in a C-terminal truncation of the receptor at
position R334 (R334X).
Aims:
In vitro assays using cell lines with exogeneous overexpression of CXCR4
WHIM variants can model the GOF cellular phenotypes of WHIM syndrome but may not entirely
mimic the condition in patients, who typically have one wildtype (WT) CXCR4 allele.
We aimed to characterize a cellular model of homozygous and heterozygous CXCR4
R334X in the endogenous locus, to better understand the pathogenic impact of harboring
mutations in one or both alleles.
Methods: CRISPR-Cas9 platform was used to establish a model of heterozygous mutations
found in patients with WHIM syndrome. Jurkat cell line (with endogenous expression
of WT CXCR4) was edited to harbor the c.1000C>T/R334X mutation in a single allele
(WT/RX) or in both alleles (RX/RX). Unedited parental Jurkat cell line and Jurkat
cells with edited silent mutations (WT/WT) were used as controls.
Results: Upon stimulation with C-X-C chemokine ligand 12 (CXCL12), RX/RX cell lines
displayed an internalization defect (65% of CXCR4 receptors remaining on the cell
surface at 100 nM CXCL12) compared to the parental (24%) and WT lines (20%). The RX/WT-expressing
cells had an intermediate phenotype (43%). We analyzed the signaling responses downstream
of activated CXCR4, for which GOF phenotypes had been reported in R334X patient cells.
Calcium mobilization in response to CXCL12 was enhanced in cells harboring the R334X
mutation, reaching a 2.5- to 3-fold higher maximum effect (Emax) compared to cells
with WT CXCR4. ERK activation downstream of CXCR4 reached a higher amplitude (8-fold
increase over baseline) and duration after stimulation with CXCL12 in all lines expressing
R334X compared to the parental and WT/WT cell lines (6-fold increase). Presence of
a single mutant allele seemed to confer the full GOF phenotype in both signaling readouts
in this cell line. Migration of cells toward CXCL12 was significantly enhanced in
R334X-expressing cells. Mavorixafor, a CXCR4 antagonist currently in clinical trials
for the treatment of patients with WHIM syndrome, was active in an unbiased manner
on a spectrum of CXCR4-related functions (ligand binding inhibition, calcium mobilization,
ERK activation and chemotaxis) with comparable biological activity and potency in
cells expressing the WT and R334X CXCR4 receptor.
Summary/Conclusion: We established the first model recapitulating the heterozygous
CXCR4WT/R334X mutations found in patients with WHIM syndrome using the CRISPR/Cas9
platform. This cellular model recapitulates the functional defects found in immune
cells from patients with WHIM syndrome. When stimulated with CXCL12, WT/RX -expressing
cells appeared to display full GOF phenotype in downstream signaling assays compared
to RX/RX-expressing cells. This observation is consistent with the dominant inheritance
pattern of WHIM syndrome. The present study brings several novel insights in CXCR4WHIM
biology and enriches the toolbox of models available for studying WHIM syndrome.
P793: SCREENING OF NATURALLY OCCURRING CXCR4 VARIANTS FOR IDENTIFICATION OF NOVEL
PATHOGENIC MUTATIONS FOR WHIM SYNDROME
S. Pawar1,*, I. Wiest1, S. Maier-Munsa1, B. Maierhofer1, N. Sondheimer2, A. G. Taveras3,
A. Badarau4, K. Zmajkovicova1
1X4 Pharmaceuticals (Austria) GmbH, Vienna, Austria; 2Division of Clinical and Biochemical
Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada; 3X4 Pharmaceuticals,
Boston, United States of America; 4Former Employee at X4 Pharmaceuticals (Austria)
GmbH, Vienna, Austria
Background: WHIM (Warts, Hypogammaglobulinemia, Infections, Myelokathexis) syndrome
is a rare, autosomal-dominant primary immunodeficiency marked by neutropenia and lymphopenia.
Classically, WHIM syndrome pathogenesis is causally linked to a variety of heterozygous
gain-of-function mutations in the C-terminus of chemokine receptor CXCR4, a master
regulator of immune cell trafficking and homeostasis. As of February 2022, 18 variants
in CXCR4 have been implicated in WHIM syndrome, while a substantial number remain
unexplored for their association with the disease. Using in vitro functional assays,
we previously found that impaired receptor internalization and enhanced chemotaxis
are conserved in all CXCR4
WHIM variants and that defective internalization correlates with neutropenia, the
most penetrant phenotype of patients with WHIM syndrome. We used this approach to
confirm functional defects in primary cells isolated from patients harboring a novel
variant, suggesting that the in vitro profiling approach can potentially drive identification
of pathogenic variants.
Aims: We aimed to functionally characterize multiple previously uncharacterized variants
of CXCR4 using in vitro assays. We also examined allele frequencies of these novel
variants to estimate the potential number of individuals harboring these variants
with a long-term goal of determining the actual prevalence of WHIM syndrome.
Methods: The CXCR4-negative K562 cell line was used as a model system to express 53
novel CXCR4 variants identified in patient and population databases (ClinVar, Ensembl,
CentoMD, GnomAD) and genetic screening initiatives (Invitae PATH4WARD) found in the
C-terminus (hotspot of CXCR4
WHIM mutations) as well as throughout the protein. The effects of these mutations
on CXCR4 internalization and chemotaxis were studied in cells stimulated with the
C-X-C chemokine ligand 12 (CXCL12). The in vitro functional parameters were compared
with known pathogenic CXCR4 variants and were used to assign a cumulative score (impaired
internalization + enhanced chemotaxis) of functional defect severity.
Results: Out of 53 selected variants, 42 were missense (ms), 6 were frameshift (fs),
3 were nonsense (ns), and 2 were transcript variants (tv). Eighteen of the screened
variants led to a decreased internalization of CXCR4 in comparison to the wild-type
(WT) receptor upon stimulation with CXCL12 while 20 variants demonstrated enhanced
chemotaxis of cells toward CXCL12. Overall, 34 variants showed defects in at least
one CXCR4-dependent marker. Correlation analysis and scoring of the functional parameters
in cells expressing mutated or CXCR4
WT revealed 17 variants (9 ms, 5 fs, 2 ns, 1 tv) that co-segregated with the known
pathogenic WHIM variants. Finally, white blood cell counts in 3 patients harboring
novel variants showed that CXCR4 internalization defects in vitro correlated with
the severity of neutropenia.
Summary/Conclusion: This is the first study characterizing functional impairments
in naturally occurring CXCR4 variants via a pipeline of assays intended to predict
pathogenicity. Seventeen CXCR4 variants were identified (several outside the C-terminus
of CXCR4) that caused in vitro functional defects resembling those exhibited by known
WHIM variants. Many (10/17) of these newly identified variants are present at high
frequencies (4.4 × 10-4–8.0 × 10-6) in genomic databases. Potential association of
these variants with WHIM syndrome is under investigation and is likely to expand current
estimates of WHIM syndrome prevalence.
P794: DIFFERENTIATION POTENTIAL AND RELATIVE GENE EXPRESSION LEVELS ARE CHANGED IN
CFU-F FROM BONE MARROW OF APLASTIC ANEMIA PATIENTS AT THE ONSET OF THE DISEASE
A. Dorofeeva1,*, I. Shipounova1, A. Luchkin1, Z. Fidarova1, E. Mikhailova1
1National Research Center for Hematology, Moscow, Russia
Background: Aplastic anemia (AA) is a heterogeneous blood system disease characterized
by the bone marrow (BM) aplasia in and pancytopenia. There are non-severe (NAA), severe
(SAA) and very severe (VSAA) forms of AA. The main cause of AA is believed to be the
damage to the hematopoietic stem cells (HSC) by autoreactive cytotoxic T-lymphocytes.
Another reason for the AA development may be a dysfunction of the BM stromal microenvironment.
One of the ways of study the BM stroma is the analysis of colony-forming units of
fibroblasts (CFU-F).
Aims: To determine the CFU-F concentration in BM of AA patients, to assess the differentiation
potential of individual colonies and the expression level of genes associated with
proliferation, differentiation, immune response, and maintenance of HSC in CFU-F from
BM of untreated AA patients.
Methods: The study included 38 AA (15 with SAA, 4 with VSAA and 19 with NAA) patients
in the debut. VSAA was combined with SAA for the analysis. The control group included
22 healthy donors. BM was aspirated at diagnostic punctures in patients and planned
exfusion from donors after informed consent. CFU-F cultures were obtained according
to the standard method. The colonies number was counted, and total RNA was isolated.
Individual CFU-F colonies were obtained by plating 15000 BM mononuclear cells per
well of a 96-well plate in aMEM with 20% FBS. Wells with individual clones had been
analyzed further. The differentiation potential of CFU-F colonies was determined by
the level of marker gene expression after incubation in a medium with inducers of
osteogenic or adipogenic differentiation. Gene expression was determined by TaqMan
RT-PCR. Data are presented as mean±standard error of the mean. Differences were considered
significant at p<0.05 using an unpaired Student’s t-test.
Results: CFU-F concentration in the BM of AA patients was not altered (33.58±9.12
per 106 BM mononuclear cells versus 34.43±8.39 in donors). The differentiation potential
of CFU-F clones from SAA patients was skewed to osteogenesis when compared with donors
and NAA patients. The proportion of CFU-F clones that failed to differentiate was
decreased in SAA patients, while in NAA patients the proportion of such clones increased
in comparison with healthy donors. These data may indicate a change in the ratio of
stromal precursors of different maturity in the BM of patients with NAA and SAA compared
with healthy donors (Table 1).
CFU-F cultures of SAA patients significantly differed from those of healthy donors
in the expression level of 10 genes (Table 2). The similar changes in 5 of these genes
(PDGFRA, PDGFRB, ANG1, CFH, NES) were also found in NAA, but, with the exception of
NES, the changes were not significant. Most of these genes are associated with the
HSC maintenance, thus, the identified changes could be the result of BM aplasia. The
expression level of the other 5 genes in CFU-F of NAA patients did not differ from
that in donors (FGFR1, IL1B, PPARG, HLA-DR) or tended to change oppositely than in
SAA (IL10). These genes are functionally associated with immune response and differentiation.
Possibly, the activity of these genes prevents the development of more pronounced
aplasia in NAA patients.
Image:
Summary/Conclusion: CFU-F of NAA and SAA patients differ from CFU-F of healthy donors
by their differentiation potential and gene expression. Changes in CFU-F of SAA patients
are more pronounced, which may be associated with deeper aplasia. This work was supported
by the Russian Foundation for Basic Research, grant no. 19-015-00280.
P795: CHARACTERIZATION OF ERCC6L2 SYNDROME PATIENTS’ TRANSCRIPTOME
S. P. Douglas1,2,*, I. Kaaja1,2, T. H. Räisänen1,2, E. Pitkänen1,3, O. Kilpivaara1,2,4,
U. Wartiovaara-Kautto1,5
1Applied Tumor Genomics Research Program, Faculty of Medicine; 2Department of Clinical
and Medical Genetics, Medicum; 3Institute for Molecular Medicine Finland (FIMM), University
of Helsinki; 4HUSLAB Laboratory of Genetics, HUS Diagnostic Center, Helsinki University
Hospital; 5Department of Hematology, Helsinki University Hospital Comprehensive Cancer
Center, University of Helsinki, Helsinki, Finland
Background: Biallelic germline mutations in ERCC6L2 cause a bone marrow failure syndrome
with a tendency for acquiring somatic TP53 mutations and possible progression to acute
myeloid leukemia with erythroid characteristics. The disease and some functions of
ERCC6L2 have only recently been characterized but a lot remains uncovered. ERCC6L2
affects DNA repair, but also plays a role in immunoglobulin class-switching and mitochondrial
function. Although the biallelic ERCC6L2 mutation is in all the patient’s cells, it
seems to affect the hematopoietic cells most drastically. However, we have seen differences
also in the metabolism and growth of the ERCC6L2-deficient patients’ skin fibroblasts.
Constitutional changes in RNA expression levels and altered metabolism due to ERCC6L2
deficiency may also affect the bone marrow niche and help us understand the disease
more profoundly.
Aims: We aimed to characterize ERCC6L2 syndrome on the transcriptome level using patient-derived
samples (blood, skin fibroblasts, lymphoblastoid cell lines [LCLs]).
Methods: 32 blood samples were collected from 15 ERCC6L2 patients (some in multiple
time points), and 6 healthy controls.
Skin fibroblast cell lines from 4 ERCC6L2 patients, and 3 healthy controls were grown
in duplicates. In total 14 samples were grown in high-glucose (25mM) media until about
80% confluence and cells were harvested after six hours of changing the media.
LCLs derived from EBV-transformed PBMCs from 6 ERCC6L2 patients and 4 healthy controls
were grown in duplicates in two different media compositions (in total 20 samples).
3’ RNA sequencing was performed on RNA extracted from all patient samples. Differential
expression analysis was done with DESeq2 and pathway enrichment analysis with PathfindR.
Results: ERCC6L2-patients and controls differ significantly in their mRNA expression
levels in multiple cell types and enriched pathways differ between cell types.
In the patient blood samples, global genome nucleotide excision repair and transcription-coupled
nucleotide excision repair (TC-NER) pathways are overrepresented compared to healthy
controls. ERCC6L2 has been suggested to be involved both in TC-NER and non-homologous
end joining of DNA double strand breaks. Also neddylation, which is a ubiquitin-like
posttranslational modification affecting gene expression, is highly overrepresented.
Cellular response to chemical stress is also upregulated, which demonstrates the reported
sensitivity of ERCC6L2-deficient cells to some chemicals.
The patients’ skin fibroblasts have different expression patterns compared to healthy
controls in pathways related to cell differentiation, response to stimuli and extracellular
matrix composition. These results support our previous observations about impaired
fibroblast growth and metabolism. Expression changes in fibroblasts can tell us about
effects that the germline ERCC6L2 mutation can have on extra-hematopoietic cells and
tissues in the absence of somatic TP53 mutations.
LCLs from ERCC6L2 patients’ cells differ from healthy controls by various immune system
pathways: the enrichment of complement activation, phagocytosis, B-cell receptor regulation,
but also in their response to metal ions.
Summary/Conclusion: Multiple cell types of ERCC6L2 patients’ cells differ significantly
from healthy controls’ cells in pathways related to DNA repair, stress response, cell
morphology and immune function. These enriched pathways demonstrate the diverse effects
the germline ERCC6L2 mutation has on the patients’ cells.
P796: ENRICHED CD16+ MONOCYTES DRIVE T LYMPHOCYTES CELL RESPONSES IN APLASTIC ANEMIA
M. Ge1,*, H. Wu1, J. Huo1, Y. Shao1, Y. Zheng1
1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science
& Peking Union Medical College, Tianjin, China
Background: Monocytes, an important component of innate and acquired immunity, encompass
three subsets, the classical monocytes (CM, CD14++CD16-), intermediate monocytes (IM,
CD14++ CD16+) and nonclassical monocytes (NCM, CD14+CD16++). IM and NCM are collectively
referred to CD16+ monocytes, which has been studied in many inflammatory diseases.
Whereas, the distribution and function of monocytes in acquired aplastic anemia (AA)
remain unclear.
Aims: This study aims to determine the distribution of peripheral blood (PB) and bone
marrow (B M) monocyte subpopulations in AA and expound the role of CD16+ monocytes
on T lymphocytes function.
Methods: The proportions of monocyte subsets in the PB and BM of AA patients and healthy
donors (HD) were analyzed by flow cytometry. CD16+ monocytes were isolated by magnetic
beads and co-cultured with CD3+ T cells, afterwards, the basic biological characteristics
of T cells including proliferation, activation and differentiation etc. were evaluated.
Results: Results showed that the percentage of CD16+ monocytes in PB and BM was significantly
increased in AA patients compared with that in HDs, and the proportion of CM was decreased.
However, there was no difference between patients with SAA and NSAA. CD16+ monocytes
were further divided into two subgroups, which named IM and NCM. In contrast with
HD, the two subgroups were both elevated in AA, mainly the IM. After treatment, NCM
and IM were reduced and CM was increased in the patients of complete and partial remission.
Then the effect of CD16+ monocytes on T cells function were elaborated. Using a co-culture
model, we discovered that CD16+ monocytes in AA patients had stronger ability to promote
the proliferation of CD3+ T, CD3+CD4+ T and CD3+CD8+ T cells than that in HDs (Fig
1A, b-d). Furthermore we found that AA-CD16+ monocytes played an important role in
the over activation of CD3+CD4+ T and CD3+CD8+ T cells (Fig 1B, b-d). As expected,
AA-CD16+ monocytes were extremely potent inducers of Th1、Th17 and cytotoxic T cells
(Tc1) differentiation in vitro (Fig 1C, b-d), nevertheless, there was no difference
on the effect of monocytes on Treg between AA and HD.
Image:
Summary/Conclusion: This study provided that the expanded CD16+ monocytes, including
NCM and IM,had a major role in driving T cell responses in AA patients.
P797: METABOLIC PROFILING IN ERCC6L2 AND SHWACHMAN DIAMOND SYNDROME
I. Kaaja1,2,*, S. P. Douglas1,2, T. H. Räisänen1,2, K. Haimilahti3,4, A. I. Nieminen5,
E. Pirinen3,6, U. Wartiovaara-Kautto1,7, O. Kilpivaara1,8
1Applied Tumor Genomics Research Program, Faculty of Medicine; 2Department of Medical
and Clinical Genetics, Medicum, Faculty of Medicine; 3Clinical and Molecular Metabolism
Research Program, Faculty of Medicine; 4Stem Cells and Metabolism Research Program,
Faculty of Medicine; 5Institute for Molecular Medicine Finland (FIMM), University
of Helsinki, Helsinki; 6Research Unit for Internal Medicine, Faculty of Medicine,
University of Oulu, Oulu; 7Department of Hematology, Helsinki University Hospital
Comprehensive Cancer Center, University of Helsinki; 8HUSLAB Laboratory of Genetics,
HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
Background: Germline mutations in genes involved in DNA repair, telomere maintenance,
and ribosome biogenesis are established causes of bone marrow failure (BMF) syndromes.
Biallelic ERCC6L2 germline mutations cause BMF and accumulation of somatic TP53 mutations
in bone marrow, contributing to the disease progressing into acute myeloid leukemia.
ERCC6L2 is tentatively indicated in DNA repair and mitochondrial function, however,
understanding its function is still in its infancy.
Aims: To particularly understand the role of mitochondrial function in ERCC6L2 syndrome,
we compared cellular metabolism in ERCC6L2 syndrome and Shwachman Diamond syndrome
(SDS), a ribosomopathy also prone to somatic TP53 mutagenesis. Our aim was to discover
any underlying alterations in mitochondrial function and to perceive any differences
between the two syndromes that are similar in phenotype but different in genotype.
Methods: To study the effect of the ERCC6L2 and SDBS germline mutations on cellular
metabolism in isolation from the effect of accumulating somatic mutations in the bone
marrow, we examined patient-derived skin fibroblasts (ERCC6L2 syndrome, n=2; SDS,
n=2) and healthy controls (n=2). Mitochondrial function was studied with Seahorse
XFe96 Analyzer where mitochondrial oxygen consumption rate (OCR) was measured under
different glucose and glutamine concentrations. Furthermore, we inhibited mitochondrial
pyruvate carrier (MPC) to examine the metabolic compensation for mitochondrial pyruvate
deficiency via Seahorse Substrate Oxidation Stress Test. Mitochondrial DNA (mtDNA)
amount was quantified in patient samples using qPCR. To further investigate the metabolism
in each BMF syndrome, untargeted metabolomic screening of ERCC6L2 (n=4), SDS (n=3)
and healthy control (n=3) fibroblasts in normal and low-glucose conditions was conducted
by liquid chromatography mass spectrometer. We cross-referenced differentially expressed
(DE) metabolites with DE genes (3’ RNAseq) in a matched set of samples to identify
important biological pathways.
Results: Our results indicate impaired mitochondrial respiration under low energy
availability in both ERCC6L2 syndrome and SDS, as both BMF syndromes showed decreased
maximal respiratory and reserve capacity in low-glucose conditions. Equal mtDNA amount
was confirmed in patient and control cells suggesting that the decline was not related
to changes in mitochondrial content. In ERCC6L2, but not in SDS cells, maximal respiratory
and reserve capacity improved upon the addition of L-glutamine. In addition, we observed
a lower glutamine concentration in both ERCC6L2 and SDS in low-glucose conditions.
MPC inhibition lowered the respiration in SDS cells, but not in ERCC6L2 cells, indicating
that pyruvate may be compensated by other substrates, such as glutamine oxidation,
in ERCC6L2 but not in SDS cells. Together, these findings imply increased consumption
of glutamine in ERCC6L2 cells and possibly impaired glutamine utilization in SDS cells.
Metabolomic screening and integration of metabolomics data with transcriptomics detected
subtle trends indicating changes in TCA metabolite and gene expression as well as
in nicotinate/nicotinamide metabolism.
Image:
Summary/Conclusion: We show altered mitochondrial function in low-glucose conditions
in two BMF syndromes, and propose glutamine-dependency as a factor for challenged
mitochondrial function under low energy substrate availability in ERCC6L2 syndrome.
Further studies on the metabolic behavior of the hematopoietic stem cell and its niche
in BMF syndromes are needed to understand the role of metabolism in the disease progression.
P799: SPATIAL ANALYSIS IDENTIFIES A SPECTRUM OF IMMUNE DYSREGULATION IN ACQUIRED BONE
MARROW FAILURE CONDITIONS
R. Koldej1,2,*, A. Prabahran1,2,3, C. W. Tan2,4, M. Davis2,4, D. Ritchie1,2,3
1ACRF Translational Research Laboratory, Royal Melbourne Hospital; 2Faculty of Medicine,
Dentistry and Health Sciences, University of Melbourne; 3Clinical Haematology, Royal
Melbourne Hospital; 4The Walter and Eliza Hall Institute of Medical Research, Melbourne,
Australia
Background: Poor Graft Function (PGF), manifested by multilineage cytopenias with
complete donor chimerism post allogeneic stem cell transplantation, and acquired Aplastic
Anaemia (AA) are acquired bone marrow failure syndromes. Both are thought to result
from T cell activation resulting in excessive interferon-γ (IFNγ) production, which
in turn suppresses haematopoiesis. In AA, this can promote clonal stem cell selection
and potentially progression to myeloid malignancy. Despite similarities in the clinical
presentation and proposed mechanism, no studies to date have compared the immunobiology
of the bone marrow (BM) microenvironment in these conditions.
Aims: To examine the immune microenvironment of the BM in archival AA and PGF BM trephines
and identify common immunopathologies.
Methods: Archival BM trephines were sourced from patients with PGF (n=20), AA at Diagnosis
(AA_DX) (n=15), AA at Progression (AA_PROG) to MDS/AML (n=15) and normal controls
(NC) (n=20). 6 x 300µm regions identified by dual CD3/CD45 immunofluorescent staining
were analysed per BM trephine using NanoString GeoMX™ digital spatial profiling for
the expression of 57 proteins with an immunology focused panel. Data was analysed
using a Limma-Voom bioinformatics pipeline.
Results: Significant dysregulation was identified across multiple proteins. Overall,
22 proteins had significantly different expression in AA_DX samples (13 up, 9 down)
compared to NC with AA_PROG and PGF having 8 (4 up, 4 down) and 14 (7 up, 7 down)
respectively. As would be expected given the hypocellular BM, expression of CD45 was
significantly reduced compared to NC across AA_DX (adj P = 5.413e-10), AA_PROG (adj
P = 8.299e-8) and PGF (adj P = 8.634e-9) groups.
When AA_DX patients were stratified into those who progressed (n=6) vs those who did
not (n=9), or when AA_DX vs AA_PROG was compared in patients with matched samples
(n=6) there were no significantly differentially expressed proteins identified. This
suggests that while AA provides an immune microenvironment that is permissive to MDS/AML
pathogenesis, it may not directly impact on clonal evolution.
Total monocytes were unchanged by CD11c expression. However, AA and PGF samples exhibited
upregulation of CD163 compared to NC (AA_DX adj P = 4.5e-6; AA_PROG adj P = 1.2e-5;
PGF adj P = 1.58e-8) suggesting an increase in monocyte/macrophage lineage cells.
Expression of CD66b was also downregulated in AA_DX (adj P = 8.459e-24), AA_PROG (adj
P = 1.193e-10) and PGF (adj P = 2.244e-14) suggesting a reduction in non-classical
monocytes, neutrophils or granulocytic MDSCs.
Importantly, VISTA was downregulated across AA_DX (adj P = 6.84e-27), AA_PROG (adj
P = 1.1e-5) and PGF (adj P = 1.58e-8) and STING was downregulated in AA_DX compared
to NC (adj P = 6.139e-13) with a trend towards reduced expression in PGF. Decreased
expression of these key immunoregulatory proteins may have wide ranging effects on
BM resident macrophages, dendritic cells, MDSCs and T cells leading to reduced immunoregulation
and increased T cell activation/IFNγ production, ultimately resulting in stem cell
depletion. Data on cell specific expression is currently being obtained to refine
this model.
Summary/Conclusion: Spatial analysis demonstrated that patients with AA and PGF exhibit
similar patterns of protein expression likely resulting in a BM immune microenvironment
of decreased immunoregulation. AA_DX samples exhibited a greater degree of dysregulation
of multiple markers compared to PGF suggesting that these diseases represent a spectrum
of immune dysregulation.
P800: BLOCKADE OF COMMON GAMMA CHAIN CYTOKINE SIGNALING WITH REGN7257, AN INTERLEUKIN
2 RECEPTOR GAMMA (IL2RG) MONOCLONAL ANTIBODY, PROTECTED MICE FROM GRAFT-VERSUS-HOST
DISEASE AND IMMUNE APLASTIC ANEMIA
A. Le Floch1,*, K. Nagashima2, T. Norton2, L. Perlee2, A. Murphy2, M. Sleeman2, J.
Orengo2
1Inflammation and Immune Diseases; 2Regeneron, Tarrytown, United States of America
Background: Pathogenic T-cell responses in T cell-mediated diseases can be driven
by cytokines of the common gamma chain (γc) cytokine family (IL2, IL4, IL7, IL9, IL15,
and IL21). γc cytokines signal through their corresponding receptors, expressed primarily
on immune cells (including T cells), that share a common coreceptor, interleukin 2
receptor subunit gamma (IL2RG) that is required for signaling.
Aims: To understand the roles of γc cytokines in driving graft-versus-host disease
(GVHD) and bone marrow failure disorders such as immune aplastic anemia (AA), we generated
REGN7257, a fully human IL2RG monoclonal antibody that inhibits γc cytokine-induced
signaling, and we tested its ability to suppress pathogenic T-cell responses in mouse
models of T cell-mediated disease.
Methods: We evaluated the efficacy of REGN7257 in a xenogeneic mouse model of GVHD,
where immunodeficient NOD-scid-IL2RGnull mice were engrafted with human peripheral
blood mononuclear cells (huPBMC). Mice were treated with REGN7257 either prophylactically
or therapeutically, and monitored for weight loss and survival, peripheral human T-cell
engraftment as well as serum pro-inflammatory cytokine levels over time. In a separate
experiment, mice were sacrificed at day 49 post-huPBMC injection (after 4 weeks of
antibody treatment) for tissue analysis (liver, lung, skin and bone marrow) of immune
cell infiltration (T cells and macrophages), inflammation and/or fibrosis.
Results: In a xenogeneic model of GVHD, both prophylactic and therapeutic γc cytokine
signaling blockade with REGN7257 effectively protected mice from weight loss and resulted
in improved survival, by reducing T-cell expansion in blood and production of pro-inflammatory
cytokines in serum. Consistent with the classic pathology of GVHD, lungs, liver and
skin of control mice were highly infiltrated by T cells, while γc cytokine signaling
blockade strongly reduced T-cell infiltration, with reduced tissue levels of pro-inflammatory
cytokines. Importantly, therapeutic γc cytokine signaling blockade in established
GVHD (i.e. when mice already showed weight loss) provided similar benefit. Blockade
of γc cytokine signaling also led to a reduction in the severity of chronic GVHD,
with decreased macrophage infiltration in liver and associated hepatic fibrosis. In
this xenogeneic model of GVHD, hemoglobin levels and platelet numbers in blood were
both reduced, indicating anemia and thrombocytopenia, respectively, which are two
complications associated with aplastic anemia. In addition to peripheral pancytopenia,
recipient mice that were engrafted with huPBMC also showed severe marrow aplasia.
Importantly, this phenotype of aplastic anemia was prevented by blockade of γc cytokine
signaling.
Summary/Conclusion: Blockade of γc cytokine signaling with REGN7257 protected against
T cell-mediated pathology in a xenogeneic GVHD mouse model that uniquely presents
hallmarks of both acute and chronic GVHD, with T-cell expansion/infiltration into
tissues and liver fibrosis, as well as hallmarks of immune aplastic anemia, with bone
marrow aplasia and peripheral cytopenia. These data provide evidence of γc cytokines
as key drivers of T cell-mediated responses, offering a potentially novel strategy
for the management of T cell-mediated diseases, such as GVHD and immune AA.
P801: EVALUATION OF COMMON GAMMA CHAIN CYTOKINE SIGNALING BLOCKADE WITH REGN7257,
AN INTERLEUKIN 2 RECEPTOR GAMMA (IL2RG) MONOCLONAL ANTIBODY, ON IMMUNE CELL POPULATIONS
ACROSS SPECIES
A. Le Floch1,*, K. Nagashima2, D. Birchard2, H. Pan2, C. Korgaonkar2, T. Norton2,
L. Perlee2, A. Murphy2, M. Sleeman2, J. Orengo2
1Inflammation and Immune Diseases; 2Regeneron, Tarrytown, United States of America
Background: The common γ chain cytokine receptor (γc; IL2RG) family of cytokines includes
interleukin 2 (IL2), IL4, IL7, IL9, IL15, and IL21. This set of cytokines exhibits
broad pleiotropic actions on both the innate and adaptive immune system with each
cytokine sharing the IL2RG chain as part of its signaling receptor complex. Mutations
in the IL2RG gene result in X-linked severe combined immunodeficiency (XSCID) in humans,
whereby patients present with dramatically diminished numbers of T cells and NK cells,
and dysfunctional B cells.
Aims: Given the crucial role for γc cytokines in the development and function of lymphocytes,
modulating their activities may offer therapeutic potential in a range of immune-mediated
diseases. To understand the effects of γc cytokine blockade on immune cells including
T-cell subsets, we utilized REGN7257, a fully human IL2RG monoclonal antibody that
inhibits γc cytokine-induced signaling, and tested its ability to suppress immune
cell populations and their functions across species (mouse, cynomolgus monkey [CM]
and human).
Methods: We evaluated the effects of γc cytokine signaling blockade with REGN7257
on immune cell populations in Il2rg
hu/hu mice and CM by flow cytometry. Mixed lymphocyte reaction assays were performed
to look at the impact of γc cytokine signaling blockade on activation/proliferation
of human T cells. To further analyze the contribution of γc cytokines to T-cell differentiation
and function, we performed in vitro transcriptomic studies on human peripheral blood
mononuclear cells stimulated with anti-CD3/CD28 beads.
Results: γc cytokine signaling blockade with REGN7257 in Il2rg
hu/hu mice efficiently reduced circulating B, NK and T cell populations, with no changes
in blood neutrophil counts. In addition, the impact of γc cytokine signaling blockade
on lymphocyte populations in vivo were investigated in CM in a single dose pharmacokinetic/pharmacodynamic
study and a repeat-dose toxicology study. Inhibition of γc cytokine signaling with
REGN7257 in CM led to similar phenotypic changes to that observed in Il2rg
hu/hu mice, with decreases in peripheral T cells and NK cells, without impacting B
cells, granulocytes, platelets or red blood cells. γc cytokine signaling blockade
led to a reduction in both CD4+ and CD8+ T cell counts with effector memory T cells
being the most impacted T-cell population studied. Furthermore, blockade of γc cytokine
signaling led to reduction in activated and proliferating T cells in CM and demonstrated
potent inhibition of allogeneic responses in mixed human lymphocyte reaction assays,
by preventing T-cell activation and proliferation. Two-tailed gene set enrichment
analysis identified 4 distinct patterns of significantly enriched gene sets that were
impacted by γc cytokine signaling upon CD3/CD28-induced activation of human T cells:
gene sets involved in inflammatory responses, differentiation and proliferation, activation
and immune responses, as well as those related to adhesion and migration. These signatures
were blocked with REGN7257 treatment.
Summary/Conclusion: Taken together, these pharmacological observations highlight the
major role for γc cytokine signaling in maintenance of lymphocyte populations (i.e.
NK cells and T cells) in mouse and CM, but not other immune cell populations (i.e.
granulocytes, platelets and red blood cells). Furthermore, our data highlight the
importance of γc cytokines in driving functions of CM and human T cells, opening a
potential new route for the management of T cell-mediated diseases.
P802: PATTERNS OF T-CELL AUTOREACTIVITY DIFFER BETWEEN PEDIATRIC APLASTIC ANEMIA AND
HEPATITIS-ASSOCIATED BONE MARROW FAILURE
M. Nováková1,2,*, M. Svatoň1,2, A. Skotnicová1, M. Suková2, L. Řezníčková1,2, T. Valová1,2,
D. Pospíšilová3, O. Fabri4, P. Švec4, J. Trka1,2, O. Hrušák1,2, J. Starý2, E. Mejstříková1,2,
E. Froňková1,2
1CLIP-Paediatric Haematology and Oncology; 2Department of Paediatric Haematology and
Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol,
Prague; 3Department of Pediatrics, Palacky University and University Hospital Olomouc,
Olomouc, Czechia; 4Department of Pediatric Hematology and Oncology, National Institute
of Children’s Diseases and Medical Faculty, Comenius University, Bratislava, Slovakia
Background: Both idiopathic aplastic anemia (AA) and hepatitis-associated bone marrow
failure (HABMF) are considered to be autoreactive T cell mediated diseases. This is
based on indirect evidence, most often the success of immunosuppression or oligoclonality
with T cell repertoire restriction.
Aims: Our questions were: Does the composition of T cells and T-cell receptor beta
(TRB) repertoire differ between HABMF, BMF without preceding hepatitis (nonHABMF)
and normal bone marrow (BM)? Are there any shared clonotypes suggesting common antigenic
stimulus? How does immunosuppressive therapy affect T-cells and does this correlate
with outcome?
Methods: In total 22 pediatric patients diagnosed in 2004-2021 with HABMF were included
in the study. Sixteen HABMF patients were treated with immunosuppressive therapy (IST)
as a frontline treatment, of which 8 patients did not respond. As a control group,
we included 10 nonHABMF patients with AA (n=6) and refractory cytopenia of childhood
(n=4), all treated with IST with poor response in 6 patients, as well as 10 samples
of healthy donor BM grafts and 8 newborn peripheral blood (NBPB) samples. We performed
flow cytometry immunophenotyping at diagnosis (D0) and on day 120 (D120) after initiation
of IST in HABMF and nonHABMF patients. We performed sequencing of the TRB gene rearrangements
according to the EuroClonality-NGS working group protocols using DNA isolated from
these samples and normalized DNA input for TRB library preparation to the equivalent
of 20 000 CD3+ cells per sample based on flow cytometry data, if possible. Whole exome
sequencing (WES) was performed from diagnostic samples of HABMF patients.
Results: Patients with HABMF had a significantly lower proportion of CD3+ T cells
in their BM compared to the nonHABMF with the predominance of CD8+ T cells and their
activation by expression of HLA-DR observed with flow cytometry. The analysis of TRB
repertoire was performed in 22 D0 and 10 corresponding D120 samples of HABMF patients
that had undergone IST and we compared the diversity and clonotype composition with
the nonHABMF patients as well as the BM graft samples and NBPB. Expansion of individual
TRB clonotypes (>5% of all reads) at D0 was observed more frequently in HABMF patients
(9 out of 22) compared to the nonHABMF patients (1 out of 10). None of these expanded
clonotypes were shared among more patients based on their nucleotide or amino acid
sequence. There was no correlation of expanded T cell clone dynamics and response
to IST. The diversity of TRB repertoire was reduced in the nonHABMF group after IST,
however we did not observe any correlation between the clinical response to the IST
and initial TRB diversity in either group of patients. WES did not reveal any pathogenic
variants in genes associated with immune dysregulation.
Summary/Conclusion: Both the proportion of T cells and T-cell composition in the BM
differed between HABMF and nonHABMF patients at diagnosis. Although we observed an
expected reduction in the CD3+ cells after IST both in the HABMF and nonHABMF group,
there was no significant difference in the TRB repertoire diversity in the HABMF patients
between D0 and D120. We did not observe any correlation between the repertoire diversity
and clonotype evolution and the clinical response to IST. Our data further support
the hypothesis that T-cells play a significant role in both AA and HABMF, but suggest
that the pathogenetic mechanism of both entities is different.
Supported by NV20-03-00284, NV19-05-00332, UNCE/MED/015, NU20J-07-00028 and GAUK 534120.
P803: TELOMERE SHORTENING IN BONE MARROW MESENCHYMAL STEM CELLS OF ACQUIRED APLASTIC
ANEMIA PATIENTS ASSOCIATE WITH ALTERED EXPRESSION OF GENES INVOLVED IN TELOMERE MAINTENANCE,
DNA DAMAGE, AND SENESCENCE
J. Srivastava1,*, P. Saxena1, N. Tripathy1, S. Nityanand1, C. P. Chaturvedi1
1Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post Graduate
Institute of Medical Sciences, Lucknow, India
Background: Telomeres are repetitive nucleotide sequences that protect the chromosome
ends from DNA damage. Compelling data suggest that telomere attrition is associated
with alteration in the DNA damage pathways, thereby inducing cell senescence. Idiopathic
acquired aplastic anemia (AA) is a bone marrow (BM) failure disease characterized
by pancytopenia and a hypoplastic fatty marrow. Several studies have reported shortening
of telomere length (TL) in hematopoietic stem cells (HSCs) and lymphocytes in AA patients.
The bone marrow mesenchymal stem cells (BM-MSC), which are the key cells of the BM
niche, have garnered lots of interest as they are found to be functionally impaired
in AA patients. However, a study highlighting telomere dysfunction in the BM-MSC of
AA patients remains obscure. Therefore, we aims to study the telomere length shortening
and its correlation with the alteration of genes involved in the telomere maintenance,
DNA damage, and cellular senescence in AA patients.
Aims: The study aims to evaluate the telomere length in BM-MSC of AA patients in comparison
to that of controls. Further, it correlates the telomere length shortening with the
alteration of genes involved in telomere maintenance, DNA damage, and cellular senescence
in Bm-MSC of AA patients.
Methods: BM-MSC were harvested from the BM of newly diagnosed idiopathic acquired
AA patients (n=20) and healthy control (n=12) after informed consent. The telomere
length and the expression of genes associated with telomere maintenance, DNA damage
responses, and cell senescence of the BM-MSC of AA patients were analyzed by real-time
quantitative-PCR (RT-qPCR). The correlation between telomere shortening and expression
of telomere maintenance, DNA damage, and cellular senescence associated genes was
done using Pearson’s correlation. Student’s t-test and Mann Whitney test were used
to compare differences between groups. All the results were analyzed using GraphPad
Prism software. The data was represented as mean ± standard deviation, and p-value
<0.05 was considered significant.
Results: Twenty patients with idiopathic acquired AA (11 non-severe aplastic anemia
(NSAA) patients and 9 severe aplastic anemia (SAA)) patients and 12 healthy controls
were included in the study. The TL was significantly shorter in the BM-MSC of AA patients
[0.77 (Range: 0.4 – 1.56) as compared to that of healthy controls [1.40 (range: 0.71
– 3.22); p=0.002] (Figure 1). The TL in BM-MSC was not influenced by age (p=0.360),
disease severity, and other hematological parameters. A significant alteration was
observed in the expression of genes involved in telomere maintenance, DNA damage,
and cell senescence with a positive correlation between the telomere length and telomere
maintenance genes TRF2 (r=0.788; p=0.007), TPP1 (r=0.636; p=0.04) and TOP1 (r=0,676;
p=0.03) as well as with gene involved in DNA damage responses CDKN1A (r= 0.667; p=0.03)
and ATM (r=0.783; p=0.007).
Image:
Summary/Conclusion: This is the first study to highlight telomere shortening in the
BM-MSC of AA patients. Furthermore, our study demonstrates that telomere shortening
is in BM-MSC of AA patients is accompanied by altered expression of genes involved
in telomere maintenance, DNA damage, and cellular senescence. These results shed new
insight into that telomere attrition in the BM-MSC of AA patients could be attributable
to DNA damage induced cell senescence or vice-versa. Overall, the findings of this
study reveal that BM-MSC of AA patients potentially contributes towards the disease
pathobiology.
P804: EX VIVO TREATMENT WITH ELTROMBOPAG RESCUES HEMATOPOIETIC STEM CELLS FROM ANTI-THYMOCYTE
GLOBULIN-RELATED DAMAGE DURING IMMUNOSUPPRESSIVE THERAPY IN PATIENTS WITH APLASTIC
ANEMIA
M. Vieri1,2,*, B. Rolles1,2, M. Crocioni1,2, S. Isfort1,2, J. Panse1,2, T. H. Brümmendorf1,2,
F. Beier1,2
1Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation;
2Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Uniklinik
RWTH Aachen, Aachen, Germany
Background: Immunosuppressive therapy (IST) with anti-thymocyte globulin (ATG) and
cyclosporine A is one first-line treatment option for patients (pts) with aplastic
anemia (AA). One side effect of ATG is the release of interferon-gamma (IFN-γ), which
is considered a major factor in the pathogenic autoimmune depletion of hematopoietic
stem cells (HSC). Recently, eltrombopag (EPAG) was introduced for therapy of refractory
AA pts due to its ability to improve HSC function. Clinical trials have evidenced
that EPAG used alongside with IST leads to a higher response rate as compared to its
later administration e.g. 2 weeks after ATG start. However, the potential underlying
cause for this difference has not been clarified to date.
Aims: We hypothesize that EPAG might protect HSC from negative effects of ATG-induced
IFN-γ release.
Methods: Sera of six pts with moderate to severe AA were collected pre-and post-ATG
administration on days +1 and +2. Bone marrow (BM) of AA pts was collected and cryopreserved
before ATG administration. In addition, we collected CD34+ cells isolated from five
healthy donors (HD) undergoing HSC mobilization. HD-derived CD34+ or AA-derived BM
cells were cultured in presence of pre as well as post-ATG serum from pts with AA,
in combination with 5 µM EPAG and/or an IFN-γ neutralizing antibody (NAB; 2.5 µg/mL).
After 48 h, colony forming unit (CFU) assay with cultured cells and intracellular
staining of phospho-SMAD2/3 (known to be down-regulated in response of IFN-γ) were
performed, while IFN-γ was measured via flow cytometry in collected sera.
Results: Serum IFN-γ levels were 0.56 ± 0.6 pg/mL (mean ± standard deviation) post-ATG,
whereas they were 0.015 ± 0.04 pg/mL pre-ATG (n= 6; p=0.08). When we tested effects
of ATG sera on the clonogenic potential (CP) of HD HSC (n=5), a significant decrease
of the number of CFU colonies (NoC) treated with post-ATG (148 ± 75) sera was found
compared to pre-ATG treatment (201 ± 75, p= 0.032). This negative effect was mitigated
by adding EPAG to the post-ATG sera (223 ± 100; p=0.036). To specifically analyze
the potential suppressive role of IFN-γ, we added a IFN-γ NAB to the post-ATG serum
(n=4), which also significantly increased colony formation (NoC 225.7 ± 33.4) compared
to 140.1 ± 9.3 post-ATG without AB (p=0.014). Furthermore, phosphorylation of phospho-SMAD2/3
was found to be significantly lower in post-ATG (2052 ± 428, mean fluorescence intensity,
n=4) compared to pre-ATG (3323 ± 32; p=0.03) treated cells. This effect was partially
rescued by adding EPAG to the post ATG sera (2247 ± 627, p=0.239). Finally, we analyzed
whether EPAG could rescue AA BM in presence of their matching serum, pre- and post-
ATG treatment (n=6). As expected for AA BM, the NoC obtained was substantially reduced.
Adding post-ATG serum resulted in further impairment of their CP (NoC: 2.0 ± 1.6)
compared to the pre-ATG serum (6.0 ± 6.7; p=0.04). Again, addition of EPAG to the
post-ATG serum was able to rescue AA BM cells’ CP from such effect (NoC: 5.6 ± 4.6;
p=0.04).
Summary/Conclusion: We provide evidence that treatment with ATG has an initial negative
impact on the CP of HSC from HD and AA pts, likely mediated through acute release
of IFN-γ. The addition of both IFN-y-NAB as well as of EPAG protects HSC from this
effect. These results indicate a potential explanation for the superior response rates
of EPAG given concurrently with ATG-based IST compared to later application and suggest
that administration of EPAG should be started simultaneously with IST in order to
maximize its beneficial effect in AA pts.
P805: ALEMTUZUMAB IN RELAPSED SEVERE APLASTIC ANEMIA: LONG-TERM RESULTS OF A PHASE
II STUDY
N. Aggarwal1,*, A. L. Manley1, J. Durrani1, R. Shalhoub2, O. Rios1, J. Lotter1, B.
Patel1, C. Wu2, N. Young1, E. Groarke1
1Hematology Branch; 2Office of Biostatistics Research, National Heart, Lung, and Blood
Institute, Bethesda, United States of America
Background: Aplastic anemia (AA) is characterized by pancytopenia and a hypocellular
bone marrow from immune-mediated bone marrow destruction. Immunosuppressive therapy
(IST) is a good alternative to hematopoietic stem cell transplantation (HSCT), but
relapse occurs in about 30% of cases, necessitating further therapy. We investigated
alemtuzumab, a humanized IgG1 monoclonal antibody targeting CD52. We initially reported
on 25 relapsed patients with 56% response. Here, we present long-term results of a
total 42 patients.
Aims: Assess the efficacy and long-term outcomes in patients with relapsed severe
AA (SAA) who received alemtuzumab.
Methods: Participants met Camitta criteria for SAA, had received at least one course
of antithymocyte globulin (ATG)-based IST, and had relapsed. Alemtuzumab was administered
intravenously (IV) (n=28) or subcutaneously (SC) (n=14). The primary endpoint was
hematologic response at 6 months, with blood counts no longer meeting SAA criteria.
Secondary endpoints included robust response (platelet or absolute reticulocyte count
>50x109/L at 6 months), relapse, clonal evolution to myelodysplasia and leukemia,
response to therapy after relapse, and survival. Patients were assessed annually,
and additional AA therapy and transfusion dependence was also recorded. Cumulative
incidence curves were used to estimate the probability of relapse among responders
and clonal evolution, with death as a competing risk. Overall survival probabilities
were evaluated using Kaplan-Meier curves. This trial is registered at clinicaltrials.gov
(NCT00195624).
Results: Patients were enrolled over 9 years with a median follow-up of 6 years. Median
age was 32 years and 57% were female. Five patients (12%) had neutrophil count <0.2x109/L
prior to receiving alemtuzumab. At 6 months, 18 (42%) patients had achieved a response,
and 12 (29%) attained a robust response. Response was achieved in 15 (54%) of those
who received alemtuzumab IV, versus 3 (21%) who received SC. Of non-responders or
those off study at 6 months (n=24), 2 achieved a late response by 1 year.
Another relapse occurred in 8 patients, and cumulative incidence at median follow-up
was 39%. Of those who were refractory to or relapsed (n=28) after alemtuzumab, 4 (15%)
proceeded to HSCT, 12 (43%) to other medical AA therapies, and 10 (37%) to both. Subsequent
treatments included eltrombopag (n=13), r-ATG (n=8) CSA (n=6), androgen (n=3) or other
(n=4); response to further treatment occurred in 2 (29%) of relapsed and 5 (38%) of
refractory patients, with response unknown in 7.
Clonal evolution occurred in 9 patients; cumulative incidence at median follow-up
was 23%. Of these, 4 were non-responders. Evolution was high risk (morphological MDS,
AML, or chromosome 7 abnormality) in 7 patients.
Overall survival was 67% at median follow-up (Figure 1). At last follow-up, 10 of
the 18 responders (56%) had durable response and were both transfusion- and AA therapy-independent.
Patients had prolonged immunosuppression after alemtuzumab, with CD4+ T cell counts
<200 in 81% of assessed patients at 6 months, 67% at 1 year, and 42% at 2 years.
Image:
Summary/Conclusion: Alemtuzumab is effective for relapsed SAA. The rate of hematologic
response was greater with IV rather than SC administration. Most responders achieved
durable long-term hematologic improvement, and the rate of clonal evolution was similar
to that seen in our SAA patients treated with rabbit ATG. Immunosuppression can persist
for years following alemtuzumab therapy, requiring long-term monitoring and antimicrobial
prophylaxis.
P806: IMPACT OF MARROW HEMOPHAGOCYTOSIS IN DIAGNOSTIC DEFINITION AND PROGNOSTIC SIGNIFICANCE
IN ADULT POPULATION
C. Almeida1,*, T. Cardoso2, A. Roque1, S. Moreira2, J. Cascais Costa2, N. Silva2,
L. Santos2, C. Geraldes1
1Hematology Department; 2Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
Background: Hemophagocytic lymphohistiocytosis (HLH) is associated with high mortality
and is an entity difficult to diagnose. Bone marrow hemophagocytosis (BMHF) is the
most easily accessible histomorphological criterion, but per si is neither specific
nor sensitive for the diagnosis of HLH and its clinical significance is still not
clear, mainly in adult population
Aims: To evaluate the clinical and prognostic significance of BMHF.
Methods: Retrospective analysis of adult patients with documentation of BMHF between
2007 and 2020. The identification of the patients was carried out by searching the
database by the following keywords: “phagocytosis”, “phagocyte” and “histiocyte”.
For the definition of HLH, the HLH-2004 and the revised HLH-2009 criteria were applied.
HScore values >169 were considered highly suggestive of HLH. Stratified models for
each of the risk scores (HLH-2004 and HLH-2009) were compared using Akaike’s information
criterion (AIC) and the Harrell’s concordance index (C-index).
Results: Ninety-five patients (pts) were included, 63.2% males with median age of
64 (20-95) years old. Twenty-two pts (23.2%) presented ≥5 HLH-2004 criteria and 24
(25.3%) HLH-2009 criteria, with only 6 patients (27.3%) with simultaneous diagnosis
by both criteria (Cohen’s kappa 0.0253). Forty-three (45.3%) patients presented ≥3
HLH-2004 criteria and 34 pts (35.8%) had Hscore>169.
After a median follow-up of 24.3 months, the median overall survival (OS) was 39.4
months. OS was significantly lower in patients with ≥5 HLH-2004 (0.9 vs 86.5 M; HR
3.56; p<0.001), with ≥3 HLH-2004 criteria (3.17 vs NR; HR 2.95; p<0.001), with HLH-2009
criteria (5.5 vs 78.0 M; HR 1.89; p=0.034), and HScore>169 (1.45 vs 86.5; HR 3.08;
p<0.001), compared with those that do not have.
Excluding variables included in the above scores, the only clinical/analytical variable
that was statistically significant for OS in univariate analysis was LDH>UNL (HR 1.02;
p<0.001).
After multivariate analysis for age, aetiology and LDH>ULN, ≥5 HLH-2004 (HR 4. p<0.001),
HLH-2009 2009 (HR 1.87 p=0.045), ≥3 HLH-2004 (HR 3.06; p<0.001) and Hscore>169 (HR
3.39; p=0.001), the four scores retain significance for OS.
In patients with BMHF, HLH-2004 criteria discriminated best between patients with
poor and favourable OS than HLH-2009 (C-index 0.6364 vs 0.5601; AIC 400.24 vs 411.53).
Concerning the aetiology, 51.6% was neoplastic (24.5% [n=12] with ≥5 HLH-2004 criteria),
13.7% infectious, 28.4% autoimmune and in 28,4% the trigger was not identified.
Summary/Conclusion: In our cohort, HLH-2004 criteria had the best predictive value
for OS, compared with HLH-2009
However, in the absence of the data need to calculate these scores, both ≥3 HLH-2004
criteria and Hscore>169 are independent predictors of OS, which, due to the deleterious
prognosis of HLH, support their utility to start treatment earlier
P807: APLASTIC ANEMIA FOLLOWING THE SARS-COV-2 VACCINE
R. Babakhanlou1,*, T. Kadia1, K. Chien1, P. Thompson1
1Leukemia, MD Anderson Cancer Center, Houston, United States of America
Background: There have been several reports describing the possible relationship between
the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccine and the development
of hematological diseases, such as autoimmune hemolytic anemias (AIHA), paroxysmal
nocturnal hemoglobinuria or immune thrombocytopenic purpura. Limited data is available
on the relationship between the SARS-CoV-2 vaccine and the development of aplastic
anemia (AA). We report a series of four cases of acquired AA developing in patients,
who had received an mRNA-based SARS-CoV-2 vaccine.
Aims: To study the temporal association of COVID-19 vaccination and acquired aplastic
anemia
Methods: We reviewed the cases of all patients who presented to MD Anderson Cancer
Center between January 2021 and December 2021 for pancytopenia. We identified patients
who were diagnosed with aplastic anemia after receiving two doses of the SARS-CoV-2
vaccine and summarized baseline characteristics and clinical course.
Results: Four patients with newly diagnosed AA were identified. All patients were
male with a median age of 56 years (range, 19-69). Two patients (50%) had received
the Pfizer-BioNTech vaccine and two (50%) the Moderna mRNA-1273 vaccine. One of the
patients had contracted COVID 8 weeks prior to his vaccination. Three patients suffered
from chronic medical problems including type 1 diabetes, hyperlipidemia, and hypertension.
None of those patients developed any symptoms after the first dose of the vaccine.
Symptoms occurred on average three weeks after the second vaccine and have been outlined
in Table 1. Most common symptoms were fatigue, bruising and bleeding, development
of petechiae, dizziness and shortness of breath. Specific autoimmune workup was done
and all patients received baseline bone marrow aspiration and biopsy with cytogenetics
and next-generation sequencing. In all four patients cytogenetics showed a diploid
karyotype with no chromosomal abnormalities. Flow cytometry and molecular diagnostics
did not show any abnormalities.
In two cases the bone marrow was hypocellular and in two cases the bone marrow was
acellular. All patients received combination immunosuppressive therapy with steroids,
horse anti-thymocyte globulin, cyclosporine and eltrombopag, three on a clinical trial
and one off protocol.
Patients were on therapy for a median of 26 weeks (16-48). One patient achieved complete
remission at 12 weeks. One patient has remained transfusion dependent, while the remaining
two patients became transfusion independent at six months.
Image:
Summary/Conclusion: Although we cannot prove a causative relationship, in rare circumstances,
the SARS-CoV-2 vaccine could be associated with the development of AA. Additional
epidemiologic and laboratory studies are planned to further evaluate any potential
causative relationship.
P808: CONGENITAL NEUTROPENIA PREVALENCE AMONG POLISH CHILDREN – SUMMARY OF A NATIONWIDE
GENETIC SCREENING CAMPAIGN
K. Bąbol-Pokora1,*, W. Dobrewa1, M. Bielska1, J. Madzio1, S. Janczar1, W. Młynarski1
1Department of Pediatrics Oncology and Hematology, Medical University of Lodz, Lodz,
Poland
Background: Severe congenital neutropenia (SCN) is a heterogenous group of rare primary
immunodeficiency disorders, characterized by impaired maturation of neutrophil granulocytes.
Patients with severe congenital neutropenia are prone to recurrent gingivitis, mouth
and rectal ulcerations, and often life-threatening bacterial infections. They are
also predisposed to myelodysplastic syndromes or acute myeloid leukemias. Nine types
of severe congenital neutropenia have been distinguished so far with the most frequent
pathogenetic defects in the ELANE gene. This study explores the distribution and mutation
spectrum of disease-causing genetic variants associated with congenital neutropenia
in the Polish pediatric cohort and the observed ten-fold increase in the referral
rate following a nationwide information campaign.
Aims: The aim of this study was to estimate prevalence of congenital and autoimmune
neutropenia among Polish children and to improve the efficiency of recruiting patients
with suspected SCN by launching the FIX-NET project with a nationwide advertising
campaign.
Methods: Molecular analyses were performed either by direct Sanger sequencing (78
patients recruited in 2008-2015) or by Targeted NGS (201 patients recruited in 2016-2021)
using Illumina platform, focused on the 54 genes related to SCN and neutropenia associated
syndromes. Overall 279 neutropenic patients were enrolled in the studies and written
informed consent has been given. The following results describe the largest cohort
of genetic variation associated with SCN in Poland.
Results: We identified pathogenic changes in genes associated to severe congenital
neutropenia or related syndromes among 66 patients, most of which were variants in
the ELANE gene (44%). There were a number of patients with pathogenic changes in CXCR4
(12%) and SBDS (11%) genes, and also in the recently described dominant SCN-related
genes – SRP54 (8%) and CLPB (5%). Novel mutations accounted for 30% of all reported
variants. In addition among 32 patients we found changes in genes that cause neutrophil
dysfunctions or may lead to autoimmune neutropenia, such as AIRE, FAS, or CTLA4.
Summary/Conclusion: Our study shows the prevalence of congenital neutropenia in Poland
in the largest pediatric neutropenic cohort described so far. It indicates that since
the introduction of NGS to the diagnosis of primary and secondary neutropenia, more
genetic changes have been detected, however, the greatest impact on increasing awareness
among doctors, and thus improving the effectiveness of recruitment had the nationwide
information campaign.
P809: MODULATION OF ARACHIDONIC ACID PATHWAY BY IBUPROFEN RESULTS IN COMPLETE HEMATOLOGICAL
RESPONSE IN GHOSAL HEMATODIAPHYSEAL DYSPLASIA WITHOUT NEED FOR CORTICOSTEROID
N. Barrett1,*, C. McDonnell2,3, M. Cotter4
1Paediatric Haematology, Royal Hospital for Children, Glasgow, United Kingdom; 2Paediatric
Endocrinology, CHI Temple Street; 3Discipline of Paediatrics, Univerisity of Dublin
Trinity College Dublin; 4Paediatric Haematology, CHI Temple Street, Dublin, Ireland
Background: Ghosal hematodiaphyseal dysplasia (GHDD) is a rare autosomal recessive
disorder characterized by pancytopenia and increased bone density of the long bones.
First described in families of Indian and Middle Eastern descent, subsequent studies
identified the genetic lesion in this disease as due to loss of function mutations
in the TBXAS1 gene encoding thromboxane synthase, a key component of the arachidonic
acid pathway responsible for the formation of thromboxane A2 from prostaglandin H2.
As well as pancytopenia, laboratory features include platelet aggregation defects
and increase in osteoblastic activity, the latter being the basis of increased bone
density in patients with Ghosal syndrome. Simultaneously, loss of thromboxane synthase
activity may be associated with shunting of prostaglandin precursors away from production
of thromboxane into production of pro-inflammatory prostaglandins. This may be the
mechanism responsible for the observed marrow suppression seen in these patients.
We describe a 3 year old female of non-consanguineous parents of Filipino origin who
presented with marked pancytopenia. Next Generation Sequencing bone marrow failure
gene panel revealed a novel homozygous mutation in TBXAS1. Subsequent DXA scanning
revealed increased bone mineral density consistent with the diagnosis (Z -score +2.4
for lumbar spine (L1-4)) although clinical sequelae of bony disease were not apparent.
Aims: While multiple case reports document that these patients have hematological
response to long term corticosteroid therapy, we hypothesized that inhibition of cyclooxygenase
(COX) upstream of thromboxane synthase in the arachidonic acid pathway may be effective
in this condition.
Methods: Patient was treated with ibuprofen at regular therapeutic doses (approximately
30 mg/kg/24 hours daily initially) and serial full blood counts were monitored.
Results: The patient exhibited a rapid and complete response to ibuprofen with normalization
of full blood counts. The dose of ibuprofen was reduced to 10 mg/kg/day, and hematologic
response is maintained on low dose after one year.The treatment is well tolerated
with no side effects.
Image:
Summary/Conclusion: Treatment of GHDD with ibuprofen is a novel approach that offers
clear potential benefits in comparison to the known adverse effects profile of long
term corticosteroids. Bone density scan and markers are in process to ascertain the
impact of this treatment on bone density, although this may take longer to show beneficial
effects. The treatment was easily tolerated and well managed by the patient and family
at home with minimum disruption to her quality of life.
P810: BONE MARROW FAILURE IN PATIENTS CARRYING VARIANTS ON CARD11 GENE.
A. Beccaria1,*, A. Grossi2, F. Fioredda3, M. Lanciotti3, E. Palmisani3, P. Terranova3,
M. Lupia3, G. Dell’Orso3, I. Ceccherini2, C. Dufour3, M. Miano3
1DOPO Clinic and Hematology Unit, Division of Pediatric Hematology and Oncology; 2Laboratory
of Genetics and Genomics of Rare Diseases; 3Hematology Unit, Division of Pediatric
Hematology and Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
Background: An accurate differential diagnosis in children with Marrow Failure (MF)
is crucial for the clinical management of the disease. In addition to the classical
congenital MF (cMF), the role of Inborn Errors of Immunity (IEI) have been recently
highlighted as potential cause of the disease. CARD11 is a membrane protein acting
as a key signaling scaffold which controls the antigen-induced lymphocyte activation
during the adaptive immune response (NF-kB, JNK and mTOR pathway). Germline CARD11
mutations can result in gain/loss of function of the protein leading to different
phenotypes (SCID, BENTA disease, atopy, CVID). Several heterozygous hypomorphic/dominant
negative (DN) variants of CARD11 have demonstrated a loss of protein function, exhibiting
high penetrance and variable expressivity. In particular, mutations in the GUK (Guanylate
Kinase) domain seem to be involved in modulating the self-inactivating capacity of
CARD11. No CARD11 variants have ever been described to be associated with MF. Herein,
we report 4 patients with MF carrying variants on the CARD11 gene.
Aims: To evaluate the clinical/immunological features and genetic profile of patients
with MF carrying CARD11 variants and followed in our Centre.
Methods: A restrospective review of clinical, immunological and genetic data was performed
from patients’ records. Molecular analysis was conducted using Next-Generation Sequencing
(NGS), targeted panels including genes involved in both cMF and IEI, or by Whole Exome
Sequencing (WES). Filtering of variants was performed according to Mendelian disease
segregation and zigosity (OMIM), in silico prediction of pathogenicity (ACMG criteria
in “Varsome”), and minor allele frequency (reported in GnomAD).
Results: Four children carrying variants in the CARD11 gene presented with MF as initial
sign of the disease (n=2) or with immune cytopenias further evolving into MF (n=2).
Their clinical/immunological features, genetic profile, treatment and follow-up are
reported in Table1. Lymphocytes subsets analysis revealed a deficiency of B memory,
Tregs and an increased of HLADR+ T cells in all patients. Two cases showed elevated
CD4-CD8- TCR αβ+ T-cells (“double negatives T-cells”). All patients showed autoimmune
stigmata and 2 of them also presented autoimmune hepatitis. No patients responded
to first-line treatment. One responded to second-line treatment with Mychofenolate-mofetil
(MMF) and periodic infusion of immunoglobulins. The remaining 3 patients underwent
hematopoietic stem cell transplantation (SCT) from haploidentical-αβCD19 depleted
transplant (n=2) or HLA-identical cousin (n=1) (Table1).
Image:
Summary/Conclusion: This report highlights novel phenotypic characteristics of patients
carrying variants in CARD11 such as MF and autoimmune hepatitis, thus widening the
clinical spectrum of the disease. This underlines the need for an enlarged molecular
analysis in pediatric cases of MF and suggests that, in some patients, immune-mediated
destruction of blood and marrow cells cooperate in generating the cytopenia. Three
variants detected in our cohort are located in the GUK domain of the gene, close to
other reported pathogenic dominant negative mutations leading to haploinsufficiency,
suggesting interference with the CARD11’s ability to self-inactivate. Functional study
will be necessary to confirm the pathogenic role of such variants. Although treatment
with MMF may be considered, SCT represents the only curative option for such patients.
P811: A SINGLE CENTER HISTORICAL CONTROL STUDY OF ELTROMBOPAG ADDED TO IMMUNOSUPPRESSIVE
THERAPY FOR SEVERE APLASTIC ANEMIA IN CHILDREN
B. yang1,*, L. fu1, H. li1, H. chen1, J. ma1
1Hematology Oncology Center, Beijing Children Hospital, Beijing, China
Background: Severe aplastic anemia is caused by immune-mediated destruction of bone
marrow. Standard immunosuppressive therapy (rabbit anti-thymocyte globulin combined
with cyclosporine) is effective, but the effect needs to be improved. A thrombopoietin-receptor
agonist, eltrombopag, added to standard immunosuppressive therapy led to clinically
significant increases in blood counts. We combined eltrombopag and standard immunosuppressive
therapy in pediatric severe aplastic anemia patients.
Aims: To evaluate the efficacy and safety of eltrombopag added to immunosuppressive
therapy for severe aplastic anemia in children.
Methods: The clinical data of patients with severe aplastic anemia and received immunosuppressive
therapy from March 2013 to July 2020 were collected and analyzed retrospectively.
We conducted a historical control study, in which the patients treated alone with
immunosuppressive therapy were used as the control group. We compared the cases between
control group and eltrombopag group, included hematological response, effective time,
relapse, clonal evolution, adverse reactions, event free survival (EFS) and overall
survival (OS).
Results: 1. A total of 115patients (60 males), median aged 5.77 years, median follow-up
time was 45 months, were enrolled in this study. All patients were diagnosed with
severe or very severe aplastic anemia, including 49 patients in the historical control
group and 66 patients in the eltrombopag group. 2. The complete response rate (CRR)
at 3 months was 49.0% in eltrombopag cohort and 8.2% in historical control. The overall
response rate (ORR) at 3months was 63.6% and 30.3% respectively. The CRR at 6 months
was 50.0% in eltrombopag cohort and 10.2% in control cohort. The ORR at 6months were
71.2% and 57.1% respectively. There were significant differences of the CRR between
two groups, there was no significant difference between two groups.3. The median effective
time of the control group and eltrombopag group was 90 days and 71 days respectively
(P = 0.007), the median complete remission time was 360 days and 98 days respectively
(P < 0.001). The median time of separated from granulocyte colony stimulating factor
(G-CSF), Red Blood Cell transfusion and Platelet transfusion in the control group
were 105 days, 46 days and 54 days respectively, and 68 days, 45 days and 45 days
in the eltrombopag group. There were significant differences in the time of separated
from G-CSF (P = 0.003), Red Blood Cell transfusion (P = 0.001) and Platelet transfusion
(P < 0.001) in the two groups. 4.The relapse rate was 4.1% (n = 2) in the control
group and 10.2% (n = 5) in the eltrombopag group. One patient (2.0%) in the control
group developed clonal evolution and progressed to myelodysplastic syndrome.5. The
common adverse reaction were transient and reversible hyperbilirubinemia (13.6%, n
= 9), elevated liver enzymes (4.5%, n = 3) and hyperuricemia (1.5%, n = 1).6. The
1-year OS and 2-year or of the control group was 100% and 98.5% respectively, comparing
to 95.9% and 97.0% in eltrombopag group. The 2-year EFS of the control group and the
eltrombopag group was 79.6% and 74.2%, respectively, and the 3-year EFS were 73.5%
and 72.7%, respectively.
Summary/Conclusion: The addition of eltrombopag to standard immunosuppressive therapy
was associated with markedly higher rates and effected more quickly of hematologic
response among pediatric severe aplastic anemia patients than in a historical cohort
and showed a reliable security.
P812: LONG-TERM COMPLEMENT INHIBITION AND SURVIVAL OUTCOMES IN PATIENTS WITH PAROXYSMAL
NOCTURNAL HEMOGLOBINURIA: AN INTERIM ANALYSIS OF THE RAVULIZUMAB CLINICAL TRIALS
A. Kulasekararaj1,*, R. Brodsky2, M. Griffin3, A. Kulagin4, M. Ogawa5, J. Wang5, A.
Mujeebuddin5, J.-I. Nishimura6, R. Peffault de Latour7, J. Szer8, J. W. Lee9
1King’s College Hospital, National Institute of Health Research/Wellcome King’s Clinical
Research Facility and King’s College London, London, United Kingdom; 2Division of
Hematology, Johns Hopkins Medicine, Baltimore, United States of America; 3St James
Hospital, NHS Teaching Hospitals, Leeds, United Kingdom; 4RM Gorbacheva Research Institute,
Pavlov University, St Petersburg, Russia; 5Alexion, AstraZeneca Rare Disease, Boston,
MA, United States of America; 6Osaka University Graduate School of Medicine, Suita,
Japan; 7Hôpital Saint-Louis AP-HP, Paris, France; 8Peter MacCallum Cancer Centre and
The Royal Melbourne Hospital, Melbourne, VIC, Australia; 9Department of Hematology,
Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea,
Seoul, South Korea
Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare, chronic, hematologic
disorder characterized by uncontrolled terminal complement activation, intravascular
hemolysis, thrombotic events and significant morbidity and mortality. Complement component
5 (C5) inhibitors, eculizumab and ravulizumab, are the current standard of care in
patients with PNH. Eculizumab has been the mainstay of treating patients with PNH
since approval by both US Food and Drug Administration (FDA) and European Medicines
Agency (EMA) in 2007. Ravulizumab (approved in 2018 [FDA] and 2019 [EMA]) is a new
treatment for patients with PNH, offering complete complement C5 inhibition throughout
the 8-week (q8w) dosing interval. The ravulizumab clinical trial program has allowed
for analysis into the long-term management of patients with PNH treated with C5 inhibitors
and their associated clinical outcomes.
Aims: To analyze patient survival using long-term data from the ravulizumab clinical
trial program.
Methods: This analysis utilized pooled long-term survival data across the ravulizumab
phase 1b, 2 and 3 trials (103 [NCT02598583], 201 [NCT02605993], 301 [NCT02946463]
and 302 [NCT03056040]) with up to 4 years of open-label extension. Complement C5 inhibitor
(eculizumab/ravulizumab) and dose received varied during the initial treatment period.
During open-label extension, patients received weight-based dosing of intravenous
ravulizumab q8w. Deaths reported, patient survival over time and adverse events resulting
in patient death were analyzed. Instances of patients that died owing to infection
or sepsis were specifically analyzed to provide insight into the events leading to
death.
Results: This analysis reported 1479.0 patient-years of follow-up in 475 patients
with PNH treated with ravulizumab. Of the 475 patients who received ravulizumab, 12
(2.5%) died during the initial treatment period and 4-year open-label extension with
an overall incidence of 0.8 per 100 patient-years. Nine of these deaths occurred within
the first 3 years of treatment. The remaining three patients died during the fourth
(n = 2) and fifth (n = 1) years of treatment. Of the 12 deaths reported, six were
attributed to infection or sepsis (Table 1). These patients were predominantly male
(83.3%), and white or Asian (50.0%, respectively), and median (range) age was 61 (43–75)
years. In addition, of these six patients, two had aplastic anemia and one patient
had pancytopenia. Most (83.3%) of these patients were hospitalized owing to onset
of infection; however, one patient (white male; 43 years old; ravulizumab to ravulizumab)
was hospitalized owing to worsening of aplastic anemia. During hospitalization, this
patient was diagnosed with acute respiratory infection and died owing to sepsis. One
patient (Asian male; 62 years old; ravulizumab to ravulizumab) died owing to meningococcal
sepsis (strain unknown). This patient was vaccinated against meningococcal groups
A, C, Y and W-135, but had not received prophylactic antibiotics. Other causes of
death included cardiac disorders (n = 1), neoplasms (n = 4; acute myeloid leukemia,
metastatic urothelial carcinoma of the kidney, metastatic malignant neoplasm of the
lung and non-small cell lung carcinoma) and respiratory disorders (n = 1).
Image:
Summary/Conclusion: This analysis reports the longest period of follow-up in 475 patients
with PNH treated with ravulizumab. Overall, long-term ravulizumab treatment was associated
with a low incidence of death and few patients died owing to infection. This analysis
supports the long-term use of ravulizumab for PNH.
P813: EFFICACY, TREATMENT ADMINISTRATION SATISFACTION AND SAFETY OF SUBCUTANEOUS RAVULIZUMAB
THROUGH 1 YEAR IN PATIENTS WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA WHO RECEIVED PRIOR
INTRAVENOUS ECULIZUMAB
M. N. Yenerel1,*, F. Sicre de Fontbrune2, C. Piatek3, F. Sahin4, W. Füreder5, M. Ogawa6,
I. Tomazos6, H. Doll7, A. Ozol-Godfrey6, J. R. Sierra6, J. Szer8
1Division of Hematology, Department of Internal Medicine, Istanbul Faculty of Medicine,
Istanbul University, Istanbul, Turkey; 2Hematology and Bone Marrow Transplant Unit,
Assistance Publique Hôpitaux des Paris, Saint Louis Hospital, Paris, France; 3Jane
Anne Nohl Division of Hematology, Keck School of Medicine, University of Southern
California, Los Angeles, CA, United States of America; 4Department of Hematology,
Ege University Bornova, Izmir, Turkey; 5Division of Hematology and Hemostaseology,
Medical University of Vienna, Vienna, Austria; 6Alexion, AstraZeneca Rare Disease,
Boston, MA, United States of America; 7Clinical Outcomes Solutions, Folkstone, United
Kingdom; 8Haematology at Peter MacCallum Cancer Centre and The Royal Melbourne Hospital,
Melbourne, VIC, Australia
Background: The efficacy of ravulizumab (intravenous [IV] formulation; administered
every 8 weeks) for the treatment of patients with paroxysmal nocturnal hemoglobinuria
(PNH) has been demonstrated in several randomized trials (NCT02946463, NCT03056040,
NCT03406507). In study 303 (NCT03748823), subcutaneous (SC) ravulizumab, administered
weekly via an on-body delivery system, showed pharmacokinetic non-inferiority to IV
ravulizumab in adult patients with PNH who were clinically stable on prior IV eculizumab
treatment. Here, we report results from the first 1 year of SC treatment, starting
at day 15 for patients who continued SC ravulizumab during the extension period (SC/SC)
and day 71 for patients who switched from IV ravulizumab to SC ravulizumab (IV/SC).
Aims: To evaluate the efficacy, treatment administration satisfaction and safety of
SC ravulizumab through the first 1 year (day 351) of treatment in adult patients with
PNH previously treated with eculizumab.
Methods: Patients (≥ 18 years) with clinically stable PNH (lactate dehydrogenase [LDH]
levels ≤ 1.5 × upper limit of normal [246 U/L]) and ≥ 3 months prior eculizumab treatment
were enrolled in the study, which consisted of a screening period (day -1 to day -30),
a 10-week randomized treatment period and an extension period of up to 172 weeks.
During the randomized treatment period, patients were assigned (2:1 ratio) to receive
either SC ravulizumab or IV ravulizumab; all patients received SC ravulizumab during
the extension period. Efficacy endpoints included: change in LDH from baseline; incidence
of breakthrough hemolysis; transfusion avoidance; and stabilized hemoglobin (avoidance
of a ≥ 2 g/dL decrease in hemoglobin in the absence of transfusion). Treatment administration
satisfaction was assessed via the Treatment Administration Satisfaction Questionnaire
(TASQ), which has been validated in a PNH population. Safety, including adverse events
(AEs), serious AEs (SAEs) and adverse device effects (ADEs), were also assessed up
to the 1-year data cut-off.
Results: In total, 128 patients received SC ravulizumab (SC/SC: n = 84; IV/SC: n =
44; mean [range] duration of SC treatment: 486.4 [37–709] days). Efficacy endpoints
(SC/SC and IV/SC) remained stable over time through 1 year of SC ravulizumab treatment.
Mean (standard deviation [SD]) percentage change in LDH from baseline to SC day 351
was 0.9% (20.5%). Breakthrough hemolysis events were infrequent: 5/128 patients (3.9%);
no event was considered free C5-related. Transfusion avoidance was maintained in 83.6%
of patients during SC treatment, and 79.7% achieved stabilized hemoglobin. Improvement
in total TASQ score with SC ravulizumab (compared with baseline IV eculizumab) was
apparent at the first post-SC treatment assessment (SC day 29) and maintained until
data cut-off (Figure). The most common AEs (reported by ≥ 10% of patients, excluding
ADEs related to device product issues) during SC treatment were headache (14.1%, all
grade ≤ 2), COVID-19 (14.1%, one death) and pyrexia (10.9%); injection site reaction
(4.7%) was the most common non-device related ADE. Treatment-emergent SAEs were experienced
by 21.1% of patients through to data cut-off. Although many patients had ≥ 1 device
issue ADE, full SC dose administration was achieved in 99.9% of attempts. ADE incidence
decreased over time.
Image:
Summary/Conclusion: The SC method of administration provides an additional treatment
option for patients with PNH receiving ravulizumab therapy. Patients may be switched
from IV eculizumab or IV ravulizumab to SC ravulizumab without loss of efficacy.
P814: CLINICAL CHARACTERISTICS AND GENE MUTATION ANALYSIS OF 148 CHILDREN WITH FANCONI
ANEMIA IN CHINA
L. Chang1,*, L. zhang1, W. An1, Y. wan1, Y. cai1, Y. Lan1, M. Ruan1, X. liu1, Y. Zou1,
X. Zhu1
1State Key Laboratory of Experimental Hematology, National Clinical Research Center
for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology &
Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical
Colleg, Tian jin, China
Background: Fanconi anemia (FA) is a rare autosomal recessive, X-linked (FANCB) or
autosomal dominant (FANCR/RAD51) disease of bone marrow failure. Pathogenesis is mainly
caused by gene mutation related to the FA pathway. At present, at least 23 genes have
been discovered to play a role in the FA pathway. The gene mutations of FA have been
reported in more and more countries. In China, there no large-scale cases have been
reported.
Aims: In this study, we analyzed the clinical characteristics of 148 FA children from
20 provinces in China from October 2003 to October 2021. We explored the relationship
between genotype and phenotype in 105 children with FA genotyping results. This is
the largest series of subtyped Chinese Fanconi anemia patients to date, and the results
will be helpful for future clinical management.
Methods: A total of 148 Fanconi anemia patients from 140 families were diagnosed by
clinical phenotype, family history, chromosome breakage test induced by mitomycin
C, Single cell gel electrophoresis experiment, targeted - sequence, whole exon sequencing,
and MLPA method from October 2003 to October 2021. Since there were no genetic test
results for the children before 2010, only the 105 patients after 2010 had gene sequencing
results, which allowed genotyping. DNA damage repair defects were detected by MMC-induced
Single cell gel electrophoresis and chromosome breakage experiments. Targeted-seq
was used to identify the FA mutations of the patients. Multiplex ligation-dependent
probe amplification (MLPA) was performed to detect large fragment deletions in patients
with negative results in the targeted-seq but conforming to the FA by MMC-induced
chromosome breakage experiments test and clinical manifestations. We performed WES
of DNA samples from five patients and their parents.
Results: 1) The most common subtype of FA in China is FA-A, followed by FA-D2 and
FA-P。2) The most common deformities in FA-A patients are finger deformities and skin
pigmentation, while heart deformities are more common in FA-D2/B/G/I/P subtypes. 3)
The common mutations of FANCA are exon23c.2101A>G, exon28c.2778 + 1G>A, exon34c.3348 + 1G>A,
exon21c.1844delC, and exon30 C2941T>G, which are different from Korea and Japan. 4)
Homozygous nonsense mutation of exon32 c.3188G>A and exon29 c.2851C>T in FA-A likely
benign, but the homozygous splicing mutation in exon29c.2852 + 1G>T is harmful. 4)
Children with splicing and Del mutations of FANCD1 gene had poor clinical prognosis,
while children with homozygous mutations of exon10c. 1792A>G is likely benign. 5)
Cases of initial diagnosis of MDS or poor disease progression tend to occur in FA-B/C/E/G/J/L/M
and FA-S subtypes. 6)Chromosome 1, 3, 7, and 8 abnormalities and mutations of SF3B1
P53 are related to disease progression.
Summary/Conclusion: We made a comprehensive description and analysis of the clinical
characteristics and mutant genes of Children with FA in China. We proposed the mutation
sites related to clinical prognosis and the cytogenetic abnormalities of disease progression,
providing new data for our comprehensive understanding of FA. Our data will be useful
for FA future menagement.
P815: PHARMACOKINETIC AND PHARMACODYNAMIC SIMILARITY OF ABP 959 AND ECULIZUMAB REFERENCE
PRODUCT: UNBOUND ECULIZUMAB AND CH50 FROM THE RANDOMIZED, DOUBLE-BLIND, SINGLE-DOSE
STUDY IN HEALTHY VOLUNTEERS
V. Hanes1, J. Pan1, D. Mytych1, V. Chow1,*
1Amgen Inc, Thousand Oaks, United States of America
Background: ABP 959 is being developed as a biosimilar to eculizumab reference product
(RP). ABP 959 and the RP are humanized recombinant IgG2/4κ mAbs which bind with specificity
to the human complement C5, blocking its cleavage to C5a and C5b and preventing the
generation of the terminal complement complex (TCC), thereby blocking cell lysis.
Excessive activation or insufficient control of C5 is believed to play a role in the
pathogenesis of paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic
syndrome (aHUS), generalized myasthenia gravis (gMG), and neuromyelitis optical spectrum
disorder (NMOSD). Drug binding to C5 blocks this biological function and contributes
to efficacy across these indications for which the RP is approved.
Aims: The goal of this analysis is to further support the previously demonstrated
pharmacokinetic (PK)/pharmacodynamic (PD) similarity of ABP 959 with eculizumab RP
by providing additional data on similarity in PK exposure with respect to unbound
eculizumab RP and 50% total hemolytic complement activity (CH50).
Methods: This was a randomized, double-blind, single-dose, 3-arm, parallel-group trial
in healthy adult male subjects, aged between 18 and 45 years, with a body mass index
of 18 to 30.0 kg/m2, who were randomized following informed consent, to receive a
300-mg IV infusion of ABP 959, or FDA-licensed eculizumab RP (eculizumab US), or EU-authorized
eculizumab (eculizumab EU). Serum samples for PK and PD evaluations were collected
over 56 days. Primary objectives were to demonstrate PK and PD equivalence of ABP
959 versus eculizumab US and ABP 959 versus eculizumab EU, as assessed by area under
the total serum concentration versus −time curve (AUC) from time 0 extrapolated to
infinity (AUCinf) and an area between the effect curve (ABEC) of 50% total hemolytic
complement activity (CH50). Secondary PK endpoints included AUC from time 0 to the
time of the last observed quantifiable concentration (AUClast) and maximum observed
concentration (Cmax). Pre-specified equivalence criterion for the primary PK and PD
parameters was 90% confidence interval (CI) for geometric means (GM) ratio within
0.80 to 1.25. Other secondary endpoints included the safety, tolerability, and immunogenicity
of the investigational products (IPs).
Results: A total of 219 subjects were randomized (ABP 959, n=71; eculizumab US, n=74;
eculizumab EU, n=74). As with PK and PD parameters of total eculizumab following a
single 300 mg IV infusion of IP,1 PK/PD parameters for unbound eculizumab were similar
between ABP 959 versus eculizumab US and ABP 959 versus eculizumab EU.
The GM and GM coefficient of variation (%CV) of AUCinf (h.µg/mL) for unbound eculizumab
was: 5072.1 (25) for ABP 959, 5527.6 (31) for eculizumab US, and 5070.3 (31) for eculizumab
EU.
The GM and %CV of ABEC of CH50 (%*h) was 17724.5 (38.75) for APB 959, 16549.4 (42.23)
for eculizumab US, and 16361.1 (36.94) for eculizumab EU. The resulting 90 and 95%
CI of GM ratios were within the bioequivalence criteria of 0.80 to 1.25.
1Eur J Haematol. 2020;105:66–74.
Image:
Summary/Conclusion: These additional clinical pharmacology results demonstrate bioequivalence
of ABP 959 to eculizumab RP with regard to unbound eculizumab and CH50. These results
further support the PK/PD similarity of ABP 959 with eculizumab RP.
P816: TRANSIENT MONOSOMY 7 IN SAMD9/9L SYNDROMES: IS IT SAFE TO WATCH AND WAIT?
M. Erlacher1,*, F. Andresen1, A. Yoshimi1, M. Sukova2, J. Stary2, B. de Moerloose3,
M. Dworzak4, S. Polychronopoulou5, G. Göhring6, C. Kratz7, J. van der Werff ten Bosch8,
M. Seidel9, B. Strahm10, M. Wlodarski10, C. Niemeyer10
1Division of Pediatric Hematology and Oncology, University Medical Center Freiburg,
Freiburg, Germany; 2Department of Pediatric Hematology and Oncology, University Hospital
Motol, Prague, Czechia; 3Department of Pediatric Hematology-Oncology, Ghent University
Hospital, Ghent, Belgium; 4Department of Pediatrics, St. Anna Children’s Hospital
and Children’s Cancer Research Institute, Vienna, Austria; 5Department of Pediatric
Hematology/Oncology, Aghia Sophia Children’s Hospital, Athens, Greece; 6Department
of Human Genetics; 7Pediatric Hematology and Oncology, Hannover Medical School, Hannover,
Germany; 8Department of Pediatric Onco-Hematology, Universitair Ziekenhuis Brussels,
Brussels, Belgium; 9Division of Pediatric Hemato-Oncology, Medical University Graz,
Graz, Austria; 10Division of Pediatric Hematology and Oncology, Medical Center, Faculty
of Medicine, University of Freiburg, Freiburg, Germany
Background: Monosomy 7 is a non-random but poorly understood somatic event in myelodysplastic
syndrome (MDS). Most enigmatic is the fact that monosomy 7 can disappear spontaneously.
Such transient monosomy 7 was recently associated with germline mutations in sterile
alpha-motif domain-containing protein 9 (SAMD9) and its paralog SAMD9-like (SAMD9L),
both located on chromosome 7q. Mechanistically, gain-of-function (GOF) SAMD9/9L mutations
inhibit proliferation and reduce survival in different cell types thus leading to
bone marrow failure, immunodeficiency and non-hematological phenotypes. Hematopoietic
cells are under a high selection pressure to inactivate this toxic gene and achieve
this by multiple mechanisms: uniparental disomy, monosomy 7, del(7q) or somatic loss-of-function
(LOF) mutations of the mutated allele. Multiple “rescued” clones exist in parallel
within the bone marrow of patients, and dominance of an UPD or somatically mutated
clone can displace the monosomy 7 clone and result in hematological remission.
Aims: We recently identified SAMD9/9L mutations in 8% of children with MDS, and a
strong association with monosomy 7 (EWOG-MDS; Sahoo et al, Nat Med, 2021). This retrospective
analysis revealed transient monosomy 7 and spontaneous hematological remission in
some infants. To prospectively study the propensity of patients to lose their monosomy
7 clone, EWOG-MDS elected to delay immediate hematopoietic stem cell transplantation
(HSCT) in children <5 years of age diagnosed of refractory cytopenia (RCC) with germline
SAMD9/9L mutation and monosomy 7.
Methods: Patients with SAMD9 or SAMD9L mutation and monosomy 7, younger than 5 years
of age, were closely monitored with bone marrow morphology, SAMD9/9L mutational analysis
and cytogenetic studies every 3-4 month. HSCT was recommended in case of severe infections,
persistent severe neutropenia, progression with additional cytogenetic aberrations
or increase in blasts (MDS-EB), or persistence of monosomy 7 beyond the age of 5 years.
Results: Eight patients diagnosed at a median age of 11 months (range 4-36 months)
with SAMD9L (n=7) or SAMD9 (n=1) germline mutation were studied. Two (P5 and P8) were
born small for gestational age, P5 presented with global developmental delay and cerebellar
atrophy. With a median follow-up of 27 months, 6 of the 8 patients are alive. One
patient succumbed to infection prior to HSCT (P4), another to transplantation-related
toxicity (P8). HSCT was performed or is currently scheduled in 4 of the 8 patients;
indications were severe neutropenia/infection (P5, P6), immunodeficiency (P7) or MDS-EB
(P8). The monosomy 7 clone regressed in 3 patients (P1-P3) and was no longer detectable
at 3 months (P1) or 1 year (P2) from diagnosis (P3 was lost to follow-up at 12 months).
Somatic SAMD9L mutations were noted in P1 and P2, both of which experienced a complete
hematological and cytogenetic recovery.
In sum, we followed 8 patients with SAMD9/9L syndrome and monosomy 7. Transient monosomy
7 with hematological recovery was observed in 2 patients and accompanied by gain of
somatic rescuing mutations.
Summary/Conclusion: Transient monosomy 7 in young children with SAMD9/SAMD9L germline
disorder and RCC with monosomy 7 is a rare event. Initiation of a watch and wait strategy
instead of timely HSCT will require a stringent surveillance strategy including repetitive
BM analyses for SAMD9/9L sequencing, cytogenetics and search for somatic oncogenic
events.
P817: SYNDROMES PREDISPOSING TO LEUKEMIA ARE A MAJOR CAUSE OF INHERITED CYTOPENIAS
IN CHILDREN
O. Gilad1,2,*, O. Dgany3, S. Noy -Lotan3, T. Krasnov3, J. Yacobovich4, R. Rabinowicz4,
T. Goldberg5, A. Kuperman6, A. Abu-Quider7, H. Miskin8, N. Kapelushnik9, N. Mandel-Shorer10,
S. Shimony11, D. Harlev12, T. Ben-Ami13, E. Adam14, C. Levin15, S. Aviner16, R. Elhasid17,
S. Berger-Achituv17, L. Chaitman-Yerushalmi18, Y. Kodman4, N. Oniashvilli19, M. Hameiri-
Grosman19, S. Izraeli4, H. Tamary1,3,20, O. Steinberg-Shemer1,3,20
1Sackler faculty of Medicine, Tel Aviv University, Tel Aviv; 2Department of Hematology-Oncology,
Scneider Children’s medical center of Israe; 3Pediatric Hematology Laboratory, Felsenstein
Medical Research Center; 4Department of Hematology-Oncology, Schneider Children’s
Medical Center of Israel; 5Department of Hematology-Oncology, Scneider Children’s
medical center of Israel, Petach Tikva; 6Blood Coagulation Service and Pediatric Hematology
Clinic, Galilee Medical Center, Nahariya; 7Pediatric Hematology, Soroka University
Medical Center, Ben-Gurion University, Beer Sheva; 8Pediatric Hematology Unit, Shaare
Zedek Medical Center, Jerusalem; 9Goldschleger Eye Institute, Sheba Medical Center,
Tel Hashomer; 10Department of Pediatric Hematology-Oncology, Rambam Healthcare Campus,
Haifa; 11Institute of Hematology, Davidoff Cancer Centre, Rabin Medical center, Petach-Tikva;
12Pediatric Hematology-Oncology Department, Hadassah University Medical Center, Jerusalem;
13Pediatric Hematology Unit, Kaplan Medical Center, Rehovot; 14Pediatric Hematology-Oncology
Department, Sheba Medical Center, Tel Hashomer; 15Pediatric Hematology Unit and Research
Laboratory, Emek Medical Center, Afula; 16Department of Pediatrics, Barzilai University
Medical Center, Ashkelon; 17Department of Pediatric Hemato-Oncology, Tel Aviv Medical
Center; 18Genoox, Health Care Technology, Tel Aviv; 19Department of Hematology-Oncology,
Schneider Children’s Medical Center of Israel; 20Department of Hematology-Oncology,
Schneider Children’s Medical Center of Israel, Petach Tikva, Israel
Background: Prolonged cytopenias are a non-specific sign with a wide differential
diagnosis. Among inherited disorders, cytopenias predisposing to leukemia require
a timely and accurate diagnosis to ensure appropriate medical management, including
adequate monitoring and stem-cell transplantation prior to the development of leukemia.
Aims: We aimed to define the types and prevalences of the genetic causes leading to
persistent cytopenias in children.
Methods: The study comprises children with persistent cytopenias, myelodysplastic
syndrome (MDS), aplastic anemia, or suspected inherited bone marrow failure syndromes,
who were referred for genetic evaluation from all pediatric hematology centers in
Israel during 2016-2019. For variant detection, we used a custom-made targeted next-generation
sequencing panel covering 226 genes known to be mutated in inherited cytopenias followed,
if needed, by whole exome sequencing. Sanger sequencing was used for validation.
Results: In total, 189 children with persistent cytopenias underwent a genetic evaluation.
Pathogenic and likely pathogenic variants were identified in 59 patients (31.2%),
including 47 with leukemia predisposing syndromes. Most of the latter (32, 68.1%)
had inherited bone marrow failure syndromes, 9 (19.1%) had inherited thrombocytopenia
predisposing to leukemia, and 3 (6.4%) had congenital neutropenia. Twelve patients
had cytopenias with no known leukemia predisposition, including nine children with
inherited thrombocytopenia and three with congenital neutropenia (not yet known to
predispose to leukemia). Importantly only 3 had myelodysplastic syndrome. Thus, the
majority of leukemia predisposition could not have been deduced from the morphological
presence of MDS
Summary/Conclusion: almost one-third of 189 children referred with persistent cytopenias
had an underlying inherited disorder; 79.7% of whom had a germline predisposition
to leukemia. Precise diagnosis of children with cytopenias should direct follow-up
and management programs and may positively impact disease outcome.
P818: NEXT GENERATION TARGETED SEQUENCING FOR ENHANCED GENOTYPING OF DIAMOND BLACKFAN
ANEMIA IN ISRAEL
T. Goldberg1,*, O. Steinberg-Shemer1,2,3, O. Dgany3, S. Noy-Lotan3, T. Krasnov3, O.
Gilad1,2, J. Yacobovich1,2, H. Tamary1,2,3
1Pediatric Hematology Oncology, Schneider Children’s Medical Center of Israel, Petah
Tikva; 2Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv; 3Pediatric Hematology
Laboratory, Felsenstein Medical Research Center, Petah Tikva, Israel
Background: Diamond Blackfan anemia (DBA), an erythroid-specific congenital bone marrow
failure syndrome, is predominantly driven by mutations in one of several genes encoding
ribosomal proteins (RP), leading to protein haploinsufficiency. In recent years, a
number of patients with “DBA-like” syndrome were found to have mutations in non-RP
genes including CECR1. Next generation sequencing (NGS) is rapidly replacing conventional
Sanger sequencing approaches in the molecular diagnosis of patients with DBA and other
inherited bone marrow failure syndromes. Targeted NGS panels enable high-resolution
deep sequencing of all known genes associated with a specific clinical presentation
such as congenital anemias.
Aims: This study aims to use a targeted NGS panel specific for inherited anemias to
genotype previously undiagnosed patients with clinical DBA in Israel. Enhanced genotyping
efficiency provided by NGS may provide an opportunity to evaluate for genotype-phenotype
correlations.
Methods: A targeted NGS panel including over 260 genes was used as an adjunct to Sanger
sequencing and multiplex ligation-dependent probe amplification for genotyping patients
from the Israeli inherited bone marrow failure registry (I-IBMFR) with a clinical
diagnosis of DBA. Over 90% of 48 patients with DBA from the Israeli registry had available
DNA for genotyping.
Results: Forty-eight patients with clinical DBA, registered to the I-IBMFR, have been
followed for a median of 12.1 years. The majority of patients presented with anemia
in the first year of life, with a median age at presentation of 3 months. Forty-four
of these patients underwent genotyping. Pathogenic RP gene alterations were detected
in 31 patients in 9 different RP genes; homozygous CECR1 mutations were found in 8
separate patients. Forty percent of these molecular diagnoses were detected by targeted
NGS panel technology. More than half of the patients exhibited at least one congenital
malformation. Cardiac anomalies were observed broadly in patients with small and large
subunit RP gene mutations. Thumb anomalies were found exclusively in patients with
RPL5 mutations; cleft lip and palate were reported only in patients with RPS10 and
RPL11 mutations. Fewer than half of the patients given a steroid trial were responsive;
patients with RPS19 mutations responded significantly better to steroid treatment
than patients with RPL5 mutations. Just under half of the patients with RP gene mutations
became red blood cell (RBC) transfusion dependent, while almost all of the patients
with CECR1 mutations required RBC transfusions. Three patients, including two genetically
undiagnosed patients, went into spontaneous remission after receiving chronic treatment.
Summary/Conclusion: Use of targeted NGS has dramatically enhanced the efficiency of
genotyping for patients with DBA and other inherited bone marrow failure syndromes,
allowing for focused management of patients and discovery of novel genotype-phenotype
correlations. Ongoing investment in registries and comprehensive genotyping are crucial
in increasing our understanding of this rare disease and optimizing patient care.
P819: TRANSPLANT, TREATMENT AND TRANSFUSION FREE (TTT-FREE) SURVIVAL AS RELEVANT CLINICAL
ENDPOINT AFTER IMMUNOSUPPRESSIVE TREATMENT FOR ACQUIRED APLASTIC ANEMIA IN ADULTS
E. Koster1, C. Halkes1,*, E. Bogers1, C. Hazenberg2, F. Heubel-Moenen3, S. Langemeier4,
E. Meijer5, E. Nur5, M. Raaijmakers6, T. Snijders7, M. de Witte8, J. Tjon1, L. de
Wreede9
1Hematology, Leiden University Center, Leiden; 2Hematology, University Medical Center
Groningen, Groningen; 3Hematology, Maastricht University Medical Center, Maastricht;
4Hematology, Radboud Medical Center, Nijmegen; 5Hematology, Amsterdam Univeristy Medical
Center, Amsterdam; 6Hematology, Erasmus Medical Center, Rotterdam; 7Hematology, Medical
Center Twente, Enschede; 8Hematology, University Medical Center Utrecht, Utrecht;
9Department of Biomedical Data Sciences, Leiden University Center, Leiden, Netherlands
Background: Acquired aplastic anemia (AA) is characterized by an aplastic bone marrow
and pancytopenia. Adult patients with AA can be treated with immunosuppressive treatment
(IST) consisting of ATGAM in combination with cyclosporin (CsA) or with an allogeneic
hematopoietic stem cell transplantation (alloSCT). Patients <40 years with an HLA-identical
sibling preferentially receive an alloSCT. Other patients are treated with IST as
1st line treatment. IST leads to transfusion independency in the majority of patients
but it can take up to 6 months before this response occurs. Some patients need long-term
treatment with CsA to maintain this response and durable efficacy is hampered by relapsing
aplasia or the development of MDS, AML or PNH. While graft versus host disease and
relapse-free survival (GRFS) is used as a favorable composite outcome after alloSCT,
a widely accepted measurement for treatment success after IST is missing.
Aims: In this study we examined Transplant, Treatment and Transfusion-free survival
(TTT-free survival) as a relevant clinical outcome after standard IST with ATGAM and
CsA in AA adult patients. We determined the 5-year TTT-free survival after standard
IST in patients ≤40 years and >40 years.
Methods: The Dutch Adult Aplastic Anemia Registry started in 2014 and contains detailed
data of all consecutive AA patients who have been treated with ATGAM-based therapy
in the participating hospitals, offering a unique possibility to evaluate real-life
long-term efficacy and safety of 1st line IST treatment in AA. To determine TTT-free
survival, a multistate model (MSM) was developed, to take into account that patients
can have transient periods of treatment success and failure. Patients started in the
dynamic state treatment & transfusion at time of the start of ATGAM. This state also
included all other (2nd line) non-alloSCT treatments (for example Eltrombopag, Rabbit-ATG
or Danazol). Other dynamic states patients could enter and leave during follow-up
were treatment & no transfusion, transfusion & no treatment and no treatment & no
transfusion (considered as TTT-free survival). Absorbing states, accessible from all
dynamic states, were death, alloSCT for AA, treatment for MDS/AML and treatment for
PNH.
Results: ATGAM with CsA as 1st line treatment was started in 117 patients (median
age 54 years, range 18-79). Overall survival after 5 year is 77% (95% Confidence Interval
(95%CI) 67-86%) Figure 1 shows the results of the MSM. After 5 years, the TTT-free
survival was 42% (95%CI: 33-55%) for the total cohort. 19% (95%CI: 12-31%) of the
patients was transfusion independent but still needed medication at this time (mainly
CsA or Eltrombopag). 15% (95%CI: 10-23%) had received an alloSCT as 2nd line treatment
for AA, 5% (95%CI: 2-13%) had started treatment for MDS/AML and 2% (95%CI: 0-11%)
had started treatment with complement inhibition for PNH. 5-year TTT-free survival
was 60% (95%CI:44-82%) in patients aged ≤40 years (n=36), but only 33% (95%CI: 23-49%)
in patients aged >40 years (n=81).
Image:
Summary/Conclusion: TTT-free survival can be used to evaluate treatment success after
IST in AA patients, allowing achievement, loss and recovery of response. We showed
that 5-year TTT-free survival is superior in patients ≤40 years compared to patients
>40 years. This MSM can be used to predict outcomes in AA patients receiving IST and
can help in the decision whether and when to offer a patient an alloSCT.
P820: PREDICTING RESPONSE TO RABBIT ATG-BASED INTENSIVE IMMUNOSUPPRESSIVE THERAPY
COMBINED WITH ELTROMBOPAG IN CHINESE ADULT PATIENTS WITH SEVERE APLASTIC ANEMIA
R. Li1, Z. Liu1, X. Chen2, Q. Long2, Y. Yang3, S. Lin4, J. Jia5, G. He1,*, J. Li1
1Hematology, the First Affiliated Hospital of Nanjing Medical University; 2Hematology,
the Second People’s Hospital of Nanjing, Nanjing; 3Hematology, the First Bethune Hospital
of Jilin University, Changchun; 4Hematology, Zhejiang Province Hospital of Traditional
Chinese Medicine, Hangzhou; 5Hematology, Peking University People’s Hospital, Beijing,
China
Background: The intensive immunosuppressive therapy (IST) consisted by antithymocyte
immunoglobulin (ATG) and cyclosporin (CsA) combining with eltrombopag (E-PAG) is the
initial therapy for adult patients with severe aplastic anemia (SAA). There is rabbit
ATG (r-ATG) rather than horse ATG in the mainland of China and E-PAG is recommended
with 75mg/d for East Asian population.
Aims: To explore factors predicting the efficacy of E-PAG (75mg/d) plus r-ATG based
IST for SAA patients in East Asia.
Methods: From February 2018 to December 2020, we conducted a retrospective, multicenter
study to analyze 58 adult SAA patients with rabbit ATG-based IST and E-PAG in the
China Eastern Cooperation Group for Anemia (CECGA). The dosage of r-ATG was 3.5mg·kg-1·d-1
for 5 days with intravenous infusion and CsA was 3-5mg·kg-1·d-1 early in the regimen
maintaining concentration at 150-200ng/ml, whose dosage could be modulated on the
basis of drug concentration and severe adverse events (SAEs). E-PAG was recommended
with 75mg/d at least 6 months. If or SAEs occurred or the count of platelet was higher
than 200×109/L, E-PAG would be discontinued.
Results: 58 patients with median age of 42.5 years (18 years to 74 years) were treated
with r-ATG based IST plus E-PAG. Cohort 1 and cohort 2 included patients with or without
a response at 6 months, whist the median age were 50.5 years (24 years to 74 years)
and 38 years (18 years to 71 years), separately. The median time between diagnosis
and treatment was 3 weeks (1 week to 106 weeks) and 3 weeks (1 week to 166 weeks),
respectively. The response rates of E-PAG combining with IST at 3, 6 and 12 months
after IST were 64%, 76% and 85% (44 of 52), while the complete response rates at 3,
6 and 12 months were 19%, 21% and 29% (15 of 52), separately. The baseline absolute
lymphocyte count (ALC) (area under a curve (AUC)=0.706, 95%CI 0.522-0.890, P=0.057),
red cell distribution width – coefficient of variation (RDW-CV) (AUC=0.722, 95%CI
0.494-0.950, P=0.040) and reticulocyte (Ret) percentage (AUC=0.798, 95%CI 0.640-0.956,
P=0.006) were highly predictive of response at 6 months (Figure 1). The tipping values
of Ret percentage, ALC and RDW-CV were 0.45%, 1.06×109/L and 11.75%, respectively.
The sensitivity and specificity of Ret percentage were 81.6% and 66.7%; RDW-CV were
94.7% and 55.6%; ALC were 55.3% and 88.9%. The 2-year overall survival was 93%.
Image:
Summary/Conclusion: The baseline Ret percentage, RDW-CV and ALC were potential factors
to predict a favorable effect in SAA patients of East Aisan.
P821: LATE ONSET AND/OR LONG LASTING NEUTROPENIA IN CHILDHOOD: CLINICAL AND HEMATOLOGICAL
CHARACTERISTICS AND OUTCOMES
C. Kelaidi1,*, K. Antoniadi1, V. Tzotzola1, V. Papadakis1, M. Ampatzidou1, C. Pontikoglou2,
I. Mavroudi2, M. Tzanoudaki3, G. Paterakis4, S. I. Papadhimitriou5, K. Tsitsikas1,
A. Bountali1, K. Manola6, H. A. Papadaki2, S. Polychronopoulou1
1Pediatric Hematology-Oncology, Aghia Sophia Children’s Hospital, Athens; 2Haemopoiesis
Research Laboratory, University of Crete, School of Medicine, Heraklion; 3Flow cytometric
cell analysis, Laboratory of Immunology, Aghia Sophia Children’s Hospital; 4Flow Cytometry
Analysis, Laboratory of Immunology; 5Laboratory of Hematology, Georgios Gennimatas
General Hospital; 6Laboratory of Health Physics, Radiobiology & Cytogenetics, National
Centre for Research Demokritos, Athens, Greece
Background: In contrast with primary autoimmune neutropenia of infants and pre-school
children, characterized by the presence of anti-polymorphonuclear antibodies (anti-PNN),
benign course and spontaneous resolution at age 3-5 years, late onset (LO) and/or
long lasting (LL) neutropenia of childhood is vaguely described.
Aims: To address characteristics and outcomes in a cohort of children with LO/LL diagnosed
and followed by our Department.
Methods: Cross-sectional study of patients aged 0-16 years with LO (age >5 years,
in accordance with Fioredda et al, 2020) or LL (duration >3 years) neutropenia (1.5
G/L) +/- leukopenia (Z-score ≤2 according to age and gender); with a first visit or
continued follow-up in our Department of Pediatric Hematology-Oncology (T.A.O.) during
the period 2015-2020. Patients with severe congenital neutropenia (SCN) were excluded.
Detection of anti-PNN using GAT and GIFT was centralized.
Results: From 1972 to 2021, 534 cases of neutropenia/leukopenia were recorded (5,
40, 82, 134, 273 during 1970-1979, 1980-1989, 1990-1999, 2000-2009, 2010-2021, respectively);
152 were included in the cross-sectional study. Sixty-one (40%) had no LO/LL (age<5
years, resolution within≤3 years), 77 (51%0 had LO and 14 (9%) had LL. Male/female
ratio was 47% vs 52% vs 28% in no LO/LL vs LO vs LL, respectively (P=0.27), median
age 1 vs 11.1 vs 3.1 years (P<10-4), median neutrophil count 0.5 vs 1.2 vs 0.6 G/L
(P<10-4), and with concomitant leukopenia in 27% vs 53% vs 36% (P=0.009) or lymphopenia
in 3% vs 10% vs 7% of cases (P=0.27). Anti-PNN were detected in 83% vs 42% vs 60%
of patients no LO/LL vs LO vs LL, respectively (P=0.12). Familial neutropenia was
observed in three pairs of siblings/cousins with LL, possible ethnic neutropenia in
7 patients, probable genetic predisposition to cancer in 4 patients LO/LL and a congenital
metabolic syndrome in one patient LL. Infection, mostly cutaneous, occurred in 17%
vs 24% vs 66% no LO/LL vs. LO vs. LL (P=0.02). Bone marrow examination was performed
in 8 patients (4 LO, 4 LL) and showed mild hypoplasia in 2 (LO). Mild peripheral blood
lymphocytic subsets abnormalities were observed in ¼ of the patients, particularly
in LO, and mild reduction of serum immunoglobulin class levels in ¼ of the patients,
without difference between no LO/LL and LO. Genetic typing performed in 9% of patients
LO/LL (targeted pediatric MDS panel N=2, primary immunodeficiency N=2, SCN N=1, whole-exome
sequencing N=2) showed a heterozygous TACI mutation in one patient with LL and frequent
infections of moderate severity. In total, MDS, immunodeficiency and autoimmunity
was noted in 1%, 5% and 4% of patients LO and immunodeficiency in 14% of patients
LL. One patient LL developed B-cell precursor ALL. One patient LO with Evans syndrome
received eltrombopag, without response, and MMF with response. All patients are alive.
Neutropenia resolved spontaneously in 54% of patients. Two-year probability of unresolved
neutropenia was 26% vs 78% vs 100% in patients no LO/LL, LO and LL, respectively (P<10-4).
Summary/Conclusion: LO/LL neutropenia might be associated with bone marrow failure
syndromes, immunodeficiency or autoimmunity. Long-term follow-up and investigations
are needed in children with LO/LL neutropenia. Genetic typing in specialized reference
networks, like EuNet-INNOCHRON or EWOG, might identify diagnostic subgroups of clinical
relevance.
P822: A PHASE 1 SINGLE ASCENDING DOSE STUDY OF CAN106, A LONG-ACTING ANTI-C5 COMPLEMENT
MONOCLONAL ANTIBODY IN CLINICAL DEVELOPMENT FOR PNH AND OTHER COMPLEMENT-MEDIATED
DISEASES
C. M. KHOO1,*, X. SONG2, Q. WU2, G. F. COX3
1Department of Medicine, National University Hospital, Singapore, Singapore; 2Clinical
Development and Medical Affair, CANbridge Pharmaceuticals Inc., Shanghai, China; 3Clinical
Development and Medical Affair, CANbridge Pharmaceuticals Inc., Cambridge, MA, United
States of America
Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare, acquired genetic
disorder caused by clonal mutations in the PIGA gene. Deficiency of PIGA protein renders
erythrocytes susceptible to complement-mediated destruction, causing intravascular
and extravascular hemolysis, which can lead to anemia, fatigue, thromboembolism, major
organ damage, and death. Despite the approval of one anti-C3 and two anti-C5 therapies
for PNH, high treatment costs limit market access in many countries. CAN106 is a novel
anti-C5 monoclonal antibody designed for optimal pH-dependent binding to C5 and enhanced
binding to FcRN to increase intracellular recycling and prolong its half-life.
Aims: This first-in-human Phase 1 study aimed to evaluate the single-dose safety and
tolerability, pharmacokinetics, pharmacodynamics, and immunogenicity of CAN106.
Methods: This was a randomized, double-blind, placebo-controlled, single-ascending-dose
study. We enrolled 31 healthy adult subjects at a single site in Singapore who received
CAN106 (mg/kg) or placebo at one of the following 6 doses (mg/kg) and ratios: (0.25,
n=1:0; 0.75, n=1:0; 2, n=3:2; 4, n=6:2; 8, n=6:2 or 12, n=6:2). The primary endpoint
was the incidence of adverse events overall and by intensity, seriousness, type, and
relatedness to study drug. Secondary endpoints were the pharmacokinetic characterization
of CAN106, the pharmacodynamic effects on free C5 (target engagement) and CH50 (serum
hemolytic activity), and the incidence of anti-drug antibodies. The original study
duration of 196 days was shortened to 112 days based upon the observed half-life of
CAN106 and the return of CH50 to normal values.
Results: The mean age of subjects was 34 years, and 97% were male. All 31 subjects
completed the study. Seven CAN106 subjects experienced 20 treatment-emergent adverse
events (TEAEs), and three placebo subjects experienced five TEAEs. Most TEAEs were
mild and not related to CAN106, and all resolved without any sequelae. Three CAN106
subjects experienced seven drug-related TEAEs at the two highest doses. Most were
mild, none was serious, and all resolved without any sequelae. These events included
a moderate infusion-related reaction that resolved upon discontinuing treatment; mild
dizziness; and mild elevations in ALT, AST, hemoglobin, hematocrit, and RBC. CAN106
exposure (Cmax and AUC) was dose-proportional, linear, and had low inter-subject variability
(<20% CV). The terminal elimination half-life was approximately 32 days. CAN106 led
to dose-dependent reductions in free C5 and CH50 within 24 hours. At the 8 and 12 mg/kg
doses, all subjects showed >99% reduction in free C5 and >90% inhibition of CH50,
with the latter sustained for 2 to 4 weeks. Two subjects in the 4 mg/kg cohort tested
positive for anti-drug antibodies at single time points (pre-dose and Day 28).
Summary/Conclusion: CAN106 was safe and well-tolerated at single doses up to 12 mg/kg
in healthy adult subjects. The dose-exposure relationship was linear, and the 32-day
half-life in healthy subjects was similar to that of ravulizumab, which is dosed every
8 weeks in patients, suggesting the potential for an extended dosing interval. CAN106
led to rapid, dose-dependent reductions in free C5 and CH50, and at the two highest
doses, achieved complete and sustained complement blockade (>90% inhibition of CH50)
for up to 4 weeks. These promising results support further clinical development of
CAN106 in PNH and other complement-mediated diseases.
P823: THE INCIDENCE, OUTCOMES, AND RISK FACTORS OF SECONDARY POOR GRAFT FUNCTION IN
HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR ACQUIRED APLASTIC ANEMIA
F. lin1,*, T. han1, Y. zhang1, Y. cheng1, Z. xu1, X. mo1, F. wang1, C. yan1, Y. sun1,
J. wang1, F. tang1, W. han1, Y. chen1, Y. wang1, X. zhang1, K. liu1, X. huang1, L.
xu1
1Peking university institute of hematology, Beijing, China
Background: Secondary poor graft function (sPGF) increases the risk of life-threatening
complications after transplantation and is an obstacle to achieving better outcomes
after hematopoietic stem cell transplantation (HSCT).
Aims: We aimed to determine the incidence, clinical outcomes, and risk factors of
sPGF in haploidentical HSCT (haplo-HSCT) for acquired aplastic anemia (AA) patients.
Methods: We retrospectively reviewed 430 consecutive AA patients receiving haplo-HSCT
in Peking University People’s Hospital between January 2006 and December 2020.
Results: We reported the estimated 3-year cumulative incidence of sPGF was 4.64%,
while no primary PGF occurred. The median time to sPGF was 121 days (range 30-626
days) after transplantation. To clarify the risk factors for sPGF, 17 sPGF cases and
382 without PGF were further analyzed. Compared to patients without PGF, the 2-year
overall survival was significantly poorer for sPGF patients (67.7% vs 90.8%, p =.002).
Twelve sPGF patients were alive until the last follow-up, and 7 achieved transfusion
independency. The multivariable analyses revealed that a history of refractory cytomegalovirus
viremia (CMViremia) (OR=7.038, p=.002) post-transplantation independently increased
the risk of sPGF. There was weak evidence that a history of grade 3-4 acute graft-versus-host
disease (aGvHD) increased the risk of sPGF (p=.092).
Summary/Conclusion: In conclusion, sPGF can develop in 4.64% of AA patients after
haplo-HSCT and significantly decreases survival. The independent hazard elements for
sPGF were a history of refractory CMViremia and grade 3-4 aGvHD. We advocated better
post-transplantation strategies to balance the risk of immunosuppression and viral
reactivation for haplo-HSCT in AA patients.
P824: HEMATOLOGIC FEATURES IN 102 FRENCH PATIENTS DIAGNOSED WITH GERMLINE MUTATIONS
OF TELOMERES RELATED GENES AGED 15 YEARS OLD OR MORE
F. Maillet1,*, J.-E. Galimard2, R. Borie3, C. Kannengiesser4, E. Lainey5, R. Peffault
De Latour1, F. Sicre de Fontbrune1
1Hematology Department, Saint Louis Hospital; 2Statistical Unit, EBMT; 3Pulmonology
Department; 4Genetics Department, Bichat Hospital; 5Hematology Department, Robert
Debré Hospital, Paris, France
Background: Short telomere syndrome (STS) is a group of genetic disorders characterized
by germline mutations in telomeres related genes (TRG) leading to premature telomere
shortening. In the recent years, the spectrum of STS diseases has widened and now
includes heterogeneous associations of hematologic, pneumologic, hepatic and osteoarticular
features, and cancer predisposition. STS clinical landscape remain unknown. We took
advantage of a large cohort of patients to describe hematologic and extra-hematologic
features of this rare disease.
Aims: The aim of this study was to describe hematologic and extrahematologic features
in patients with STS diagnosed at age 15 or more and followed in adult centers.
Methods: All data were obtained from the French National reference Center for Aplastic
Anemia. Between 2003 and 2021, we retrospectively included all patients 15 years old
or older at diagnosis of STS, with pathogenic germline variant(s) in TRG, and at least
one bone marrow assessment (bone marrow aspiration or biopsy). Overall survival (OS)
was calculated from date of diagnosis to death or last follow-up using the Kaplan-Meier
estimator.
Results: One hundred and two patients were included: 89 (87%) were propositus and
13 were diagnosed after genetic counseling. The median follow-up after diagnosis was
3.6 years. The 4 years OS of the index cases was 63% (95% CI: 49 – 74).
62 (70%) and 6 (46%) patients were male, and median age at diagnosis was 42 years
(IQR 28-56) and 27 years (IQR 21-48) for index cases and non-index cases, respectively.
Median age at onset of first symptoms was 28 years (IQR 20-49) and 29 years (IQR 18-38),
with a median duration from first symptoms to STS diagnosis of 5 years (IQR 2-11)
and 6 years (IQR 0-7) for index cases and non-index cases, respectively. In index
cases, TRG mutations involved TERT in 43 patients (49%), TERC in 24 patients (28%),
RTEL1 in 11 patients (13%), DKC1 in 3 patients (3%), and in 1 patient each for NHP2,
CTC1, PARN, TINF2, TERT plus TERC (0.9%). In index cases, mutations were heterozygous
in 92%, homozygous in 6%, and X-linked in 2%. Among 68 patients with available telomere
evaluation, the median telomere length was 6.2% (IQR 4.8-7.4%), whereas the median
telomere length at first percentile was 7.6%. Telomere length was shorter than first
percentile in 81.5% of patients. 69% of index cases had at least 1 relative with STS.
Among index cases, 73 (82%) presented an hematological involvement. At diagnosis,
the hematological presentation was BMF in 49 patients (70%), MDS in 17 (24%) and AML
in 2 (3%), 1 CVID (1.5%), 1 lymphoma (1.5%) and not specified for 3 patients. During
follow-up, evolution toward MDS was observed in 2 patients, and toward AML in 4. 14
patients received allo-HSCT (13.7%): 9 for BMF and 5 for MDS.
Extra hematologic features among index cases: 49 patients (55%) had at least one clinical
feature of dyskeratosis congenita. Pulmonary involvement was present in 55 patients
(62%), of those 93% had diffuse interstitial pneumonia. 12 patients underwent lung
transplantation. Hepatic involvement was present in 42 patients (47%), including nodular
regenerative hyperplasia (29%), cirrhosis (33%) and portal hypertension (43%). 3 patients
underwent liver transplantation. Cancer occurred in 7 patients: 4 solid cancers and
3 lymphomas.
Summary/Conclusion: This study in STS patients allows a better understanding of the
spectrum of this disease. Hematological presentation was the main feature, followed
by lung and hepatic involvement. A long-term multidisciplinary close follow-up is
mandatory in this population of patients.
P825: FACTOR D INHIBITION WITH ORAL BCX9930 MONOTHERAPY LEADS TO SUSTAINED CONTROL
OF HEMOLYSIS AND SYMPTOMS OVER 48 WEEKS IN SUBJECTS WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA
NAÏVE TO C5 INHIBITORS
A. McDonald1,*, J. L. R. Malherbe2, A. Kulasekararaj3, W. Füreder4, M. Griffin5, M.
Cornpropst6, M. K. Farmer6, D. Kargl6, D. Collins6, W. Sheridan6, A. Risitano7
1Netcare Pretoria East Hospital, Moreleta Park; 2University of the Free State, Bloemfontein,
South Africa; 3Kings College Hospital NHS Foundation Trust, London, United Kingdom;
4Medical University of Vienna, Vienna, Austria; 5Leeds Teaching Hospitals NHS Trust,
Leeds, United Kingdom; 6BioCryst Pharmaceuticals, Durham, United States of America;
7AORN Moscati, Avellino, Italy
Background: Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by hemolysis
due to uncontrolled activity of the alternative pathway (AP) of complement. BCX9930
is an oral Factor D inhibitor that targets the rate-limiting enzyme of AP with the
potential to prevent both intravascular hemolysis and extravascular hemolysis. We
previously reported safety and effectiveness of BCX9930 at 22 weeks in subjects naïve
to complement inhibitors (CI; McDonald et al. EHA 2021). Here, we report further long-term
safety, tolerability, and effectiveness of BCX9930.
Aims: This analysis evaluated effectiveness of BCX9930 in subjects completing at least
48 weeks of treatment at target dose of 400 mg BID or 500 mg BID. Additionally, safety
data are presented for all subjects (n=16, including 6 subjects with an inadequate
response to C5 inhibitors) enrolled in an open-label, dose-ranging study (NCT04330534)
and extension study (NCT04702568).
Methods: Study BCX9930-101 enrolled subjects with PNH with either no history of C5
inhibitor use or those with an inadequate response to C5 inhibitors (transfusion within
3 months of screening or Hb <10 g/dL). Subjects in each of three multiple dose cohorts
received BCX9930 at 50 to 400 mg orally twice daily (BID) titrated to target dose
of 400 or 500 mg BID. CI-naïve subjects deriving clinical benefit after 28 days of
treatment could receive up to 48 additional weeks of BCX9930 before continuing into
an extension study.
Safety and effectiveness outcomes were evaluated monthly. Effectiveness endpoints
were change from baseline (CFB) to 48 weeks in hemoglobin (Hb), red blood cell (RBC)
transfusions, lactate dehydrogenase (LDH), absolute reticulocyte count (ARC), and
% PNH Type II+III RBC clone size relative to PNH white blood cell (WBC) clone size.
CFB in fatigue was assessed by Functional Assessment of Chronic Illness Therapy (FACIT)
Fatigue Scale (clinically important difference ≥3-point change, higher scores indicate
less fatigue).
Results: Ten CI-naïve adult subjects enrolled in BCX9930-101 and 9 completed at least
48 weeks of treatment. Mean (SD) age and time since PNH diagnosis were 29.2 (8.09)
years and 2.6 (1.70), mean (SD) number of RBC units transfused in 12 months prior
to study entry was 7.6 (8.2), and median (range) treatment exposure at 400 mg or 500 mg
BID was 470 (350, 533) days. Mean (SEM) Hb CFB to Week 48 was 3.75 (0.89) g/dL (8.25
[0.60] g/dL to 12.0 [0.85] g/dL), and all 9 subjects were transfusion-free versus
2 of 9 in 12 months prior to study. At Week 48, mean (SEM) ARC CFB was – 57 (21.8)
103/µL (178 [20.7] vs. 121 [15.5] 103/µL), mean (SEM) % PNH RBC/WBC clone size increased
from 51.9% (4.4%) to 93% (2.7%), and mean (SEM) LDH CFB was –70.5% (6.18%). Clinically
meaningful improvements in FACIT-Fatigue scores were observed over 48 weeks, with
mean (SEM) CFB of 5.3 (1.82) points (mean score 40.7 vs 46.0). The most common adverse
events in all subjects (n=16) were headache (38%), self-limited rash (25%), and abdominal
pain (19%).
Summary/Conclusion: BCX9930 at doses up to 500 mg BID was generally well-tolerated.
Notable improvements from baseline to 48 weeks were observed in Hb, LDH, ARC, and
fatigue scores, with concomitant increases in PNH RBC clone size. Proximal AP inhibition
of Factor D by BCX9930 demonstrated sustained relief of anemia and fatigue, and freedom
from transfusions, supporting ongoing pivotal trials of BCX9930 monotherapy for treatment
of PNH.
P826: FACTOR D INHIBITION WITH ORAL BCX9930 LEADS TO SUSTAINED CONTROL OF HEMOLYSIS
AND SYMPTOMS OVER 48 WEEKS IN SUBJECTS WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA INADEQUATELY
CONTROLLED ON C5 INHIBITORS
A. Kulasekararaj1,*, W. Füreder2, A. McDonald3, J. L. R. Malherbe4, S. Gandhi1, D.
Collins5, M. Cornpropst5, S. Dobo5, M. K. Farmer5, D. Kargl5, W. Sheridan5, M. Griffin6,
A. Risitano7
1Kings College Hospital NHS Foundation Trust, London, United Kingdom; 2Medical University
of Vienna, Vienna, Austria; 3Netcare Pretoria East Hospital, Moreleta Park; 4University
of the Free State, Bloemfontein, South Africa; 5BioCryst Pharmaceuticals, Durham,
United States of America; 6Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom;
7AORN Moscati, Avellino, Italy
Background: Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by hemolysis
due to uncontrolled activity of the alternative pathway (AP) of complement. BCX9930
is an oral Factor D inhibitor that targets the AP rate-limiting enzyme with the potential
to prevent both intravascular hemolysis and extravascular hemolysis. We previously
presented safety and effectiveness data for BCX9930 at 11 weeks in subjects with an
inadequate response (IR) to C5 inhibitors (C5 INH; Kulasekararaj et al. EHA 2021).
Here, we report further long-term safety, tolerability, and effectiveness of BCX9930.
Aims: This analysis evaluated effectiveness of BCX9930 in subjects completing at least
48 weeks of treatment at target dose of 400 mg BID or 500 mg BID. Additionally, safety
data are presented for all subjects enrolled (n=16, including 10 subjects naïve to
C5 INH) in an open-label, dose-ranging study (NCT04330534) and extension study (NCT04702568).
Methods: Study BCX9930-101 enrolled subjects with PNH with either no history of C5
INH use or those with an IR to C5 INH (defined as having a transfusion within 3 months
of screening or Hb <10 g/dL). For subjects with an IR to C5 INH treatment, BCX9930
was added to C5 INH and subjects were treated with 200 mg orally twice daily (BID)
escalated to 400 mg BID, or 400 mg BID escalated to 500 mg BID. Subjects deriving
clinical benefit could rollover into an extension study after 28 days, and C5 INH
could be withdrawn.
Safety and effectiveness outcomes were evaluated monthly. Effectiveness endpoints
were change from baseline (CFB) to 48 weeks in hemoglobin (Hb), red blood cell (RBC)
transfusions, lactate dehydrogenase (LDH), absolute reticulocyte count (ARC), and
% PNH RBCs relative to PNH white blood cells (WBC). CFB in fatigue was assessed by
Functional Assessment of Chronic Illness Therapy (FACIT) Fatigue Scale (clinically
important difference ≥3-point change, higher scores indicate less fatigue).
Results: Six adult subjects with an IR to C5 INH enrolled in Study BCX9930-101 and
rolled over into the open-label extension study, and 4 completed 48 weeks of treatment
(3 of 4 on BCX9930 monotherapy at Week 48, mean [range] duration on monotherapy 107
[85, 129] days). Mean (SD) age and time since PNH diagnosis were 31 (9.42) and 7.7
(5.80) years, mean (SD) number of RBC units transfused in 12 months prior to study
entry was 5.5 (4.4), and median (range) treatment exposure at 400 mg or 500 mg BID
was 413.5 (337, 420) days. Mean (SEM) Hb CFB to Week 48 was 2.69 (0.97) g/dL (8.91
[0.56] g/dL to 11.60 [1.27] g/dL). 3 of 4 subjects were transfusion-free at Week 48
versus 1 of 4 in 12 months prior to study. At Week 48, Mean (SEM) ARC CFB was –63
(17.1) 103/µL (204 [15.5] vs. 141 [19.6] 103/µL), and mean (SEM) % PNH RBC/WBC clone
size increased from 59% (4.4%) to 92% (3.8%,). Clinically meaningful improvements
in FACIT-Fatigue scores were observed over 48 weeks, with mean (SEM) CFB of 10.7 (6.0)
points (mean score 37.3 vs. 47.9). The most common adverse events for all subjects
(n=16) were headache (38%), self-limited rash (25%), and abdominal pain (19%).
Summary/Conclusion: BCX9930 at doses up to 500 mg BID was generally well-tolerated.
Notable improvements were observed from baseline to 48 weeks in Hb, ARC, and fatigue
scores, with concomitant increases in PNH RBC clone size. Proximal AP inhibition of
Factor D by BCX9930 in combination with C5 INH (with 3 of 4 subjects transitioning
to monotherapy) led to sustained relief of anemia, fatigue, and reduction of transfusion
burden, supporting ongoing pivotal trials of oral BCX9930 monotherapy for treatment
of PNH.
P827: EFFICACY AND SAFETY OF ROMIPLOSTIM ADDED TO IMMUNOSUPPRESSIVE THERAPY AS A FIRST-LINE
TREATMENT IN PATIENTS WITH APLASTIC ANEMIA: A PHASE 2/3 CLINICAL TRIAL
H. Yamazaki1,*, J. W. Lee2, J. H. Jang3, M. Sawa4, M. Kizaki5, Y. Tomiyama6, K. Nagafuji7,
K. Usuki8, J.-P. Gau9, Y. Morita10, J.-L. Tang11, H. Chang12, M. Noshiro13, A. Matsuda14,
K. Ozawa15, K. Mitani16, Y. Kanda15, S. Nakao1
1Kanazawa University Hospital, Ishikawa, Japan; 2Seoul St. Mary’s Hospital; 3Samsung
Medical Center, Seoul, South Korea; 4Anjo Kosei Hospital, Aichi; 5Saitama Medical
Center, Saitama; 6Osaka University Hospital, Osaka; 7Kurume University Hopital, Fukuoka;
8NTT Medical Center Tokyo, Tokyo, Japan; 9Taipei Veterans General Hospital, Taipei,
Taiwan; 10Kindai University Hospital, Osaka, Japan; 11National Taiwan University Hospital,
Taipei; 12Chang Gung Medical Foundation- Linkou Branch, Taoyuan, Taiwan; 13Kyowa
Kirin Co.,Ltd., Tokyo; 14Saitama Medical University International Medical Center,
Saitama; 15Jichi Medical University Hospital; 16Dokkyo Medical University Hospital,
Tochigi, Japan
Background: Romiplostim, a thrombopoietin (TPO) mimetic protein, has been shown to
promote tri-lineage hematopoiesis in patients with acquired aplastic anemia (AA) refractory
to immunosuppressive therapy (IST) or eltrombopag; however, its effectiveness in the
combination therapy with antithymocyte globulin (ATG) plus cyclosporine (CsA) as a
first-line treatment remains unknown.
Aims: To clarify this issue, we conducted a phase 2/3 clinical trial to evaluate the
efficacy and safety of romiplostim combined with rabbit ATG plus CsA in patients with
AA requiring transfusions who had not been exposed to IST.
Methods: This study was a multi-national, open-label study with romiplostim in IST-naïve
adult patients with AA (NCT03957694). 10 µg/kg of romiplostim was started on day 1
of ATG therapy (2.5mg/kg/day for 5 days) and was weekly given for the first 4 weeks.
The dose was adjusted from 5 to 20 µg/kg according to the prespecified dose adjustment
procedure. Treatment lasted until Week 26. For those who intend to continue treatment,
romiplostim administration was extended to Week 52. The primary endpoint was the hematological
response rate (HRR) at Week 27 based on the response assessment criteria (Table 1).
The secondary endpoints included the HRR at Week 14; the time to the hematological
response; the proportion of patients who achieved transfusion independence or who
showed a reduction of transfusion requirement among patients who had received a transfusion
within 8 weeks prior to the first romiplostim administration. The bone marrow and
cytogenetic analyses were performed prior to enrollment and at Week 27 and 53.
Results: Twenty-six patients were screened, of which 17 (9 Japan, 7 Korea, and 1 Taiwan;
5 transfusion dependent non-severe AA, 6 severe AA (SAA) and 6 very SAA) were enrolled
in the study. The median age was 44.0 years (range: 25-70). Two patients discontinued
romiplostim before Week 27. The HRR (CR or PR) at Week 27 was 76.5% (95% CI: 50.10%,
93.13%), of which 6 (35.3%) achieved CR. The HRR at Week 14 was 41.2% (95% CI: 18.44%,
67.03%). The median day to achieve the first hematological response was 93.0 (range:
63-181). Of the 16 patients who depended on platelet transfusion before romiplostim
administration, 14 (87.5%) achieved transfusion independence or showed a reduction
of transfusion requirement at Week 27. In addition, of 16 who required erythrocyte
transfusion at baseline, 13 (81.3%) achieved transfusion independence or showed a
reduction of transfusion requirement at Week 27.
The frequently reported adverse events (AEs) were constipation (41.2%) followed by
headache (35.6%). The frequently reported drug-related AEs were headache (12.9%) followed
by muscle spasms (9.7%), alanine aminotransferase increased, fibrin D dimer increased,
malaise, and pain in extremity (each 6.5%). In bone marrow examination, increases
of reticuline grade (grade 1 to 2) were observed in 3 patients at Week 27 and 1 patient
at Week 53. No chromosomal abnormality or transformation into MDS and/or AML was observed.
Image:
Summary/Conclusion: This is the first clinical trial of romiplostim combined with
rabbit ATG plus CsA in patients with acquired AA requiring transfusions who were previously
untreated with IST. This regimen produced higher HRR at Week 27 (76.5%) than those
of historical control received rabbit ATG plus CsA (approximately 50% at 6 months),
with manageable AEs, and could therefore serve as a new first-line treatment option
in patients with AA.
P828: NORMALIZATION OF HEMATOLOGIC AND HEALTH-RELATED QUALITY OF LIFE MARKERS IN PATIENTS
WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA TREATED WITH PEGCETACOPLAN AND BASELINE HEMOGLOBIN
AT OR ABOVE 10 G/DL
J. Panse1,*, N. Daguindau2, S. Okuyama3, R. Peffault de Latour4, P. Schafhausen5,
N. Straetmans6, M. Al-Adhami7, T. Ajayi7, E. Persson8, M. Yeh7, R. Wong9
1Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation,
University Hospital RWTH Aachen, Aachen, Germany; 2Hematology Department, CH Annecy
Genevois, Annecy, France; 3Hematology/Oncology Division, Denver Health Medical Center,
Denver, United States of America; 4French Reference Center for Aplastic Anemia and
Paroxysmal Noctural Hemoglobinuria, Assistance Publique - Hôpitaux de Paris; Université
de Paris, Paris, France; 5Medical Clinic and Polyclinic II - Hematology, Oncology,
University Medical Center of Hamburg-Eppendorf, Hamburg, Germany; 6Department of Hematology,
Cliniques Universitaires Saint-Luc, Woluwe-Saint-Lambert, Belgium; 7Apellis Pharmaceuticals,
Inc., Waltham, United States of America; 8Swedish Orphan Biovitrum AB, Stockholm,
Sweden; 9Sir Y.K. Pao Centre for Cancer & Department of Medicine and Therapeutics,
Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong
Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare life-threatening disease
characterized by chronic complement-mediated hemolysis and thrombosis. Patients living
with PNH can experience anemia with associated fatigue, dyspnea, and require blood
cell transfusions which can negatively impact quality of life (QoL). Pegcetacoplan
(PEG), is a C3 complement-inhibitor approved by the FDA/EMA for adults with PNH.
Aims: This post hoc analysis evaluated hemoglobin (Hb), lactate dehydrogenase (LDH),
absolute reticulocyte count (ARC), and Functional Assessment of Chronic Illness Therapy
(FACIT)-Fatigue normalization rates and safety after PEG treatment in a subgroup of
patients with PNH and baseline Hb levels ≥10.0g/dL from the PADDOCK (NCT02588833),
PEGASUS (NCT03500549), and PRINCE (NCT04085601) studies.
Methods: PADDOCK evaluated PEG treatment (270-360mg/day subcutaneously [sc]) in complement-inhibitor
naïve patients. PEGASUS enrolled patients with Hb levels <10.5g/dL at screening despite
stable eculizumab (ECU) treatment (≥3 months). PEGASUS patients were randomized 1:1
to ECU or PEG (1080mg sc 2x weekly) during the randomized controlled period (RCP)
through Week 16. Patients who received PEG during the RCP continued with PEG monotherapy
(PEG-to-PEG) and ECU patients switched to PEG monotherapy (ECU-to-PEG) during the
open-label period (OLP) through Week 48. PRINCE compared PEG treatment (1080mg sc
2x weekly) in complement-inhibitor naïve patients with PNH and Hb levels below the
lower limits of normal (males: ≤13.6g/dL; females: ≤12.0g/dL) to control treatment
(CTRL; excluding complement-inhibitors i.e., ECU/ravulizumab). The post hoc analysis
included adult patients with PNH with baseline Hb levels ≥10.0g/dL and no transfusions
within 14 days of the baseline measurement. Hb, LDH, and ARC normalization, as well
as FACIT-Fatigue normalization (defined as increase to population norm) was evaluated
after 16 weeks of PEG monotherapy for all three studies. Patients who withdrew or
were lost to follow-up without providing efficacy data at the specified timepoints
were classified as non-responders. Safety endpoints included incidences of adverse
events (AEs).
Results: Overall, 22 patients were included in the post hoc analysis: 5 PADDOCK, 9
PEGASUS, and 8 PRINCE patients. After 16 weeks of pegcetacoplan treatment, patients
in all three studies showed improvements in Hb normalization, LDH normalization, and
ARC normalization (Table 1). Additionally, improvements in FACIT-Fatigue normalization
were demonstrated in PADDOCK, PRINCE, and PEGASUS (PEG-to-PEG) patients (Table 1).
No incidences of serious AEs were reported in PADDOCK and PRINCE subgroups. One PEGASUS
patient had a serious AE of breakthrough hemolysis (severe, unrelated to PEG), following
an upper respiratory infection which resolved and did not lead to study discontinuation.
Incidences of injection site reactions and infections and infestations occurred at
similar rates between subgroups in all three studies (Table 1). No thrombotic events
occurred in this patient population.
Image:
Summary/Conclusion: While this patient population had near normal Hb at baseline (median:
10.4g/dL), they also had elevated ARC and LDH prior to PEG therapy, suggesting ongoing
hemolysis which was normalized in most patients after PEG treatment initiation. Overall,
these results suggest PEG can be efficacious long-term in patients with less severe
anemia, regardless of prior complement-inhibitor treatment, resulting in further clinical
improvements in markers of hemolysis and QoL. The safety profile of PEG was consistent
with results from previous studies.
P829: A PHASE III RANDOMIZED CLINICAL TRIAL COMPARING SB12 (PROPOSED ECULIZUMAB BIOSIMILAR)
WITH REFERENCE ECULIZUMAB IN PATIENTS WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA
J. H. Jang1,*, R. Demichelis Gomez2, H. Bumbea3, L. Nogaieva4, L. L. L. Wong5, S.
M. Lim6, J. Park7, Y. Kim8, S. Cho7
1Hematology-Oncology, Samsung Medical Center, Seoul, South Korea; 2Hematology, Instituto
Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico; 3Hematology,
Bucharest Emergency University Hospital, Bucharest, Romania; 4Regional Treatment-Diagnostic
Hematology Center, Communal Nonprofit Enterprise Cherkasy Regional Oncology Dispensary
of Cherkasy Oblast Council, Cherkasy, Ukraine; 5Hematology Unit, Hospital Queen Elizabeth,
Kota Kinabalu; 6Department of Medicine, Hospital Sultanah Aminah, Johor Bahru, Malaysia;
7Clinical Development; 8Biometrics, Samsung Bioepis, Incheon, South Korea
Background: SB12, a proposed eculizumab biosimilar, is a humanized monoclonal antibody
that blocks complement C5 cleavage, thereby inhibits terminal complement-mediated
intravascular hemolysis.
Aims: The randomized, double-blind, multicenter, cross-over study was aimed to demonstrate
comparable clinical efficacy by evaluating the lactate dehydrogenase (LDH), safety,
pharmacokinetics (PK), pharmacodynamics (PD), and immunogenicity of SB12 and reference
eculizumab (ECU) in paroxysmal nocturnal hemoglobinuria (PNH) patients (NCT04058158).
Methods: A total of 50 patients aged ≥ 18 years with a confirmed diagnosis of PNH
and ≥ 1.5 upper limit of normal range (ULN) of LDH without previous exposure to a
complement inhibitor were included. All patients provided written informed consents
and were randomized (1:1) to treatment sequence I (TS1: SB12 to ECU, n=25) or II (TS2:
ECU to SB12, n=25). Patient received 600 mg of SB12 (TS1) or ECU (TS2) intravenously
every week for first 4 weeks (initial phase) and 900 mg for the fifth week, followed
by 900 mg every 2 weeks thereafter (maintenance phase). The treatment was switched
to ECU (TS1) or SB12 (TS2) at Week 26, and switched treatment was provided until Week
50.
Primary endpoints were LDH level at Week 26 and time-adjusted area of under the effect
curve (AUEC) of LDH from Week 14 to Week 26 and Week 40 to Week 52. Equivalence was
declared for LDH level at Week 26 if the two-sided 95% confidence interval (CI) of
the mean difference in between SB12 and ECU lied within the pre-defined equivalence
margin of [−1.2 × ULN, 1.2 × ULN] = [−337.2, 337.2], where ULN = 281 U/L. Equivalence
was declared for time-adjusted AUEC of LDH if the two-sided 90% CI of the ratio of
geometric means between SB12 and ECU lied within the pre-defined equivalence margin
of [0.77, 1.29]. Secondary endpoints were LDH profile over time and number of units
of packed red blood cells (pRBCs) transfused throughout the study period, safety,
PK, PD, and immunogenicity.
Results: Of the 50 randomized patients, 49 patients received the study treatment and
46 patients completed the study. Baseline demographic and disease characteristic were
comparable between the two treatment sequences. The 95% CI of mean difference in LDH
level at Week 26 between SB12 and ECU (SB12 − ECU: 34.48, 95% CI [−47.66, 116.62])
lied within the pre-defined equivalence margin. The 90% CI of ratio of time-adjusted
AUEC of LDH between SB12 and ECU (SB12/ECU: 1.08, 90% CI [0.95, 1.23]) lied within
the pre-defined equivalence margin. The overall LDH profile during the study period
was also comparable. The mean number of units of pRBCs transfused during Period 1
(TS1: 1.1 U; TS2: 0.9 U, respectively) and Period 2 (TS1: 1.1 U; TS2: 1.0 U, respectively)
was comparable.
Treatment-emergent adverse events (TEAEs) was reported in 72.3% for SB12 and 68.1%
in ECU. Three patients in SB12 and 2 patients in ECU had serious TEAEs. None of three
serious TEAEs reported in SB12 were related to the treatment. Two of three TEAEs (cellulitis;
infusion site hypersensitivity) reported in ECU were treatment-related, and 1 death
due to major adverse vascular event (portal vein thrombosis) not related to the treatment
was reported in ECU. The mean eculizumab serum trough concentrations and the mean
terminal complement activities in both treatment sequences were comparable. No patient
developed anti-drug antibodies during the study period.
Image:
Summary/Conclusion: SB12 showed clinical equivalence to ECU measured by LDH. SB12
and ECU were comparable in terms of safety, PK, PD, and immunogenicity.
P830: CROVALIMAB MAINTAINS CLINICAL BENEFIT OVER LONG-TERM TREATMENT IN PATIENTS WITH
PAROXYSMAL NOCTURNAL HEMOGLOBINURIA: RESULTS FROM THE COMPOSER TRIAL OPEN-LABEL EXTENSION
A. Röth1,*, M. Egyed2, S. Ichikawa3, Y. Ito4, J. S. Kim5, Z. Nagy6, N. Obara7, J.
Panse8, H. Schrezenmeier9, S. Sica10, J. Soret11, K. Usuki12, S.-S. Yoon13, K. Benkali14,
M. Buri15, P. Lundberg15, H. Patel16, K. Shinomiya17, S. Sreckovic15, J.-I. Nishimura18
1Department of Hematology and Stem Cell Transplantation, West German Cancer Centre,
University Hospital Essen, University of Duisburg-Essen, Essen, Germany; 2Kaposi Mor
Oktato Korhaz, Kaposvar, Hungary; 3Tohoku University Hospital, Miyagi; 4First Department
of Internal Medicine, Hematology Division, Tokyo Medical University, Tokyo, Japan;
5Yonsei University College of Medicine, Severance Hospital, Seoul, South Korea; 6Semmelweis
University, Budapest, Hungary; 7Department of Hematology, Faculty of Medicine, University
of Tsukuba, Tsukuba, Japan; 8University Hospital RWTH Aachen, Aachen; 9Institute of
Transfusion Medicine, University Hospital Ulm, Ulm, Germany; 10Fondazione Policlinico
Universitario A. Gemelli-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS),
Rome, Italy; 11Centre d’Investigation Clinique, Hôpital Saint-Louis, Paris, France;
12NTT Medical Center Tokyo, Tokyo, Japan; 13Department of Internal Medicine, Seoul
National University Hospital, Seoul, South Korea; 14Certara, Inc., Paris, France;
15F. Hoffmann-La Roche, Ltd., Basel, Switzerland; 16Genentech, Inc., South San Francisco,
CA, United States of America; 17Chugai Pharmaceutical Co., Tokyo; 18Graduate School
of Medicine, Osaka University, Osaka, Japan
Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a potentially life-threatening
acquired hematopoietic stem-cell disorder, resulting in hematuria, anemia, and acquired
thrombophilia. In the Phase I/II COMPOSER trial (NCT03157635), patients (pts) with
PNH who were treatment-naive or switched from eculizumab were treated with crovalimab,
a novel anti-C5 monoclonal antibody designed with Sequential Monoclonal Antibody Recycling
Technology (Röth et al. Blood 2020).
Aims: To report the long-term efficacy, safety, pharmacokinetic (PK) and pharmacodynamic
(PD) outcomes in pts with PNH treated with crovalimab in the open-label extension
(OLE) period of the COMPOSER trial.
Methods: COMPOSER consists of four sequential parts followed by an OLE period. Pts
with PNH who were treatment-naive or switched from eculizumab in Parts 2, 3, and 4,
who completed the primary treatment period through Week 20 and derived benefit from
crovalimab treatment, were eligible to enter the OLE (Röth et al. EHA 2019). Long-term
efficacy, safety, PK and PD data are presented for the OLE. Efficacy endpoints include
change in lactate dehydrogenase (LDH), transfusion avoidance and hemoglobin stabilization
(both assessed in 24-week intervals), and breakthrough hemolysis (BTH). PK and PD
endpoints include assessment of serum concentrations of crovalimab, complement activity
by liposome immunoassay (LIA), and free C5 over time.
Results: Overall, 43 of 44 pts entered the OLE and at the clinical cutoff (Nov 1,
2021), 38 pts were ongoing.
Mean normalized LDH was maintained at ≤1.5 × upper limit of normal (ULN) during the
OLE, and 80% to 100% of evaluable pts achieved LDH ≤1.5 × ULN at each visit (Figure).
Transfusion avoidance was achieved in 83% to 92% of pts over each 24-week interval
in the OLE. Similarly, hemoglobin stabilization remained relatively stable during
the OLE, with 79% to 88% of pts achieving hemoglobin stabilization across 24-week
intervals. There was a total of five BTH events during the OLE across all pts (none
leading to withdrawal) for an overall BTH rate of 0.05 events per pt-year (95% confidence
interval: 0.01, 0.11).
The incidence of adverse events (AEs) was reported cumulatively from baseline. Median
treatment duration (range) for treatment-naive and switched pts on the study was 3.4
years (0.9-4.4) and 2.9 years (0.4-3.9), respectively. Any grade AEs occurred in 95%
(42/44) of pts; none led to withdrawal from treatment. Severe AEs occurred in 17%
(3/18) of treatment-naive pts and 19% (5/26) of switched pts. Serious AEs occurred
in 32% (14/44) of pts (33% [6/18] in treatment-naive and 31% [8/26] in switched pts),
of which 5% (2/44) were assessed as related to study treatment by the investigator.
No meningococcal infections or deaths were reported.
Crovalimab concentrations were stable and within the expected range over time. Terminal
complement activity by LIA was near the lower limit of quantification (10 U/mL) and
free C5 levels were maintained at low levels (<0.001 g/L) in most pts. PK/PD relationships
between free C5 or LIA with crovalimab confirmed that crovalimab levels of ≈100 μg/mL
resulted in complete inhibition of terminal complement activity.
Image:
Summary/Conclusion: Treatment-naive and switched pts with PNH treated with crovalimab
in the COMPOSER trial maintained disease control over long-term follow-up. No new
safety signals were identified, and complete terminal complement inhibition was maintained
during the OLE. These data support the continued clinical development of crovalimab
in PNH.
P831: PATIENT-REPORTED OUTCOMES IN PATIENTS WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA
TREATED WITH CROVALIMAB: RESULTS FROM THE COMPOSER TRIAL
A. Röth1,*, J. Panse2, B. Gentile3, M. Buri4, H. Patel3, Z. Nagy5
1Department of Hematology and Stem Cell Transplantation, West German Cancer Center,
University Hospital Essen, University of Duisburg-Essen, Essen; 2University Hospital
RWTH Aachen, Aachen, Germany; 3Genentech, Inc., South San Francisco, CA, United States
of America; 4F. Hoffmann-La Roche Ltd., Basel, Switzerland; 5Semmelweis University,
Budapest, Hungary
Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a potentially life-threatening
acquired hematopoietic stem cell disorder, resulting in anemia, hemoglobinuria, and
acquired thrombophilia. Patients with PNH can experience a variety of disease-related
symptoms, particularly fatigue, that can negatively impact their day-to-day functioning
and health-related quality of life. In the Phase I/II COMPOSER trial (NCT03157635),
patients with PNH who were treatment-naive or switched from eculizumab were treated
with crovalimab, a novel anti-C5 monoclonal antibody designed with Sequential Monoclonal
Antibody Recycling Technology (Röth et al. Blood 2020).
Aims: To explore the efficacy of crovalimab from the perspective of patients with
PNH using patient-reported outcome (PRO) tools to assess fatigue, functioning, and
global health status (GHS)/quality of life (QoL).
Methods: COMPOSER included treatment-naive (Parts 2 and 4 Arm A [4A]) and switched
(Parts 3 and 4 Arm B [4B]) patients who received different crovalimab dosing regimens
for a 20-week treatment period (Figure). Paper PROs were completed at baseline and
at scheduled visits during the treatment period (up to Week 10 for Part 2 and Week
20 for Parts 3 and 4). Fatigue was assessed with the Functional Assessment of Chronic
Illness Therapy (FACIT) Fatigue scale. Additional symptoms, functioning, and GHS/QoL
were assessed with the European Organisation for Research and Treatment of Cancer
Quality of Life Questionnaire Core 30 (EORTC QLQ-C30).
Results: The PRO completion rate at baseline was 100% of evaluable patients for Parts
2 (n=10), 3 (n=19), and 4A (n=8), and 86% for 4B (n=7). Completion rates remained
≥86% for all follow-up visits.
Mean FACIT-Fatigue scores at baseline among treatment-naive patients in Parts 2 (29.7,
standard deviation [SD]=13.7) and 4A (33.9, SD=8.7) were below or within the range
for severe fatigue (30-34; Eek et al. J Patient Rep Outcomes 2021). Mean scores improved
while on treatment with crovalimab, from baseline to Week 10 in Part 2 patients (8.8,
SD=10.2) and to Week 20 in Part 4A patients (6.0, SD=6.1), exceeding the threshold
for clinically meaningful improvement (5 points; Cella et al. ASH 2021). Switched
patients in Parts 3 and 4B had less fatigue on average at baseline (37.3, SD=12.6;
37.0, SD=9.9, respectively) compared with treatment-naive patients, and showed largely
minor changes at each follow-up. Similar results were observed for the EORTC QLQ-C30
Fatigue scale. Part 2 and Part 4A patients by Week 10 and 20, respectively, had mean
fatigue severity levels comparable to (FACIT-Fatigue) or lower than (EORTC QLQ-C30
Fatigue) mean baseline values in switched patients.
Mean EORTC QLQ-C30 scores in all functioning domains (i.e., physical, role, social,
emotional, and cognitive) improved from baseline to Week 10 in Part 2 patients. Improvements
in physical, role, and social functioning only were observed in Part 4A patients at
all scheduled follow-up visits. Patients in both treatment-naive cohorts exhibited
improvements in GHS/QoL. Switched patients, on average, exhibited limited change from
baseline in EORTC QLQ-C30 scores, with mean improvements only observed for emotional
functioning in Part 4B patients.
Image:
Summary/Conclusion: Treatment-naive patients experienced improvements from baseline
in fatigue, functioning, and GHS/QoL whereas switched patients maintained baseline
levels. These data provide supportive evidence for the treatment efficacy of crovalimab
from the patient perspective.
P832: CLINICAL OUTCOMES AND IMMUNE RESPONSES TO SARS-COV-2 VACCINATION IN PATIENTS
WITH SEVERE APLASTIC ANEMIA
R. Rajput1,*, X. Ma2, K. L. Boswell3, M. Gaudinski3, E. M. Groarke1, J. Lotter1, O.
Rios1, I. Darden1, C. O. Wu2, R. A. Koup3, N. S. Young1, B. Patel1
1Hematology Branch, National Heart Lung and Blood Institute; 2Office of Biostatistics
Research; 3Vaccine Research Center, National Institute of Allergy and Infectious Diseases,
National Institute of Health, Bethesda, Maryland, United States of America
Background: Coronavirus Disease 2019 (COVID-19) caused by SARS-CoV-2 has led to significant
morbidity and mortality in patients with hematological disease. Severe aplastic anemia
(SAA) is a life-threatening bone marrow failure disorder that presents with pancytopenia
and a hypocellular marrow due to immune-mediated destruction of hematopoietic stem
cells (HSC). Patients are at high risk for infection due to neutropenia and treatment
with immunosuppressive therapy (IST). Acute COVID-19 infection has been reported to
cause worsening underlying marrow failure and death in unvaccinated SAA patients.
Patients with SAA are often not vaccinated to protect against routine viruses due
to concerns of ineffective protective specific antibody response to viral antigens
and potential pathogenic global immune system activation. To date, the clinical impact
and efficacy of COVID-19 vaccination in SAA has not been described.
Aims: Assess the impact of SARS-CoV-2 vaccination on SAA disease status and to determine
the humoral and cellular response to vaccination.
Methods: SAA treated with IST including horse antithymocyte globulin (h-ATG), cyclosporine
(CSA), and eltrombopag (EPAG) at the National Institute of Health with confirmation
of SARS-CoV-2 vaccination between January and November 2021 were included. Current
SAA disease status at the time of vaccination was recorded as complete response (CR),
partial response (PR) and non-response (NR). Univariate analysis was performed using
the Student paired t-test to compare available blood counts. Peripheral mononuclear
blood cells (n=27) and serology (n=12) after completion of vaccination series were
analyzed to assess cellular and humoral response, respectively.
Results: Blood samples from fifty patients with SAA with median age of 42 years (9-78),
29 females, and 21 males, were studied. At time of vaccination, 15 patients (30%)
were receiving CSA. Forty-seven of 50 (94%) of patients did not have any changes in
disease status after vaccination. Among 40 patients with available data 3 months prior
to and after completion of vaccination series, there was no significant difference
in HGB (p=0.52) PLT (p=0.67), ANC (p=0.98), and ALC (p=0.42; Figure 1A-C). Relapse
after vaccination, defined as progressive and substantial decline in blood counts,
was noted in 3 (6%) cases. All 3 relapsed patients were deemed weak PR at time of
vaccination (PLT < 50 K/μL) and were 6 months, 3 years, and 4 years from initial IST.
Two of the 3 patients received an initial Pfizer inoculation and did not receive the
second dose because of declining blood counts. Relapse occurred 4 weeks after completion
of a full Moderna series in the third patient, who had demonstrated a decline in counts
6 months prior to vaccination. Vaccinated SAA patients all had adequate antibody production
to the SARS-CoV-2 spike protein. They also exhibited a CD4+ Th1 dominant response,
similar to healthy controls, even when receiving CSA at time of vaccination (Figure
D). CD8+ responses to vaccination were observed, but patients with SAA appear to have
qualitatively blunted responses compared to controls.
Image:
Summary/Conclusion: SARS-CoV-2 vaccination in SAA may result in transient decline
in blood counts but true relapse is rare. Relapse may be more likely in patients whose
blood counts are declining prior to vaccination. Appropriate humoral and cellular
responses are seen in patients with SAA. A risk versus benefit assessment to vaccinate
against SARS-CoV-2 in SAA may be warranted in patients with weak partial response
to initial therapy or whom have clinical or laboratory evidence suggestive of active
disease.
P833: CATEGORIZING HEMATOLOGICAL RESPONSE TO PEGCETACOPLAN IN PATIENTS WITH PAROXYSMAL
NOCTURNAL HEMOGLOBINURIA: A POST HOC ANALYSIS OF THE PHASE 3 PRINCE STUDY DATA
A. Risitano1,*, R. Wong2, M. Al-Adhami3, J. Savage3, R. Horneff4, R. Peffault de Latour5
1Hematology and BMT Unit, AORN, San Giuseppe Moscati, Avellino, Italy; 2Sir Y.K. Pao
Centre for Cancer & Department of Medicine and Therapeutics, Prince of Wales Hospital,
The Chinese University of Hong Kong, Sha Tin, Hong Kong; 3Apellis Pharmaceuticals,
Inc., Waltham, United States of America; 4Swedish Orphan Biovitrum AB, Stockholm,
Sweden; 5French Reference Center for Aplastic Anemia and Paroxysmal Noctural Hemoglobinuria,
Assistance Publique - Hôpitaux de Paris; Université de Paris, Paris, France
Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare complement-mediated
hematological disorder characterized by anemia, hemolysis, and bone marrow failure.
Pegcetacoplan (PEG), an FDA/EMA-approved targeted C3 therapy for the treatment of
PNH, can control intravascular and prevent extravascular hemolysis. A prior post hoc
analysis of PEGASUS reported a higher proportion of patients presenting good or better
hematologic responses to PEG versus eculizumab (Risitano A, et al., Blood, 2021;138
[Supplement 1]: 1104). The PRINCE study demonstrated efficacy/safety of PEG compared
to control treatment (CTRL; supportive treatment of PNH without complement inhibitors)
in complement-inhibitor naïve patients with PNH.
Aims: This post hoc analysis categorized the hematologic response to PEG and CTRL
at baseline and Week 26 in the PRINCE Trial (phase 3, multicenter, randomized, open-label,
controlled trial [NCT04085601]).
Methods: Adult complement-inhibitor naïve (no complement-inhibitor treatment within
3 months prior to screening) patients with PNH who had low hemoglobin (Hb) levels
and high lactate dehydrogenase levels were included. Patients were randomized 2:1
to receive PEG or CTRL through Week 26. Hematologic response to PEG and CTRL was categorized
as complete, good, partial, or minor at baseline and at Week 26 using the number of
packed red blood cells transfusions needed in the last 6 months and Hb level at the
specified timepoints (revised categorization criteria per Debureaux PE, et al. Bone
Marrow Transplant. 2021; 56(10):2600-2602). Hematologic response categorization was
performed according to the following criteria: Complete response: no transfusions,
no anemia (≥12 g/dL); Good response: no transfusions, but with chronic mild anemia
(10-12 g/dL); Partial response: no/occasional transfusions (≤2 units/6 months) and
chronic moderate anemia (8-10 g/dL); Minor response: regular transfusions (≥3 units/6
months) and chronic moderate/severe anemia (<10 g/dL), or no/occasional transfusions
(≤2 units/6 months) and chronic severe anemia (<8 g/dL). Any patients with missing
data at Week 26 were evaluated using the most recent value within the 6 weeks prior.
Statistical tests assessed significance for responses of ‘Good’ or ‘Complete’, collectively
(Fisher’s Exact Test between PEG and CTRL groups at baseline; McNemar’s test within
PEG group at baseline vs. Week 26).
Results: In this study, 53 patients were randomized to PEG (n=35) or CTRL (n=18).
At baseline, most patients had a partial or minor response to supportive therapy prior
to randomization, with only 5.7% of patients in the PEG group and 0.0% in the CTRL
group presenting a Good or Complete hematologic response (p= 0.5428). Hematologic
response categorization for patients in the PEG group are shown for baseline and Week
26 (Figure). At Week 26, 80.0% of patients in the PEG group exhibited a Good or Complete
response. The proportion of patients in the PEG group who demonstrated a Good or Complete
response was significantly higher at Week 26 as compared to baseline (p<0.0001). One
patient treated with PEG had missing data at Week 26 due to being lost to follow-up
and was not evaluated.
Image:
Summary/Conclusion: This post hoc analysis of PRINCE trial data revealed that a substantial
proportion of patients achieved good or complete hematologic response to PEG after
26 weeks. These findings further support the positive efficacy of PEG on hematologic
parameters and provide evidence for the role of proximal complement inhibition on
hematological and clinical improvements for patients with PNH.
P834: REAL-WORLD EVIDENCE OF SAFETY AND EFFECTIVENESS OF ECULIZUMAB AND SWITCH TO
RAVULIZUMAB IN A SWISS PATIENT POPULATION WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA
A. Rovó1,*, L. Simeon2, M. Gavillet3, K. Samii4, N. Cantoni5, D. Heim6, G. Stüssi7,
M. Bissig8
1Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University
Hospital, Bern; 2Department of Hematology and Central Hematology Laboratory, Cantonal
Hospital Lucerne, Luzern; 3Service and Central Laboratory of Hematology, Department
of Oncology and Department of Laboratory Medicine and Pathology, Lausanne University
Hospital (CHUV), Lausanne; 4Hematology Division, Department of Oncology, Geneva University
Hospitals, Geneva; 5Division of Oncology, Hematology and Transfusion Medicine, Kantonsspital
Aarau, Aarau; 6Department of Hematology, University Hospital Basel, Basel; 7Clinic
of Hematology, Oncology Institute of Southern Switzerland, Bellinzona; 8Department
of Medical Oncology and Hematology, University Hospital of Zurich, Zurich, Switzerland
Background: Eculizumab (ECU) a complement component 5 (C5) inhibitor, is the current
standard of care for patients with paroxysmal nocturnal hemoglobinuria (PNH). Ravulizumab
(RAV) is engineered from ECU with the main advantage of having an extended 8-week
(vs 2-week with ECU) dosing interval, which decreases treatment burden markedly. Hence,
RAV is expected to become the new standard of care to treat PNH. In Switzerland, RAV
was approved for PNH treatment in 2020.
Aims: The objective of this study was to evaluate data from the PNH Swiss Soliris
and Ultomiris Reimbursement Registry (SSURR) to assess key parameters for safety and
effectiveness of ECU and to assess the adoption of RAV for PNH treatment by physicians
under real-world conditions.
Methods: The SSURR was designed in 2012 as a prospective, longitudinal, multi-center
registry study in Switzerland. Patients were recruited and followed in 9 centers.
Health-related data were collected at the first consultation (baseline) and during
follow-up visits. In this report, results are based on data collected from 23 February
2012 to 30 September 2021. Data were summarized using descriptive analysis. Ethics
approval was obtained. All participants provided written informed consent.
Results: In total, 56 patients with a median age of 51 (range 20-84) years were enrolled
in the Registry, 52% were female. For 42 patients, the 12-month follow-up (FU) was
available: 39 patients on ECU and 3 on RAV (switched from ECU). Median FU was 27 (range
12-108) months. Hemoglobin (Hb) mean was 63 ± 47 g/L at baseline (n=41), and Hb mean
was 105 ± 25 g/L (n=26) at the last visit. The lactate dehydrogenase (LDH) decreased
from a mean 1457 ± 706 U/L (baseline, n=42) to 403 ± 212 U/L after 12 months FU (n=40;
65% of patients < 1.5 × upper limit of normal [ULN]), and was for 88% of the patients
< 1.5 × ULN at their last visit (n=24). Thrombosis events (TE) were reported in 33%
of patients prior to treatment start (n=42) and 3 (7%) had thrombosis during FU of
which 2 patients had no prior history of TE. Within 12 months prior to treatment start,
21 patients (50%) received one or more red blood cell (RBC) transfusions. At 12 months
of FU, 91% (n=42) were RBC transfusion-free. RBC transfusions remained reduced (82%;
n=27 at last visit). After 12 months FU, 19% (n=8) of the patients had withdrawn from
the registry due to allogenic bone marrow transplantation, low PNH clone size, transformation
to hypoplastic MDS, and 5 non-disease related reasons.
Eight patients from the 56 enrolled received RAV as initial treatment. They were all
C5-inhibitor treatment-naïve with a mean LDH level of 1481 ± 1432 U/L at inclusion,
which decreased to 230 ± 62 U/L at the 6-month visit (n=4; all below 1.5 × ULN). Of
the 56 enrolled patients with PNH, 43% were switched from ECU to RAV. These patients
(58% female) had a median age of 55 (range 26-85) years and 75% of them had LDH <
1.5 × ULN at their last LDH measurement on ECU before RAV administration. There were
7 patients with a 6-month FU after switching at which 6 of them had a LDH below 1.5
× ULN.
Summary/Conclusion: The current analysis provides real world evidence for the safety
and effectiveness of ECU in the Swiss PNH patient population, demonstrated by pronounced
reduction in LDH, low need for RBC transfusion and stable hemoglobin as well as reduced
occurrence of thrombosis. The fast adoption of RAV demonstrates RAV may likely become
the new standard of care for patients with PNH. In time, more data will become available
for safety and effectiveness of RAV under real-world conditions.
P835: GENETIC BACKGROUND AND CLINICAL CHARACTERISTICS OF CONGENITAL NEUTROPENIAS IN
ISRAEL
O. Steinberg-Shemer1,2,3,*, L. Yeshareem4, S. Noy-Lotan3, O. Dgany3, T. Krasnov3,
G. Berger Pinto1, B. Bielorai2,5, A. A. Kuperman6,7, R. Laor8, N. Mandel-Shorer9,10,
A. Ben Barak9, C. Levin10,11, A. Mahdi12, H. Miskin12, S. Revel-Vilk13,14, D. Levin15,
M. Benish15, T. Zuckerman10,16, O. Wolach2,17, I. Pazgal2,18, O. Gilad1,2, T. Goldberg1,
J. Yacobovich1,2, S. Izraeli1,2, H. Tamary1,2,3
1Hematology-Oncology, Schneider Children’s Medical Center of Israel, Petach Tikva;
2Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv; 3Pediatric Hematology
Laboratory, Felsenstein Medical Research Center; 4Pediatrics A, Schneider Children’s
Medical Center of Israel, Petach Tikva; 5Pediatric Hematology-Oncology, Sheba Medical
Center, Tel Hashomer; 6Blood Coagulation Service and Pediatric Hematology Clinic,
Galilee Medical Center, Nahariya; 7Azrieli Faculty of Medicine, Bar-Ilan University,
Safed; 8Hematology Service, Bnei Zion Medical Center; 9Department of Pediatric Hematology-Oncology,
Ruth Rappaport Children’s Hospital, Rambam Healthcare Campus; 10Rappaport Faculty
of Medicine, Technion-Institute of Technology, Haifa; 11Pediatric Hematology Unit
and Research Laboratory, Emek Medical Center, Afula; 12Pediatric Hematology, Soroka
University Medical Center, Ben-Gurion University, Beer Sheva; 13Pediatric Hematology/Oncology
Unit, Shaare Zedek Medical Center; 14Faculty of Medicine, Hebrew University, Jerusalem;
15Pediatric Hemato-Oncology, Tel Aviv Medical Center, Tel Aviv; 16Hematology and Bone
Marrow Transplantation Institute, Rambam Healthcare Campus, Haifa; 17Institute of
Hematology, Davidoff Cancer Center, Rabin Medical Center; 18Comprehensive Center of
Thalassemia, Hemoglobinopathies & Rare Anemias, Institute of Hematology, Beilinson
Hospital, Rabin Medical Center, Petach Tikva, Israel
Background: Congenital neutropenias comprise a heterogenous group of disorders causing
low neutrophil counts with a variable clinical presentation. Patients may suffer from
severe infections and have a high risk of myeloid transformation. To date, over 30
genes are known to cause congenital neutropenias, with a diverse relative frequency
in different ethnicities. Understanding the spectrum of genetic causes of neutropenia
in ethnically diverse populations and the related clinical presentation may direct
the molecular work-up and clinical care. Israel is enriched with multiple ethnicities
and high consanguinity.
Aims: To evaluate the clinical and genetic spectrum of patients with congenital neutropenias
in Israel.
Methods: All patients diagnosed with congenital neutropenia from the Israeli Inherited
Bone Marrow Failure registry were included. Data were extracted from the registry
and patients’ charts. Sanger sequencing was first performed for ELANE or G6PC3, followed
by less commonly mutated genes. In recent years, patients with wild-type ELANE/G6PC3
were referred for targeted next generation sequencing, followed by whole exome sequencing.
Results: The cohort encompasses 66 patients, from 50 families, including 7 with cyclic
neutropenia. Fifty patients (76%) were genetically diagnosed; 22 (33% of the cohort)
had mutations in ELANE, 9 (13.6%) had homozygous G6PC3 mutations, 7 (10.6%) SBDS mutations,
6 (9%) patients had SRP54 mutations, two each had heterozygous GFI1 and homozygous
JAGN1 mutations and one each had TAZ and SLC37A1 mutations. All patients with G6PC3
and GFI1 mutations were of consanguineous Muslim Arab origin. Extra-hematopoietic
manifestations were common among patients with mutations in G6PC3. Most patients (82%)
presented with severe infections and this was also the most common cause of death.
Eight patients (12%) developed myeloid transformation; in 6 of those a somatic molecular
work-up was performed, and truncating mutations in the granulocyte-colony stimulating
factor were detected in 4. Eighteen (27%) patients underwent hematopoietic stem cell
transplantation, most often due to insufficient response to treatment with granulocyte-colony
stimulating factor.
Summary/Conclusion: The prevalence of genes causing congenital neutropenias in Israel
is unique, as it includes a relatively high frequency of patients with mutations in
G6PC3 and an absence of patients with HAX1 mutations, thus directing the genetic work-up
and the clinical care. Similar to other registries, a molecular diagnosis was not
achieved for 24% of the patients, suggesting yet unknown genetic causes.
P836: LONG-TERM DYNAMICS OF CLONAL HEMATOPOIESIS IN CHRONIC IDIOPATHIC NEUTROPENIA
(CIN)
G. Tsaknakis1,2,*, S. Papadakis1,2, P. Kanellou3, E. Boutakoglou1,2, I. Mavroudi1,2,
C. Pontikoglou1,2, E. Papadaki1,2
1Haemopoiesis Research Laboratory, University of Crete School of Medicine; 2Department
of Hematology, University Hospital of Heraklion; 3Department of Hematology, Venizeleion
General Hospital, Heraklion, Greece
Background: We have previously performed next generation sequencing (NGS) analysis
of genes recurrently mutated in myeloid malignancies in a cohort of CIN patients.
We have estimated for the first time the frequency of clonal hematopoiesis in these
patients and found that clonal CIN patients have significantly higher risk to develop
MDS/AML compared to non-clonal CIN patients.
Aims: To conduct longitudinal follow-up NGS analyses of myeloid genes in CIN patients
in order to assess clonal dynamics and evolution of clonal hematopoiesis in these
patients.
Methods: We have performed longitudinal NGS analyses in 53 patients with CIN diagnosis
(mean absolute neutrophil counts -ANCs- 1320±460/μL; median 1500, range 200-1700/μL)
according to previously published criteria i.e no evidence of any underlying condition
related to neutropenia after an extended clinical/laboratory investigation including
bone marrow biopsy, karyotype, immunophenotype. Genomic DNA was extracted from bone
marrow or peripheral blood samples at baseline and follow-up timepoints, sequencing
libraries were prepared and subjected to targeted NGS on an Ion S5 Prime Sequencer
(Thermo Fisher Scientific) using a panel of 38 myeloid genes.
Results: Longitudinal NGS analysis in the patients was performed over a period of
4-173 months (median 31 months). The incidence of clonal hematopoiesis was similar
at baseline (16/53 i.e. 30.2%) and follow-up (16/53 i.e. 30.2%) (Figure 1A). The mutation
spectrum (i.e. absence of mutations, multiple mutated genes or presence of a single
mutated gene) at baseline (Figure 1B) was comparable to that at follow-up (Figure
1C). In total, 28 mutations were detected in 17 CIN patients at one or both time-points.
The variant allele frequencies (VAF) did not show a significant difference at baseline
(9.68%) and follow-up (13.23%) and correlation analysis showed that VAFs at both points
strongly correlated (r=0,59; P<0.01), with the majority of CIN patients showing a
stable clone size (Figure 1D). We performed the same analysis for the most frequently
mutated genes in our cohort of patients, namely DNMT3A, TET2, SRSF2 (Figure 1E-G).
Overall, DNMT3A mutations did not show any positive selection over time, with a median
increase in VAF of 1.78% (P=0.67) during the follow-up period (median period of 47.5
months, range 12-164). Similarly non-significant increases in VAF was seen for TET2
(0.49%) and SRSF2 (0.04%), within a median period of 26 months (range 22-111) and
55 months (range 12-98), respectively. One patient with newly developing clonal CIN
and one patient with disappearance of clonal hematopoiesis were found at follow-up
after 4 and 98 months, respectively. Two patients acquired a second mutation at follow-up.
At baseline they carried mutations in SRSF2 and ZRSR2, respectively and at follow-up
they both developed TET2 truncating mutations. No significant changes at baseline
and follow-up in ANCs were observed in patients with clonal disease. Overall, four
patients (7.55%) transformed to AML/MDS.
Image:
Summary/Conclusion: In the majority of CIN patients tested for clonal evolution over
time, most mutant hematopoietic clones appeared to be remarkably stable, with limited
VAF expansion over the follow-up period and no acquisition of new molecular alterations.
Only two patients acquired an additional mutation over time and this propensity was
associated with mutations in spliceosome genes. None of the individuals bearing DNMT3A
or TET2 mutations acquired additional mutations. This study contributes to the better
understanding of clonal hematopoiesis in CIN.
P837: ELTROMBOPAG (EPAG) INDUCES HEMATOLOGIC RESPONSES FOR PATIENTS WITH POST-ALLOGENIC
HEMATOPOIETIC STEM CELL TRANSPLANT POOR GRAFT FUNCTION (POST-ALLO HSCT PGF): RESULTS
OF THE ELTION STUDY IN SPAIN
C. Vallejo1,*, I. García2, F. Sanchez Guijo3, M. Cuesta4, C. Solano5, A. Sampol6,
T. Torrado7, M. Vendranas8, M. Marigil8
1University Hospital Donostia, San Sebastian, Spain, University Hospital Donostia,
San Sebastian, Spain, Santiago de Compostela; 2Department of Hematology, University
Hospital Santa Cruz y San Pau, Barcelona; 3Department of Hematology, University Hospital
of Salamanca, Salamanca; 4Department of Hematology, Carlos Haya University Hospital,
Malaga; 5Department of Hematology, Hospital Clinico de Valencia, Valencia; 6Department
of Hematology, University Hospital Son Espases, Palma de Mallorca; 7Department of
Hematology, University Hospital Alvaro Cunqueiro, Vigo; 8Medical Affairs Department,
Novartis, Barcelona, Spain
Background: Persistent cytopenia due to poor graft function (PGF) is a life-threatening
complication in patients undergoing allogeneic HSCT (allo-HSCT). Several therapeutic
approaches have been tested in this subset of patients with poor clinical results.
Aims: The objective of this multicenter open-label interventional prospective phase
II Novartis study (ELTION, ClinicalTrials.gov id: NCT03718533), was to analyze efficacy
and safety of EPAG in patients with post-allo-HSCT poor graft function.
Methods: Adult patients diagnosed with PGF (defined as severe cytopenia after day
+30 post-transplant, with two or more of the following: platelets <20000/µL -mandatory-,
ANC <1000/µL, hemoglobin< 10 g/dL), and full donor chimerism, were eligible to enter
the trial. Study treatment consisted of EPAG. at 150 mg/day administered up to 36
weeks; dose adjustments were contemplated as per protocol on an individual basis.
The primary efficacy endpoint was the overall hematologic response (partial and complete),
as determined by platelet, hemoglobin and neutrophil counts by 16 weeks after the
initiation of EPAG.
Results: Although the aim of the study was to include 33 patients, recruitment stopped
prematurely due to the difficulties for hospital visits posed during COVID-19 pandemic,
and eventually only 10 patients were included. The decision for this premature termination
is not related to any safety concern related to the drug. Patient characteristics
are shown in the Table 1 attached below. At EPAG. initiation, all 10 patients showed
thrombocytopenia (<20000/µcL), 5 presented with anemia (Hgb <10 g/dL), and 4 had neutropenia
(ANC <1000/µcL). Four patients discontinued EPAG before week 12 due to: disease progression/relapse
(2 patients), protocol deviation (1 patient), and CMV infection (1 patient). In none
of the cases, the event was related to study drug. At week 16, 4 patients (4/10, 40%)
and at week 24, 5 patients, showed improvement in at least one of the 3 hematologic
cell lines (partial response), respectively. Counts pre- and post-EPAG and global
response in patients who stayed on treatment > 12 weeks are displayed below:
Image:
Summary/Conclusion: In our experience, EPAG worked well in subjects with PGF, an otherwise
life-threatening condition for patients, and its use at 150 mg/day is safe and well
tolerated in this setting. Our data suggest that eltrombopag might improve hematologic
cell counts in patients with PGF, especially in those patients who remained on treatment
at week 24, however further research is warranted to extend its applicability for
larger cohorts
P838: PEGCETACOPLAN RAPIDLY STABILIZES COMPLEMENT INHIBITOR NAÏVE PATIENTS WITH PAROXYSMAL
NOCTURNAL HEMOGLOBINURIA EXPERIENCING HEMOLYSIS WITH ACUTE HEMOGLOBIN DECREASES; PRINCE
TRIAL POST HOC ANALYSIS
R. Wong1,*, M. Al-Adhami2, J. Savage2, R. Horneff3, M. Yeh4, T. Dumagay5, D. Gomez-Almaguer6
1Sir Y.K. Pao Centre for Cancer & Department of Medicine and Therapeutics, Prince
of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong; 2Apellis
Pharmaceuticals, Inc., Waltham, United States of America; 3Swedish Orphan Biovitrum
AB, Stockholm, Sweden; 4Apellis Pharmaceuticals, Waltham, Waltham, United States of
America; 5Department of Cellular Therapeutics, Makati Medical Centre, Makati, Philippines;
6Hematology Service, Hospital Universitario “Dr José Eleuterio González”, Universidad
Autónoma de Nuevo León, Monterrey, Mexico
Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare and life-threatening
disease characterized by complement-mediated hemolysis and thrombosis. Pegcetacoplan
(PEG) is the first FDA/EMA-approved C3 complement-inhibitor that provides broad hemolysis
control in patients with PNH.
Aims: This is the first report from the PRINCE trial on the efficacy of PEG in control
arm rescue patients using a post hoc time-aligned analysis.
Methods: PRINCE is a phase 3, randomized, open-label study, that evaluates the efficacy
of PEG in improving lactate dehydrogenase (LDH) and hemoglobin (Hb) levels in patients
with PNH. 53 adult (≥18 years old), complement-inhibitor naïve (no prior eculizumab/ravulizumab)
patients with LDH ≥1.5x the upper limit of normal (ULN) (≥339 U/L) and Hb below the
lower limits of normal (LLN) (males: ≤13.6 g/dL; females: ≤12.0 g/dL), were enrolled.
Patients were randomized 2:1 to receive PEG (1080 mg subcutaneously twice weekly,
n=35; PEG arm) or to the Control arm (trials were in regions where C5 inhibitors were
not available, n=18) for 26 weeks. Patients in the Control arm had the option to escape
to PEG treatment (Rescue arm) should they exhibit signs of hemolysis and acute Hb
decline by ≥2 g/dL from their baseline. Key endpoints included both changes from baseline
in LDH and Hb levels. Because patients could be rescued at any point, a post hoc analysis
was performed to time-align the Rescue patients (n=11), based on the start of PEG
dosing, with patients in the PEG arm, to compare changes in LDH and Hb levels over
time by PEG exposure.
Results: 61% (11/18) of Control arm patients experienced acute Hb decreases allowing
rescue by PEG treatment. All Rescue patients had a pre-rescue LDH ≥2X ULN (mean 2424.0 U/L)
and all had a rapid response to PEG treatment with a significant decrease in LDH level
by Week 1 (mean 422.1 U/L) with 5/11 patients achieving ≤1.5X ULN; by Week 2 mean
LDH levels were in the normal range (186.6 U/L) with 8 patients below ULN and 2 patients
below 1.5X ULN, suggesting complete control of intravascular hemolysis. These results
are similar to PEG arm patients (150.4 U/L) (Figure A).
Seven of the eleven patients showed a significant increase in Hb level (Week 2 mean
Hb for Rescue patients: 10.9 g/dL; PEG patients: 11.19 g/dL); four patients showed
no change, potentially due to underlying bone marrow dysfunction (3 had a history
of aplastic anemia). Three patients had a transfusion within 5 days of starting PEG
treatment; 1 of them remained transfusion dependent after rescue and did not have
a Hb change.
The time-align plots (Figures A and B) show Rescue patients had rapid improvements
in LDH to normal levels and Hb improvements similar to PEG arm patients.
PEG demonstrated a favorable risk-benefit profile. No deaths or serious adverse events
deemed related to PEG occurred and no drug discontinuations due to PEG-related adverse
events were reported. The most common AEs were injection site reaction (PEG [including
Rescue patients for all AEs], 30.4%, n=14; Control arm, 0.0%), hypokalemia (PEG, 13.0%,
n=6; Control arm, 11.1%, n=2), dizziness (PEG, 10.9%, n=5; Control arm, 0.0%), and
fever (PEG, 8.7%, n=4; Control arm, 0.0%).
Image:
Summary/Conclusion: PEG treatment was able to rapidly stabilize complement-inhibitor
naïve patients experiencing acute hemolysis with substantial Hb decrease, and normalize
LDH and raise Hb levels. The treatment effect of PEG on both LDH and Hb levels over
time was largely similar between the PEG arm patients and Rescue patients. PEG represents
a new effective therapeutic option with a favorable safety profile for PNH patients.
P839: PATIENTS WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA TREATED WITH PEGCETACOPLAN
SHOW IMPROVEMENTS IN D-DIMER NORMALIZATION AND DECREASE IN INCIDENCE OF THROMBOSIS
I. Weitz1,*, M. Al-Adhami2, J. Min2, E. Persson3, M. Yeh2, J. Savage2, D. Dingli4
1Jane Anne Nohl Division of Hematology, Keck-USC School of Medicine, Los Angeles;
2Apellis Pharmaceuticals, Waltham, United States of America; 3Swedish Orphan Biovitrum
AB, Stockholm, Sweden; 4Mayo Clinic, Rochester, United States of America
Background: Thrombosis is the main life-threatening complication of paroxysmal nocturnal
hemoglobinuria (PNH), a rare acquired hematologic disease. Pegcetacoplan (PEG) is
a C3 complement-inhibitor approved by the FDA/EMA for treatment of adults with PNH.
Aims: This post hoc analysis examined incidence of thrombosis, anti-thrombotic therapy
(ATT), and D-dimer normalization in adult patients with PNH treated with PEG in the
Phase 3 PEGASUS (NCT03500549) and PRINCE (NCT04085601) studies.
Methods: PEGASUS enrolled adult patients with PNH with prior suboptimal response to
eculizumab (ECU) despite stable ECU treatment (≥3 months) and hemoglobin (Hb) levels
<10.5 g/dL at screening. For the randomized controlled period (RCP), patients were
randomized 1:1 to ECU (n=39) or PEG (n=41; 1080 mg subcutaneously [sc] 2x weekly)
for 16 weeks. Thereafter, ECU patients were switched to PEG monotherapy and PEG RCP
patients remained on PEG during the open-label period (OLP, n=77) through Week 48.
PRINCE compared PEG treatment (n=35; 1080 mg sc 2x weekly) in complement-inhibitor-naïve
patients to patients receiving control treatment (CTRL; excluding complement-inhibitors
i.e., ECU/ravulizumab; n=18). CTRL patients had the option to escape to the PEG group
if Hb levels decreased ≥2 g/dL from baseline (n=11). Safety analyses in all trials
included monitoring of thrombotic events. D-dimer normalization, defined as D-dimer
level less than the upper limit of normal (0.5 µg/mL) was examined at baseline, Week
8, and the end of the study period (PEGASUS: 16 weeks [RCP] and 48 weeks [OLP], PRINCE:
26 weeks).
Results: Prior to study entry, 31% (n=25) of PEGASUS patients (6 of these while on
ECU therapy) and 9% (n=5) of PRINCE patients experienced at least one type of thrombosis.
During the PEGASUS OLP, 2 of 77 patients experienced a thrombotic event, one in the
setting of non-Hodgkin’s lymphoma and one during sepsis; neither event was deemed
related to PEG. No patients in the PEGASUS RCP or PRINCE trial experienced a thrombotic
event.
Pooled analysis of completed clinical trials of PEG including PEGASUS and PRINCE indicated
there were 2 cases of thrombosis in 164 patients comprising 130 patient-years (1.54
events per 100 patient-years), compared to 1.77 events per 100 patient-years for patients
on ECU treatment before entry into PEGASUS.
In PEGASUS, 34% (n=27) of patients were on ATT prior to study entry. During the PEGASUS
RCP, concomitant ATT was observed in 37% (PEG, n=15) and 33% (ECU, n=13) of patients,
and during the PEGASUS-OLP in 23% (n=18) of patients (PEG monotherapy). In PRINCE,
concomitant ATT decreased from 21% (n=11) prior to the study to 7% (PEG, n=3) and
0% (CTRL, n=0) during the study.
In PEGASUS, D-dimer normalization in the PEG arm increased from 73% (baseline, n=30)
to 89% (Week 8, n=33) and was largely sustained at Week 16 and Week 48 (Table 1).
D-dimer normalization in the ECU arm increased once ECU patients switched to PEG monotherapy,
from 76% (Week 16, n=29) to 89% (Week 48, n=17) (Table 1). In PRINCE, D-dimer normalization
in the PEG arm increased from 51% (baseline, n=18) to 67% (Week 8, n=18) and increased
further to 68% (n=19) at Week 26 (Table 1).
Image:
Summary/Conclusion: Overall, these results demonstrate that PEG treatment can increase
D-dimer normalization and reduces incidence of thrombotic events in patients with
PNH who are complement-inhibitor naïve or remained anemic after ECU treatment, suggesting
that PEG treatment is as effective as ECU in these outcomes.
P840: A MATCHING-ADJUSTED INDIRECT COMPARISON OF THE EFFICACY OF PEGCETACOPLAN USING
PRINCE TRIAL DATA VERSUS RAVULIZUMAB AND ECULIZUMAB IN COMPLEMENT-NAÏVE PATIENTS WITH
PAROXYSMAL NOCTURNAL HEMOGLOBINURIA
R. Wong1, J. Fishman2,*, K. Wilson3, M. Yeh2, Z. Hakimi3, C. Yee4, L. Huynh4, M. S.
Duh4
1Sir Y.K. Pao Centre for Cancer & Department of Medicine and Therapeutics, Prince
of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong; 2Apellis
Pharmaceuticals, Waltham, United States of America; 3Swedish Orphan Biovitrum AB,
Stockholm, Sweden; 4Analysis Group, Boston, United States of America
Background: Pegcetacoplan (PEG) is a PEGylated peptide targeting proximal complement
protein C3 that has been shown to control both intravascular and extravascular hemolysis
and recently approved by the FDA and EMA for the treatment of paroxysmal nocturnal
hemoglobinuria (PNH). PRINCE (NCT04085601), was a phase-3, randomized, open-label
trial assessing the efficacy and safety of PEG compared to control (CTL) treatment,
excluding complement inhibitors among treatment naïve patients with PNH. PEG demonstrated
superior efficacy to CTL by improving hemoglobin (Hb) levels and reductions in lactate
dehydrogenase (LDH) levels in patients with PNH. To date, no head-to-head clinical
trials exist comparing PEG to complement inhibitors in treatment naïve patients.
Aims: To assess the comparative effectiveness of PEG for complement-naïve patients
with PNH against ravulizumab (RAV) and eculizumab (ECU) using matching-adjusted indirect
comparison (MAIC) methodology to conduct cross-trial comparisons.
Methods: Individual patient data from PRINCE as well as aggregated published results
of RAV and ECU in the ALXN1210-PNH-301 (“301”) study (Lee et al. 2019) were used.
To adjust for cross-study differences, propensity score weighting was developed using
logistic regression analyses and used to balance demographic and clinical characteristics
between the PEG arm and RAV and ECU arms from 301. Outcomes were compared at 26 weeks
using unanchored MAIC between PEG versus RAV and PEG versus ECU and included mean
and percent change in LDH levels from baseline, LDH normalization (defined as LDH
levels dropping <1xULN [246 U/L] in the absence of transfusions during the randomized
controlled period), Hb stabilization (defined as the avoidance of a ≥2 g/dL decrease
in hemoglobin level in the absence of transfusions during the randomized controlled
period), and transfusion avoidance. Weighted Wald tests and 95% confidence intervals
were computed for comparisons of categorical and continuous outcomes.
Results: A total of 34 patients were included in the PEG arm, 125 patients in the
RAV arm, and 121 patients in the ECU arm. Prior to matching, significant differences
existed between arms in the proportion of patients with White and American Indian
or Alaska Native race, and mean LDH (all p<0.001). After matching the PEG to the RAV
and ECU arms, separately, most patient characteristics were well balanced with a mean
age of 43.8 years, 44.8 years, 46.2 years, respectively, 43.0-50.0% females, and a
mean Hb of 9.4-9.6g/dL across arms; however, there were significant differences in
Asian race and baseline LDH (all p<0.01). At week 26, patients in the PEG arm showed
a significantly greater improvement in their LDH levels from baseline, with a mean
of -523 U/L (-11.2%) compared to RAV and -578 U/L (-12.0%) compared to ECU (all p<0.001;
Figure 1). Patients receiving PEG also showed significant improvements in LDH normalization
(35.6% compared to RAV, p<0.001; 39.6% compared to ECU, p<0.001), Hb stabilization
(28.2% compared to RAV, p<0.001; 31.7% compared to ECU, p<0.001), and transfusion
avoidance (22.6% compared to RAV, p<0.001; 30.1% compared to ECU, p<0.001; Figure
1).
Image:
Summary/Conclusion: In the absence of head-to-head comparisons, this unanchored MAIC
study shows that PEG is more efficacious versus RAV and ECU in complement inhibitors
treatment naïve PNH patients in terms of significant improvements across all evaluated
endpoints, Hb stabilization, LDH improvement, LDH normalization and transfusion avoidance.
P841: EXTRACELLULAR VESICLE PROTEINS AS BIOMARKER OF OUTCOME IN MULTIPLE MYELOMA -
A REAL-WORLD STUDY
E. Arnault Carneiro1,*, B. Ferreira1,2,3, C. Pestana1,4, F. Barahona1,3, J. Caetano1,3,5,
R. Lopes1,6, P. Lúcio5, M. Neves5, H. C. Beck7, A. S. Carvalho8, R. Matthiesen8, B.
Costa-Silva9, C. João1,3,5
1Myeloma Lymphoma Research Group, Champalimaud Foundation, Lisbon; 2Hemato-Oncology
Unit, Champalimaud Foundation, Odense; 3NOVA Medical School, NOVA Medical School;
4Centre of Statistics and its Applications, Faculty of Sciences, University of Lisbon;
5Hemato-Oncology Unit, Champalimaud Foundation; 6Faculty of Medicine, University of
Lisbon, Lisbon, Portugal; 7Centre for Clinical Proteomics, Odense University Hospital,
Odense, Denmark; 8Computational and Experimental Biology, CEDOC, NOVA Medical School;
9Systems Oncology Group, Champalimaud Foundation, Lisbon, Portugal
Background: Multiple myeloma (MM) is a hematological malignancy characterized by clonal
plasma cells accumulation within the bone marrow. MM diagnosis and prognosis rely
on bone marrow biopsy. Given clonal plasma cell patchy distribution, bone marrow biopsies
may not reflect disease heterogeneity whilst submitting patients to invasive procedures.
Liquid biopsies, such as extracellular vesicles (EV) from peripheral blood, may overcome
these limitations. In MM, there is evidence that EV intervene in key processes such
as tumor progression and drug resistance. Most studies analyzing EV from MM patients
focus on genome, and real-world studies on EV proteome are scarce.
Aims: We characterized blood and bone marrow samples from the precursor stage, MGUS
and from MM patients to determine whether EV content can discriminate between diagnosis
and predict patient prognosis. These results were compared to healthy donor samples.
Methods: After informed consent signed, a cohort of 102 patients and 19 healthy donors
were followed in median time of 25 months. Blood and BM samples were collected at
diagnosis, response and relapse of patients. EV were isolated by ultracentrifugation.
Protein and EV particle concentrations were quantified. Proteomic analysis was performed
by mass spectrometry (LC-MS/MS). Diagnostic and prognostic impacts of EV characteristics
were investigated. A multivariable longitudinal logistic regression model was built
to determine the association with common myeloma-related blood parameters.
Results: We report a set of peripheral blood EV-proteins (PDIA3, C4BPA, BTN1A1, APRIL,
PSMB8 and PDE8B) that have the potential to be used as new diagnosis biomarker for
myeloma patients. Functional enrichment analysis revealed that proteins differentially
expressed in patient vs healthy donors are strongly related to immune response, supporting
increased immune dysfunctions in patients with active multiple myeloma.
We identified that the level of EVcargo (protein/particle ratio) is significantly
associated to patient overall survival. High EVcargo patients (>0.6 µg/108 particles)
had a shorter overall survival compared to low EVcargo patients (≤0.6 µg/108 particles).
High EVcargo is associated with high sFLC lambda, IgA immunoparesis and shorter time
in response. The downregulation of IGHA1/IGHA confirmed patients immunoparesis. Upregulation
of Ig lambda production proteins confirmed the increased presence of sFLC in the same
patients. Also, we report an association between the progression free survival time
and expression level at diagnosis of SERPINA2, RPS26 and UBBP4, suggesting their potential
as prognosis biomarkers.
Image:
Summary/Conclusion: To our knowledge, this is the first report of EV protein content
from a real-world MGUS and MM patient cohort followed for more than 2 years. Our findings
show that PDIA3, BTN1A1, APRIL and complement proteins should be further explored
in myeloma, as peripheral blood biomarkers of diagnosis. Circulating EVcargo is a
promising prognostic biomarker for MM patients. Patients with high EVcargo had 12
times increased risk of dying and high EVcargo is associated with important prognostic
features such as immunoparesis, sFLC or duration of treatment response. EVcargo is
an indirect and more affordable measure to infer EV protein load compared to mass-spectrometry.
Our results show that EV have the potential to be used as diagnosis biomarker in MGUS
and MM patients and as an added approach to discriminate patients with poor survival.
Our results substantiate the interest of EV as liquid biopsies in myeloma and future
validation in independent clinical settings is urged.
P842: NOVEL MULTIFUNCTIONAL TETRAVALENT CD38 NKP46 FLEX-NK ENGAGERS ACTIVELY TARGET
AND KILL MULTIPLE MYELOMA CELLS
L. Lin1, H.-M. Chang1, C. Nakid2, S. Frankel1, D. Wu3, J. Kadouche4, D. Teper4, O.
Mandelboim5, J.-C. Bories2, A. Arulanandam1,*, W. Li1
1Cytovia Therapeutics, Natick, United States of America; 2INSERM U976 and Université
de Paris, Paris, France; 3Lynx Biosciences, San Diego; 4Cytovia Therapeutics, Aventura,
United States of America; 5Hebrew, University of Jerusalem, Jerusalem, Israel
Background: Given that CD38 is a clinically validated target for natural killer (NK)
cell mediated cytotoxicity in multiple myeloma (MM), we sought to leverage our FLEX-NKTM
platform to create a NK engager antibody targeting CD38. FLEX-NKTM is a proprietary
platform for production of tetravalent IgG1-like multifunctional NK engager antibodies
with a novel FLEX-linker to allow for simultaneous binding of both the targeted cancer
cells and NK cells via the activation receptor NKp46.
Aims: Derive preclinical proof of concept with CD38 targeted NK-cell engager bispecific
antibody in Mulitple Myeloma
Methods: CYT-338 was expressed in CHOZEN cells and purified by step column chromatography.
PB-NK cells were purified from healthy donor blood. PB-NK cytotoxicity against MM
tumors was evaluated in the presence of CYT-338 or isotype control antibodies at a
fixed E/T ratio. CYT-338 pharmacokinetics in mice was evaluated following a single
intravenous injection. Anti-tumor efficacy of CYT-338 was evaluated in NSG-mice injected
with PB-NK cells and MM cell line MM1S expressing luciferase. CYT-338 mediated fratricide
was evaluated in purified PB-NK’s. CYT-338 cytokine release assessments and hemato-cytotoxicity
was evaluated in-vitro in human PBMC assays.
Results: CYT-338 showed dose dependent binding to CD38 expressing MM cell lines MM1S
and KMS11 and no binding to a CD38 knock out MM.1S cell line. CYT-338 bound MM cell
lines with ~ 2-fold higher mean fluorescence intensity than anti-CD38 monoclonal antibody
(mAb) or daratumumab alone. Epitope mapping studies for our CD38 mAb binder using
alanine scanning mutagenesis showed that amino acid S274 on CD38, critical for binding
to daratumumab, is not important for CD38 binding, suggesting a distinct epitope detected
by our CD38 binder. CYT-338 showed greater dose dependent NK cell redirected cytolysis,
degranulation and cytokine production against MM1S cells compared to daratumumab.
The CYT-338 pharmacokinetics study in mice showed a terminal half-life of 41 hrs.
Preliminary in-vivo tumor model studies in NSG mice using PB-NK and CYT-338 injections
showed tumor growth inhibition of MM1S tumor cells. Low NK cell fratricide was observed
with CYT-338 while daratumumab showed significantly higher NK cell fratricide. In
peripheral blood mononuclear cell hemato-toxicity studies, depletion of monocytes
and NK cells were observed with daratumumab but no significant depletion was observed
with CYT-338. Cytokine release assessment of CYT-338 in the human PBMC assay showed
no evidence of cytokine release while high levels of cytokine release was observed
with daratumumab, anti-CD28 (TGN1412) and CD3 antibody controls.
Summary/Conclusion: These results suggest that the CYT-338 engager has a favorable
NK cell engager profile for targeting CD38 expressing multiple myeloma distinct from
daratumumab.
P843: METABOLOMIC CHARACTERIZATION OF HUMAN MULTIPLE MYELOMA CELL LINE TO STUDY TUMOR
RESISTANCE TO DIFFERENT CLASSES OF THERAPEUTIC AGENTS
A. Steer1,*, D. Chemlal1, E. Varlet2, A. Machura1, A. Kassambara1, E. Alaterre2, G.
Requirand3, N. Robert3, C. Hirtz4, H. De Boussac1, A. Bruyer1, J. Moreaux2
1Diag2Tec; 2Institute of Human Genetics, UMR 9002 CNRS-UM; 3Laboratory for Monitoring
Innovative Therapies, Department of Biological Hematology, CHU Montpellier; 4Clinical
Proteomics Platform, LBPC, IRMB, CHU, UM Montpellier, Montpellier, France
Background: Multiple myeloma (MM) is the second most common hematological malignancy,
characterized by the abnormal accumulation of plasma cells in the bone marrow. Although
the latest treatments, including proteasome inhibitors, immunomodulating agents, or
immunotherapy, have greatly improved patient survival, a residual subset of cells
remains resistant to therapies and usually causes relapses.
Aims: The drug resistance could be explained, among others, by the interactions with
the microenvironment, MM’s high molecular heterogeneity, or the appearance of adaptive
survival mechanisms after treatment exposure. However, a better understanding of the
mechanisms involved in drug resistance remains of significant interest. Among the
factors influencing the resistance of cancer cells, the “metabolic plasticity” of
the tumor and, therefore, its ability to adapt to stress conditions is a mechanism
increasingly studied in recent years in cancer. Although measuring mitochondrial metabolism
has been identified as a major factor influencing response to treatments in several
cancers, few studies have been documented in MM.
Methods: Here, we characterized the metabolic profiles of a panel of 20 human MM cell
lines (HMCL) representative of the molecular heterogeneity found in MM patients. This
panel included 10 commercial HMCL and 10 HMCL derived in our laboratory. First, oxygen
consumption rates (OCR), extracellular acidification rate (ECAR), and spare respiratory
capacity (SRC) of our HMCL panel were assessed in a Mito Stress Assay using a Seahorse
XFe96 analyzer.
Results: Interestingly, the metabolic activities were shown very heterogeneous in
HMCLs with a part of cell lines more dependent of the glycolysis activity and another
part more dependent of the mitochondrial respiration. By integrating the HMCL’s metabolic
profiles with their respective transcriptomic data (RNAseq), we defined a metabolomic
score to classify the HMCL into different groups and represent their glycolysis level
or mitochondrial activity. The score was calculated from the expression of 112 genes
involved in the electron transport chain (Oxphos) or in glycolysis. For a given gene,
the expression values were first log2-transformed and then normalized by subtracting
the average. The score is computed as the difference between the Oxphos and glycolytic
genes’ average expression. Interestingly, high significant correlations between the
HMCL’s functional metabolic profiles and their calculated metabolomic score were identified.
Furthermore, we investigated the potential prognostic value of this gene-based metabolomic
score in the MMRF CoMMpass cohort (newly diagnosed MM patients, n=674) using the maxstat
algorithm, which segregated the cohort into two groups with a significantly different
outcome. We identified that 32% of patients with a high gene-based metabolomic score,
were associated with poor overall survival (P = 3.1x10-6). We then validated this
prognostic value in a second cohort of MM’s patients: the cohort of 206 patients (ArrayExpress
public database under accession number E-MTAB-362). Moreover, we performed correlation
analyses between the metabolic profiles of those HMCL and their respective response
to the conventional treatments (IC50). We found a significant correlation between
a high mitochondrial ATP production and the resistance to proteasome inhibitor (P
= 0.023, n= 12).
Summary/Conclusion: Altogether, we demonstrated that metabolomic deregulation could
participate in drug resistance in MM. Inhibitors targeting metabolic activities may
be of therapeutic interest to overcome drug resistance in MM.
P844: CHARACTERIZATION OF MULTIPLE MYELOMA CELL LINES WITH ACQUIRED-RESISTANCE TO
PROTEASOME INHIBITORS HIGHLIGHTS A LINK BETWEEN RESISTANCE AND METABOLIC DEREGULATION
H. De Boussac1,*, A. Machura1, A. Steer1, A. Kassambara1, M. Gely1, D. Chemlal1, C.
Gourzones2, G. Requirand3, N. Robert3, L. Vincent4, C. Herbaux4, A. Bruyer1, J. Moreaux5
1Diag2Tec; 2UMR 9002 CNRS-UM; 3Laboratory for Monitoring Innovative Therapies, Department
of Biological Hematology; 4CHU Montpellier; 5Institute of Human Genetics, UMR 9002
CNRS-UM, Montpellier, France
Background: Characterized by an abnormal clonal proliferation of malignant plasma
cells, Multiple myeloma (MM) is the second most common hematological malignancy. Novels
agents have significantly improved clinical outcomes, but most of the MM patients
eventually relapse. A better understanding of the drug resistance mechanisms remains
of significant interest to improve the treatment of patients
Aims: We aimed to investigate the mechanisms involved in the resistance to proteasome
inhibitors (PI).
Methods: For that purpose, we have derived and characterized 6 Bortezomib (Btz)-resistant
human myeloma cells lines (BR-HMCLs) from different molecular subgroups and still
dependent on the addition of IL-6 including XG1BR t(11;14), XG2BR t(12;14), XG7BR,
XG20BR, XG24BR t(4;14) and XG19BR t(14;16). These cell lines were cultured continuously
with escalating concentrations of Btz during 12 months.
Results: Interestingly, BR-HMCLs demonstrated significant resistance to Btz compared
to their parental counterparts (CTR-HMCLs) (mean IC50: BR-HMCLs =5nM vs CTR-HMCL=2.3nM,
p<0.05). In addition, these BR-HMCLs also showed significant cross-resistance to Carfilzomib
(Cfz) and Ixazomib (Ixa) PIs (p<0.05 for Cfz or Ixa). Finally, no significant cross-resistance
was observed with other therapeutic agents (melphalan, dexamethasone or IMIDs), indicating
that the observed drug resistance mechanisms are especially related to PIs.
We then used genomic approaches (whole genome sequencing and transcriptomic analyses)
to understand the PIs resistance mechanisms acquired by MM cells. Remarkably, as observed
in relapsed MM patients, among the 40 mutations identified in BR-HMCLs compared to
the CTR-HMCLs, we identified a mutation residing in the Btz-binding pocket of the
proteasome beta5-subunit (PSMB5), that reduces the PI binding capacity, thus preventing
inactivation of the catalytic activity of the 20S proteasome.
Further transcriptomic analyses on BR-HMCLs underlined significant deregulation of
genes involved in cell metabolism and drug clearance that could allow the BR-cells
to maintain metabolic homeostasis and survival in stringent redox conditions. Thus,
in the BR-HMCL we identified an upregulation of enzymes directly involved in glycolytic
and energy metabolism (ALDOC, ENO3, HK1, PDK1/3, PFKB3/4, PFKL, SLC2A1 (FC>1.5 p-value
<0.05), but also a significant downregulation of 8 solute carrier protein (SLC) intake
transporters together with a significant upregulation of xenobiotic receptors (FC>
or < 1.5; p-value <0.05).
We then tested the metabolomic of BR-HMCL using the Seahorse assay. This technology
allows to analyze the glycolysis via the extracellular acidification rate (ECAR) and
cell respiration via the mitochondrial oxidative phosphorylation based on the oxygen
consumption rate (OCR). Following the increased gene expression of several glycolytic
enzymes found in the BR-HMCLs, we observed that while CTR and BR displayed equivalent
respiration and glycolytic activities, their treatment with Btz induces major metabolic
modifications, including a significant increase of the glycolytic and mitochondrial
activity of the BR-HMCL while it decreases it in the CTR-HMCL, suggesting a higher
capacity of the BR-HMCL to respond to cell stress.
Summary/Conclusion: Altogether, we developed acquired PIs resistant HMCLs that exhibit
PSMB5 mutation as observed in patients, and we identified pathways linked to metabolism
deregulation in these cell lines. These results make our PI-resistant models, an attractive
preclinical model to test new therapeutic strategies to overcome PI resistance in
MM.
P845: IDENTIFICATION OF MULTIPLE MYELOMA BIOMARKERS VIA LIQUID BIOPSY BASED ON CELL-FREE
DNA USING A CAPTURE-HYBRIDIZATION PANEL
N. Buenache Cuenda1,*, A. Sánchez-delaCruz1, I. Cuenca Navarro1, L. Rufián Vázquez1,
V. Garrido García1, A. Giménez Sánchez1, R. Alonso Fernández2, J. M. Sánchez Pina2,
Y. Ruiz Heredia1, S. Barrio García1, I. Rapado Martínez1,2, R. Ayala Díaz1,2,3,4,
J. Martínez-López1,2,3,4, J. M. Rosa-Rosa1
1Department of Translational Haematology, Research Institute Hospital 12 de Octubre
(i+12) Haematological Tumors Clinical Research Unit H12O-CNIO; 2Department of Translational
Haematology, Haematology Service, Hospital 12 de Octubre; 3Department of Medicine,
Faculty of Medicine, Complutense University; 4CIBERONC, Madrid, Spain
Background: Multiple myeloma (MM) is a haematological malignancy characterized by
clonal proliferation of pathogenic CD138+ plasma cells (PPCs) in bone marrow (BM).
In recent years, the treatment of MM has shown great evolution. However, most patients
who achieve a complete response eventually relapse. For that, an early detection of
PPCs could be very beneficial for MM patients, and therefore, a sensitive detection
of this stage could allow early therapeutic interventions. Liquid biopsy using cell-free
DNA (cfDNA) as a minimally invasive approach could be an alternative not only for
the diagnosis but also for the detection of recurrences.
Aims: The main objective of this study was the quantification of MM patient-specific
biomarkers in cfDNA compared to PPCs and BM samples.
Methods: Patients with active MM were treated with VRD, autologous stem cell transplantation
and maintenance. We studied gDNA from 23 PPCs from BM aspirates, 16 gDNA from whole
BM and 12 cfDNA from peripheral blood samples obtained at diagnosis. Capture panel
was aimed to detect most common and relevant aberrations known in MM such as coding
regions of 37 genes involved in MM progression and drug resistance, canonical IGH
and IGK to capture IGH rearrangements and regions to cover traditional translocations.
Capture libraries were generated with SureSelect Reagent kits (Agilent Technologies).
We used 50 ng of genomic DNA for both PCCs and BM and for cfDNA a median of 10-200 ng
input. Final libraries were run on an Illumina NextSeq 500 platform. A specific bioinformatics
pipeline was applied: alignment to the hg38 genome, SNVs and Indels identification
by combination of variant callers, and IGH/K rearrangements with MiXCR.
Results: A total of 36 different mutations were identified in the PPCs with frequencies
ranging from 0.011-0.570. Comparison between PPCs and cfDNA showed that 20 out of
23 (87%) mutations identified in the PPCs were observed in the cfDNA, whereas 32 out
of 35 (91%) mutations were observed in BM. Moreover, 6 out of 11 (55%) mutations identified
in the PPCs were observed in cfDNA and BM (Figure 1A). In 5 out of 5 patients at least
1 mutation of those evaluated in PPCs was detected in cfDNA. Interestingly, for the
classical oncogenes (NRAS, KRAS and BRAF), 5 of 8 (63%) hotspot mutations were observed
in cfDNA and 7 of 8 (88%) in BM. Using the ratio between the observed frequency in
cfDNA/BM and that observed in PPCs, we estimated an average tumour burden of 6% in
cfDNA and a mean clonality of ~30% in BM. In addition, 5 out of 5 translocations identified
in PPCs were observed in BM and unexpectedly 1 out of 2 was observed in cfDNA. Regarding
IGH/K rearrangements, 50% of the patients presented at least one rearrangement in
the cfDNA fraction that was identified in the PPCs, and 93% in the BM fraction. Furthermore,
the average reduction in frequency observed in cfDNA and BM was ~12% compared to the
frequency seen in PPCs. Finally, 2 out of 5 patients showed the same IG rearrangement
in the three fractions studied[jm1] (Figure 1B).
Image:
Summary/Conclusion: Most of molecular alterations identified in PPCs seemed to be
present in peripheral blood of patients with MM, surprisingly, even translocation
breakpoints could be seen in cfDNA. However, liquid biopsy monitoring is limited by
signal dilution, as an approximately 1-log reduction in cfDNA was observed compared
to PPCs, which should be considered in bioinformatic analyses. On the other hand,
IGH/K rearrangements could be identified in fewer patients, but the signal reduction
was only ~12%, confirming IG rearrangements as strong biomarkers for MM in cfDNA.
P846: EXPRESSION SIGNATURE OF TP53 BIALLELIC INACTIVATION IDENTIFIES A GROUP OF MULTIPLE
MYELOMA PATIENTS WITHOUT THIS GENETIC CONDITION BUT WITH DISMAL OUTCOME.
C. De Ramón1,*, E. A. Rojas1, I. J. Cardona-Benavides1, M. V. Mateos1, L. A. Corchete1,
N. C. Gutiérrez1
1Hematology, University Hospital of Salamanca, IBSAL, Cancer Research Center-IBMCC
(USAL-CSIC), Salamanca, Spain
Background: Cytogenetic abnormalities remain the most relevant prognostic factors,
especially those related to TP53 gene. Biallelic inactivation of TP53, included in
the definition of double-hit (DH) MM, entails an ominous prognosis, although is present
in less than 5% of newly diagnosed MM (NDMM). However, ultra-high-risk MM, defined
as those patients with a median survival less than 24 months, represents 15-20% of
the MM population. While other high-risk cytogenetic abnormalities may account for
this adverse prognosis, these kind of cytogenetic alterations are not present in all
patients with such an unfavorable outcome. On the other hand, p53 can be deregulated
by other mechanisms different from changes in DNA gene sequence, such as epigenetic
regulation or altered expression of its regulators.
Aims: To define the transcriptional signature of DH-TP53 and to find out if it was
present in other patients who did not have biallelic inactivation of TP53.
Methods: We analyzed RNA-seq, whole-genome and whole-exome sequencing data from 660
newly diagnosed MM (NDMM) patients from the MMRF (Multiple Myeloma Research Foundation)
CoMMpass study to characterize the transcriptional signature of DH-TP53 MM. This gene
signature was used to build a score based on a Spearman correlation coefficient and
a scaled GINI index. Survival data were retrieved from the IA16 release and the analysis
was performed using the Kaplan-Meier estimator and log-rank test. Multivariable Cox
models were fitted in R. GSE4581 and GSE136400 gene expression series from GEO were
used as validation cohorts.
Results:
TP53 biallelic inactivation, defined in this work as DH-TP53 group, was found in 23
out of 660 patients (3.5%) and was associated with an exclusive gene expression signature
consisted of 78 genes. Based on these genes, we calculated the DH-TP53 score, which
identified a subgroup of 50 patients that shared the same transcriptional profile
(DH-TP53-like group). The prognosis of this group was particularly unfavorable with
a median overall survival (OS) of 23 months; and a progression-free survival (PFS)
of 15 months, even worse than that described for DH-TP53 patients (HR = 1.83 [95%
CI, 1.00-3.35], p = 0.046). The prognostic value of the DH-TP53 was confirmed as an
independent prognostic factor for PFS (HR 3.84 [95% CI 2.51-5.88], p < 0.001) and
OS (HR 3.32 [95% CI, 2.31-4.77], p < 0.001) in the multivariable analysis. We also
observed that survival for any of the cytogenetic abnormalities was significantly
shortened in the DH-TP53-like group (p < 0.05). Furthermore, the prognostic value
of the DH-TP53 score was externally validated using gene expression data obtained
by microarray analysis
Image:
Summary/Conclusion: The expression signature of DH-TP53 MM was shared by other MM
patients without TP53 biallelic inactivation (DH-TP53-like group). The DH-TP53 score
identified an ultra-high-risk group of MM patients with a median overall survival
below 24 months. In addition, this score refined the prognostic stratification of
cytogenetic abnormalities, and was externally validated using expression data analyzed
by microarrays.
Funding: This study has been funded by ISCIII (PI16/01074 and PI19/00674) (co-funded
by FEDER), Castilla y Leon (GRS 2058/A/19 and GRS 2331/A/21) and AECC (PROYE20047GUTI).
CDR, EAR and IJCB were supported by the AECC (CLJUN18010DERA), the “Consejería de
Educación de Castilla y León” and the ISCIII (FI20/00226), respectively.
P847: MRD BY MASS SPECTROMETRY IN PERIPHERAL BLOOD AND NEXT GENERATION SEQUENCING
IN BONE MARROW IN A PHASE 2 STUDY OF DARATUMUMAB, CARFILZOMIB, LENALIDOMIDE, AND DEXAMETHASONE
FOR MULTIPLE MYELOMA
B. Derman1,*, J. Rosenblatt2, D. Avigan2, A. Major1, M. Rampurwala1, D. Barnidge3,
A. Stefka1, K. Jiang1, A. Jakubowiak1
1Section of Hematology/Oncology, University of Chicago, Chicago; 2Beth Israel Deaconess
Medical Center, Boston; 3The Binding Site Group, Rochester, MN, United States of America
Background: Measurable residual disease (MRD) as assessed by next generation sequencing
(NGS) using bone marrow (BM) aspirate carries prognostic significance in multiple
myeloma (MM). Though its analytical sensitivity reaches 10-6, MRD assessed from BM
may be vulnerable to false negatives due to patchy or extramedullary disease, or due
to inadequate aspirate sample. Mass spectrometry (MS) is a promising peripheral blood
(PB) assay that may approximate current methods of MRD detection.
Aims: In this phase 2 study evaluating the safety and efficacy of extended daratumumab,
carfilzomib, lenalidomide, and dexamethasone (Dara-KRd) without autologous stem cell
transplant (ASCT) in newly diagnosed MM, we are evaluating the concordance of MRD
by NGS in BM and by MS in PB.
Methods: Forty-one patients have been enrolled from two MM Research Consortium sites
into this phase 2 study (planned enrollment 45 patients). All patients receive 24
cycles of Dara-KRd in 28-day cycles without ASCT. With optional stem cell collection
for ASCT-eligible candidates after cycle 4. MRD by NGS was assessed from BM aspirates
by the clonoSEQ® assay (Adaptive Biotechnologies) with a limit of detection <10-6.
MRD by MS was evaluated using both an automated MALDI-TOF (EXENT) and liquid-chromatography-MS
(LC-MS) by the Binding Site Group (assays under development). Paired PB MS and BM
NGS samples were available for 13 patients at early (post-cycle 4) and 18 patients
at later (cycles 8-24) timepoints. There were 44 paired NGS/EXENT samples (14 post-cycle
4) and 42 paired NGS/LC-MS samples (13 post-cycle 4). Cohen’s kappa test was used
to assess concordance between MS and NGS samples.
Results: For the early post-cycle 4 timepoint, there was low agreement between NGS
(sensitivity threshold 10-6) and MS; there was 46% agreement (Cohen’s kappa -0.18)
between NGS and EXENT and 54% agreement (Cohen’s kappa -0.15) between NGS and LC-MS.
Of the discordant cases for NGS and EXENT, 4/7 (57%) were NGS-/EXENT+. Two of these
4 patients converted to EXENT- at C8. For discordant NGS/LC-MS cases, 5/6 (83%) were
NGS-/LC-MS+. None converted to LC-MS-. The one NGS+/LC-MS- case was from a patient
with kappa light chain MM.
For the later timepoints (cycles 8-24), there was higher concordance between NGS and
MS. There was 63% agreement (Cohen’s kappa 0.27) between NGS and EXENT and 59% agreement
between NGS and LC-MS (Cohen’s kappa 0.13). Of the discordant cases for NGS and EXENT,
4/11 (36%) were NGS-/EXENT+ and 1 of these cases was followed by conversion to EXENT-.
For discordant NGS/LC-MS cases, 9/12 (75%) were NGS-/LC-MS+. None converted to LC-MS-.
Two of the three NGS-/LC-MS+ cases were from a patient with kappa light chain MM.
With median follow-up of 10 months, there have been no progressions or deaths among
these patients. The prognostic significance of persistent NGS+ or LC-MS+ patients
cannot be determined at this time and requires longer follow-up.
Image:
Summary/Conclusion: MRD assessment by MS (EXENT and LC-MS) in PB and NGS in BM serve
complementary roles. Early in treatment, MS positivity may represent false positives
due to immunoglobulin recycling. EXENT from PB appears to more closely approximate
the sensitivity of MRD by NGS at a sensitivity threshold of 10-5, while LC-MS from
PB appears to reach and possibly exceed the sensitivity of MRD by NGS in BM at a sensitivity
threshold of 10-6. The prognostic significance of persistent LC-MS positivity is unclear
and requires longer follow-up.
P848: TINOSTAMUSTINE (EDO-S101), AN ALKYLATOR AND HISTONE DEACETYLASE INHIBITOR, ENHANCES
THE EFFICACY OF DARATUMUMAB IN PRECLINICAL MODELS OF MULTIPLE MYELOMA
A. Diaz-Tejedor1,*, L. San-Segundo1, L. A. Corchete1, L. González-Méndez1, M. Martín-Sánchez1,
M. Lorenzo-Mohamed1, M. González-Rodríguez1, M. Cruz-Hernández1, M. Sánchez-Blázquez1,
N. C. Gutiérrez1, M.-V. Mateos1, M. Garayoa1, E. M. Ocio2, T. Paíno1
1Department of Hematology, Cancer Research Center-IBMCC; University Hospital of Salamanca-IBSAL,
Salamanca; 2Department of Hematology, University Hospital Marques de Valdecilla (IDIVAL);
University of Cantabria, Santander, Spain
Background: Tinostamustine (EDO-S101) is a novel alkylating and deacetylase inhibiting
molecule designed to improve drug access to DNA strands, induce DNA damage and counteract
its repair in cancer cells. It has shown anti-myeloma (MM) activity in different preclinical
models both in monotherapy and in combination with bortezomib.
Aims: To evaluate whether tinostamustine enhances the anti-myeloma effect of daratumumab.
Methods: Tinostamustine was provided by Mundipharma and daratumumab was obtained from
pharmacy surplus of the University Hospital of Salamanca. Mechanisms of action of
daratumumab were studied by flow cytometry (FCM) in MM cell lines pretreated with
tinostamustine and cultured as specified: 1) with F(ab)2 fragments (direct apoptosis
via crosslinking); 2) with 10% human serum (CDC assays); 3) co-cultures of MM and
NK cells (ADCC assays). Expression of CD38 and NKG2D ligands (MICA and MICB) after
tinostamustine treatment was evaluated by FCM, WB and qPCR. The effects of tinostamustine
+ daratumumab combination were also studied ex vivo in bone marrow (BM) samples from
MM patients, and in vivo in two plasmacytoma models developed in CB17-SCID and NK-cell-humanized
NSG mice.
Results: Pretreatment of U266, JJN3, MM.1S, NCI-H929, RPMI-8226 and MOLP-8 cell lines
with tinostamustine (1-2.5 μM) for 48 h increased CD38 expression. Tinostamustine-pretreated
myeloma cells showed a higher level of daratumumab binding, as demonstrated by anti-human-IgG1-AF488
staining. Also, tinostamustine (1-2.5 μM) increased MICA and MICB expression in MM
cell lines. In ex vivo cultures, tinostamustine increased expression of CD38 in 4
out of 10 patients and MICA in 3 out of 5 patients.
Next, the influence of tinostamustine pretreatment (2.5 μM) on several daratumumab
mechanisms was evaluated. In apoptosis via crosslinking, the percentage of apoptotic
cells in presence of daratumumab was higher in tinostamustine-pretreated MOLP-8 cells
vsDMSO-pretreated cells (90.57%±7.35 vs 60.36%±2.86; p<0.001). Likewise, pretreatment
with tinostamustine increased the percentage of Annexin-V+/7AAD+ MOLP-8 cells in CDC
assays with daratumumab, although not reaching statistical significance (93.22%±1.05
vs73.39%±11.93). In MM-NK co-cultures in absence of daratumumab (NK cell-mediated
direct cytotoxicity), the percentages of apoptotic cells in tinostamustine-pretreated
vs DMSO-pretreated cells were: 83.64%±5.28 vs 61.58%±6.81 in MOLP-8 cells (p<0.05);
54.27%±5.99 vs 34.24%±7.37 for RPMI-8226 (p<0.05); and 26.15%±4 vs 12.32%±1.97 in
MM.1S (p<0.05). However, in the presence of daratumumab (ADCC) these percentages showed
a similar tendency although not significant: 88.57%±3.77 vs 78.89%±5.57 in MOLP-8;
70.75%±5.69 vs 58.28%±7.52 for RPMI-8226; and 56.74%±7.25 vs 45.68%±7.1 in MM.1S.
Ex vivo experiments with patients’ BM samples (n = 10) showed that simultaneous combination
of daratumumab + tinostamustine significantly increased the percentage of eliminated
MM cells compared to individual treatments, with an acceptable toxicity on healthy
lymphocytes (Figure 1a).
Finally, in the CB17-SCID model, tinostamustine treatment (24 h) followed by daratumumab
administration controlled tumor growth significantly better than daratumumab alone
(Figure 1b). The combination prolonged the median survival as compared to vehicle,
and monotherapy with tinostamustine or daratumumab (Figure 1b). Similar results were
obtained in the NSG model.
Image:
Summary/Conclusion: Our preclinical data demonstrate that tinostamustine increases
the anti-myeloma effect of daratumumab presumably due to enhanced expression of CD38
and MICA.
P849: DEPLETION OF DIS3 IN MULTIPLE MYELOMA LEADS TO PERTURBATION IN RNA METABOLISM,
CELL CYCLE PROGRESSION AND MITOTIC CHECKPOINT.
V. Favasuli1,*, I. Silvestris1, K. Todoerti2, D. Ronchetti1, S. Erratico3, D. giannandrea4,
Y. Torrente5, R. Chiaramonte4, S. Fabbris1, N. Bolli1, L. Baldini1, A. Neri1, E. Taiana1
1Oncology and Hemato-oncology, University of Milano; 2Phatology and Molecular Oncology,
IRCCS Istituto Nazionale dei Tumori; 3Novystem Spa, Novystem Spa; 4Department of Health
Sciences; 5Stem Cell Laboratory, Unit of Neurology, University of Milano, Milan, Italy
Background: MM is a genetically heterogeneous neoplasm in which the co-occurrency
of multiple genomic events play a crucial role in tumor development, progression and
drug resistance. DIS3 gene maps at 13q22 and encodes for a highly conserved ribonuclease
involved in the RNA processing, quality control and degradation. Deletion of DIS3
gene is a common characteristic of MM patients (approximately 50% of cases); moreover,
DIS3 mutations occur in about 11% of MM cases and are frequently (70%) associated
with loss of the gene. DIS3 mutations along with del(13q) have been recently described
by us as associated with a strong negative effect on clinical outcome.
Aims: The purpose of this study is to shed light on the biological role and impact
of DIS3 transcript in MM.
Methods: LNA gapmeRs were used to silence DIS3 in HMCLs by gymnotic delivery in four
human myeloma cell lines, two of which showing a monoallelic del(13q). qRT-PCR has
been used to evaluate DIS3 silencing efficiency. Cell viability and cellular growth
was evaluated by viable counts and by Cell Titer Glo assay. Modulation of cell cycle
phases distribution was assessed by flow cytometry. Cell cycle synchronization was
obtained using SynchroSet kit. Transcriptional analysis was performed using Affymetrix
microarray. Protein expression was evaluated by Western Blot. Mitotic spindle organization
was analyzed by immunofluorescence (IF).
Results:
DIS3 silenced HMCLs show a significant decrease of cellular growth right after 4 days
from LNA gapmeR delivery. DIS3depleted cells showed a significant perturbation of
cell cycle in all the HMCLs tested. Analysis of synchronized HMCLs revealed a more
pronounced modulation of all cell cycle phases distribution confirming the involvement
of DIS3 in cell cycle regulation. Accordingly, DIS3 silencing lead to an important
modulation of cell cycle checkpoint proteins and mitotic spindle checkpoints. Confocal
microscopy analysis performed in DIS3 silenced HMCLs revealed the presence of mitotic
defects and microtubule organization abnormalities. Transcriptional profile analysis
of DIS3-KD samples is associate to a strong modulation of cell cycle regulation and
mitotic spindle related pathways, together with the expected, strong modulation of
RNA degradation pathway.
Summary/Conclusion: Our data indicate that DIS3 silencing in HMCLs leads to a cell
cycle perturbation accompanied to an increase of mitotic defects. To the best of our
knowledge, this is the first evidence in the context of the pathological role of DIS3
in MM. These results should be considered of interest considering that DIS3 abnormalities
are a common feature of MM patients. Moreover, the transcriptional analyses may contribute
to identify putative DIS3-related pathways and genes (with coding or non-coding potential)
involved in tumorigenic mechanisms in MM and possibly implicated in the therapeutic
settings.
P850: THE HUMORAL AND CELLULAR RESPONSE TO SARS-COV-2 MRNA VACCINATION IN PATIENTS
WITH MULTIPLE MYELOMA AND PRE-MALIGNANT MONOCLONAL GAMMOPATHIES: IMPACT OF OMICRON
VARIANT
P. Storti1,*, V. Marchica1, R. Vescovini1, V. Franceschi1, L. Russo1, V. Raimondi1,
D. Toscani1, J. Burroughs Garcia1, N. T. Iannozzi1, B. Dalla Palma2, M. T. Giaimo1,2,
L. Notarfranchi1, G. Donofrio1, N. Giuliani1,2
1Department of Medicine and Surgery, University of Parma; 2Hematology, “Azienda Ospedaliero-Universitaria”,
Parma, Italy
Background: Multiple myeloma (MM) patients have a high risk of infections with a possible
reduced response to vaccines including anti-SARS-CoV-2 one. Currently, the impact
the emerging new variants in MM patients underwent to anti-SARS-CoV-2 vaccination
is still unknown.
Aims: The humoral and cellular response to SARS-CoV-2 mRNA full vaccination and booster
dose and the impact of new spike-mutated variants, including Omicron have been investigated
in a cohort of MM patients and those with pre-malignant monoclonal gammopathies.
Methods: We enrolled 38 COVID-19-naïve patients: 6 monoclonal gammopathies of undetermined
significance (MGUS), 10 smoldering myeloma (SMM), 9 newly diagnosed MM (NDMM) and
13 relapsed MM (MMR). Peripheral blood (PB) samples were collected before the first
dose and 14±2 days after the second dose (POST) of the mRNA BNT162b2 vaccine. In a
subset of 16 patients with MM (MMD and MMR), PB samples were also collected after
14±2 days of a heterologous booster dose (BOOSTER) with mRNA-1273 vaccine. SARS-CoV-2
spike IgG antibodies (Abs) were measured by the COVID-SeroIndex Kantaro SARS-CoV-2
IgG test. The fraction of neutralizing Abs (NAbs) against SARS-CoV-2 spike of Wuhan-Hu-1
strain and five variants (alpha, beta, gamma, delta and Omicron) was assessed in the
collected sera by a SARS-CoV-2 pseudovirus neutralization assay. Finally, the collected
PB-mononuclear cells were stimulated, with overlapping peptides pools spanning SARS-CoV-2
spike glycoprotein and SARS-CoV-2 spike-specific CD4+ and CD8+ T cells were identified
by intracellular cytokine staining for Interferon-gamma (IFN-γ), interleukin-2 (IL-2),
tumor necrosis factor-alpha (TNF-α), and CD107a+ using flow cytometry assay.
Results: The seropositivity rate for SARS-CoV-2 spike IgG Abs in the total cohort
was 86.8% with 13.2% patients exhibiting no detectable Abs. We found that MMR patients
had significantly lower SARS-CoV-2 spike IgG Abs and NAbs compared with MGUS, SMM
and NDMM patients after full vaccination. All the analyzed variants, remarkably Omicron,
had a significant negative impact on the neutralizing ability of the vaccine-induced
Abs in all patients with MM and also with SMM. We also observed a variable spike-specific
CD4+ and CD8+ T cell responses. We found that both SMM and NDMM patients had significantly
reduced spike-specific IL-2+CD4+ T cell responses compared to MGUS patients. MM patients
showed reduced IFN-γ+ and TNF-α+CD8+ T cells, compared to MGUS. In BOOSTER samples,
MMR patients retained significantly lower anti-spike-IgG-Abs levels compared to MMD
patients. After the booster dose, MMD patients lost the negative impact of the spike
variants seen after full vaccination, due to a significant increase of the NAbs titers.
On the other hand, MMR patients retained NAbs titers against Beta, Delta and Omicron
variants significantly lower than against Whuan-Hu-1 spike. The booster dose increased
SARS-CoV-2 spike-specific T cells in both MMD and MMR reaching the 100% of responder
patients with an increased percentage of IL-2+CD4+ T cells compared to full vaccination.
Summary/Conclusion: Our study underlines the negative impact of Omicron variants on
the neutralizing ability of the vaccine-induced Abs in MM patients as well as in SMM
after a full vaccination. Booster dose immunization improved spike humoral and cellular
responses in MMD patients and in most, but not all, MMR patients suggesting these
patients need to be considered still at risk of Omicron SARS-CoV-2 infection with
a clinically relevant disease.
P851: TARGETING GENE DEPENDENCIES IN MYC OVEREXPRESSING MULTIPLE MYELOMA
L. Hasan Bou Issa1,*, L. Fléchon1, W. Laine1, A. Ouelkdite1, G. Escure2, S. Gaggero1,
T. Ingegnere1, R. Sklavenitis Pistofidis3, I. M. Ghobrial3, T. Facon2, S. Mitra1,
J. Kluza1, B. Quesnel1,2, S. Manier1,2
1Univ Lille, Canther, INSERM UMR-S1277 - CNRS UMR9020; 2CHU Lille, Department of Hematology,
Lille, France; 3Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA,
United States of America
Background: Multiple myeloma (MM) is an incurable hematological malignancy characterized
by a proliferation of clonal plasma cells in bone marrow. MM progresses from precursor
stages, named monoclonal gammopathy of undetermined significance (MGUS) and smoldering
MM (SMM), to the symptomatic myeloma, MM. MYC abnormalities play a critical role in
the disease progression. However, MYC is not therapeutically targetable due to its
nuclear localization and the short half-life of the protein.
Aims: To overcome this, we hypothesized that the proliferative advantage promoted
by MYC overexpression induces differential genomic dependencies on other signaling
pathways thus creates vulnerabilities that can be exploited therapeutically.
Methods: We searched for genetic vulnerabilities associated with MYC overexpression
by leveraging genome-scale pooled short hairpin RNA screening data for 236 cancer
cell lines from Project Achilles. We generated an isogenic model of MYC overexpression
(OE) in U266 cell line using EF1A-C-MYC lentiviral vector. Then, we performed RNA-seq,
quantitative proteomics by Tandem Mass Tag mass spectrometry (TMT-MS) and a drug screening
with ~2000 compounds. For validation, we performed pharmacological inhibition of glutamine
catabolism as well as shRNA-mediated GLS1 knockdown. To determine the functional mechanisms,
we used capillary electrophoresis-mass spectrometry (CE-MS) and Agilent Seahorse XF
analyzer.
Results: Achilles analysis revealed main dependencies associated with MYC overexpression
on glutamine metabolism and specifically GLS1 (glutaminase) and SLC1A1 (glutamine
transporter). Using a screening of 2000 small molecules, we further observed that
inhibitors of NAD synthesis and mTORC1, which rely on the intracellular glutamine
pool, had preferential effect on the proliferation of U266/MYC. Both RNA-seq and quantitative
proteomics showed no significant upregulation of glutaminolysis related genes, suggesting
a non-oncogenic dependency. Our validation tests confirmed this metabolic dependency,
MYC OE cell lines failed to proliferate in the context of glutamine starvation and
showed higher sensitivity to CB-839 and V-9302 inhibiting GLS1 and SLC1A5, respectively.
Using the Seahorse analyzer, we measured the oxygen consumption rate (OCR) induced
by glutamine. U266/MYC cells possess the ability to oxidize glutamine at a higher
rate compared to U266/Ctrl. Additionally, higher sensitivity of U266/MYC to CB-839
was observed on both baseline and FCCP-induced OCR highlighting the role of glutamine
in controlling mitochondrial OXPHOS in MYC OE cells. To understand the differential
metabolic rewiring in MYC OE cells, we performed metabolomic analysis and observed
higher GLS1 activity in U266/MYC presented by elevated glutamine to glutamate flux.
We also identified the enriched metabolic pathways under GLS1 inhibition. Notably,
CB-839 in U266/MYC results in more pronounced changes in TCA cycle and energy debt.
This effect was blunted by the co-incubation with a-ketoglutarate, in a rescue experiment.
Interestingly GLS1 inhibition was not limited to this effect, but extended to redox
balance. CB839 significantly decreased glutathione level in U266/MYC. Furthermore,
the intracellular concentrations of the glutamate-dependent amino acids were more
depleted under GLS1 inhibition in U266/MYC compared to U266/Ctrl.
Summary/Conclusion: Combining these observations, we were able to identify vulnerabilities
to glutamine metabolism in MYC overexpressing cells. These results may lead to developing
new therapeutic strategies to target MYC in clinical practice.
P852: NEOANTIGENS PREDICTED BY THE GNE NEOANTIGEN PIPELINE IN PATIENTS DIAGNOSED WITH
DE NOVO MULTIPLE MYELOMA
Y. Jiang1, C. Henneges2,*, S. Ruppert3, P. Kimes2, A. Amitai2, A. Qamra2, C. Dos Santos3,
J. N. Paulson2, A. Bazeos1
1Product Development, Oncology, Roche Products Limited, Welwyn, United Kingdom; 2Product
Development Data Sciences; 3Oncology Biomarker Development, gRED, Genentech, Inc.,
South San Francisco, United States of America
Background: Neoantigens are tryptic peptides expressed and presented on MHC-I characteristic
for specific tumor mutations. They enable immune surveillance against cancer and may
be potential targets for individualized cancer immuno-therapy.
Aims: Here we present neoantigen evaluations from reprocessing de novo Multiple Myeloma
patients recruited into the CoMMpass study.
Methods: Whole Exome/Genome Sequencing data as well as RNA-sequencing data was provided
by the Multiple Myeloma Research Foundation CoMMpass (SM) Longitudinal Study (research.themmrf.org)
[NCT01454297]. Data was processed with the GNE/Roche neoantigen pipeline applying
Mutect/Strelka for mutation calling, RNAseq and hlahd for HLA haplotyping prior to
predicting peptide binding affinity and rank using NetMHCpan 4.0. Only strong binding
peptides (rank score <0.5) are displayed here and analyzed using the nested-model
approach: the baseline model of primary therapy (PI, IMiD, both) was extended by a
term for presence of individual neoantigens in a gene (prevalence >=5%) for modeling
Progression-free (PFS) and Overall Survival (OS) and Number of Progression Events
during Observation period. Neoantigens are reported if the Likelihood-Ratio-Test was
statistically significant for the Cox PH model or a Poisson regression at the 5% level,
or if the AIC was better than the reference model. We also report the c-Index and
calibration slope, estimated on a predefined training:validation split.
Results: In N=730 patients, we identified nine proteins with high affinity neoantigens
in total (LRT p-value<0.05): MT-CYB, IGHJ5, MAGEC1, IGKV1_5, MT_ND1, IGLV3_1, KRAS,
AC245369.1, and NRAS. All, except NRAS (p=0.055), had a statistically significant
Risk-Ratio when included in the Poisson regression model. Of these, IGHJ5 and IGKV1-5
correlated with PFS; MT-ND1 and KRAS correlated with OS. HR and Risk-Ratios with 95%
CI in Table. Neoantigens occur at different frequencies ranging from 4.5% to 31.1%
in the study population. The most commonly shared neoantigens are from IGLV3-1 (31.1%).
Mapping neoantigens to protein domains using the PFAM database, they are present in
domains related to energy turn-over (PF00032, PF00033), RAS signaling (PF00071) or
in the Ig-like domain (PF07686).
Image:
Summary/Conclusion: We identified nine neoantigens, including two pairs each affecting
PFS and OS, that had an effect on the number of progression event rate. Most were
associated with immuno-globulin chain mutations, four were found as mitochondrial
mutations and or being part of signaling. More data is needed to determine the correlations
of these neoantigens with clinical and cytogenetic risk factors in patients with multiple
myeloma. In addition, further studies are needed to determine factors that affect
the commonality of neoantigens in this population.
P854: HEXABODY-CD38 INDUCES TROGOCYTOSIS AND EFFECTIVELY INDUCES COMPLEMENT-MEDIATED
TUMOR CELL LYSIS AFTER TREATMENT WITH DARATUMUMAB OR ISATUXIMAB IN VITRO
I. H. Hiemstra1,*, K. Santegoets1, M. L. Janmaat1, W. Ten Hagen1, S. Van Dooremalen1,
B. E. De Goeij1, S. Bosgra1, A. K. Sasser2, E. C. Breij1
1Genmab BV, Utrecht, Netherlands; 2Genmab US, Inc, Plainsboro, NJ, United States of
America
Background: HexaBody-CD38 (GEN3014) is a next-generation CD38-specific IgG1 antibody
with a hexamerization-enhancing mutation. HexaBody-CD38 is designed to induce strong
anti-tumor activity in patients with CD38-expressing hematological malignancies through
potent complement-dependent cytotoxicity (CDC) and other Fc-mediated effector functions.
The safety and preliminary efficacy of HexaBody-CD38 are currently being evaluated
in a first-in-human trial in relapsed/refractory multiple myeloma (MM) patients (NCT04824794).
Trogocytosis has been suggested as effector mechanism of daratumumab: reduction of
CD38 expression on tumor and immune cells is thought to reduce local immunosuppression
and contribute to improved adaptive immune responses against MM cells (Krejcik, 2018,
Oncotarget 9, 33621).
Aims: The present study aimed to increase our understanding of the MoA of HexaBody-CD38,
by studying its capacity to induce trogocytosis as well as its capacity to induce
CDC in CD38+ tumor cells in the presence of daratumumab or isatuximab in vitro.
Methods: Trogocytosis was evaluated as the amount of membrane transfer in absence
of transfer of cytoplasm in a flow cytometry-based assay using Wien-133 cells as target
cells and healthy donor monocytes as effector cells. Binding of HexaBody-CD38 to CD38+
cell lines SU-DHL-4, NCI-H929, and Wien-133 that were pre-treated with daratumumab
or isatuximab was allowed for 15 min, 1 h, 4 h or 24 h, while daratumumab or isatuximab
remained present at saturating concentrations. HexaBody-CD38 mediated CDC of daratumumab
or isatuximab-opsonized Wien-133 cells, which were insensitive to CDC induction by
daratumumab or isatuximab, was assessed by flow cytometry at 45 min, 4 h, or 24 h
after adding human complement in the presence of daratumumab or isatuximab.
Results: HexaBody-CD38 induced dose-dependent transfer of plasma membrane from CD38+
tumor cells to human monocytes (n=7). The mean EC50 for trogocytosis activity of HexaBody-CD38
was 8.79 ± 2.49 ng/mL (0.018 ± 0.014 nM). This was in the same range as EC50s of daratumumab
and the HexaBody-CD38 parental antibody without E430G mutation, suggesting that the
hexamerization mutation has limited impact on trogocytosis induction.
HexaBody-CD38 was found to compete with daratumumab and isatuximab for binding to
CD38. Binding of HexaBody-CD38 to CD38+ cells increased in time and with increasing
concentration, generally faster and more extensive in the presence of isatuximab compared
to daratumumab. At equimolar concentrations, near-complete displacement of isatuximab
and daratumumab was observed after incubations ≥1 h and ≥4 h, respectively. Accordingly,
after 24 h HexaBody-CD38 induced comparable maximum CDC of CD38+ cells in presence
or absence of daratumumab. Comparable CDC activity in the presence or absence of isatuximab
was already observed after 4 h.
Summary/Conclusion: HexaBody-CD38 was shown to induce efficient monocyte-mediated
trogocytosis of CD38+ tumor cells in vitro, suggesting HexaBody-CD38 may reduce CD38-associated
immunosuppression in the tumor microenvironment. In addition, it has previously been
reported that the main differentiating effector mechanism activity of HexaBody-CD38
compared to daratumumab and isatuximab is its increased capacity to induce CDC. Here
we confirmed the CDC potency of HexaBody-CD38 even in the presence of saturating concentrations
of daratumumab and isatuximab. This suggests that HexaBody-CD38 is capable of inducing
CDC of CD38+ myeloma cells in patients who have received prior anti-CD38 mAb treatment
with residual daratumumab or isatuximab present in their circulation.
P855: SYNERGISTIC EFFICACY OF COMBINED SUMOYLATION AND PROTEASOME INHIBITION IN MULTIPLE
MYELOMA
U. Keller1,*, G. Heynen1, F. Baumgartner1, M. Heider2, U. Patra3, J. Braune1, P. Mertins4,
S. Müller3, F. Bassermann2, J. Krönke1, M. Wirth1
1Hematology, Oncology and Cancer Immunology, Charite - Universitätsmedizin Berlin,
Berlin; 2Medicine III, Technische Universität München, Munich; 3Biochemistry II, Goethe
Universität Frankfurt, Frankfurt; 4Proteomics Core, Max-Delbrück Center for Molecular
Medicine, Berlin, Germany
Background: Multiple myeloma (MM) is a genetically and clinically heterogeneous neoplastic
plasma cell disorder. Treatment regimens for MM include proteasome inhibitors (PIs)
which are routinely combined with dexamethasone and chemotherapeutics or immunomodulatory
drugs (IMiDs). While such regimens have resulted in significant improvement of disease
control and survival, the development of drug resistance remains a major clinical
problem. Relapsed/refractory MM (r/r MM) is associated with particularly poor prognosis.
New therapeutic strategies to further improve the management of MM and to overcome
drug resistance are therefore urgently needed. SUMOylation is a post-translational
protein modification pathway closely related to ubiquitylation. Hyperactive SUMOylation
(SUMO) signalling is involved in both cancer pathogenesis and cancer progression.
A state of increased SUMOylation has been associated with aggressive cancer biology.
Aims: We sought to identify stress response mechanisms associated with adverse prognosis
and PI resistance. The overall goal was to develop novel therapy combinations targeting
molecular vulnerabilities associated with advanced and/or PI-resistant MM.
Methods: RNA-sequencing, unbiased mass spectrometry-based proteomics and bioinformatics
analyses were used to interrogate the biology and in particular stress response pathways
in MM. Cell culture finding were informed with primary MM patient data from gene expression
studies and and survival data from clinical trials. Novel drug combination treatments
were tested in MM cell-based models in vitro, in MM xenograft models in vivo, and
in primary MM patient samples.
Results: Here, we found that r/r MM is characterized by a state of activated SUMOylation,
a ubiquitin-related post-translational protein modification pathway. High expression
of the SUMO E1 activating enzyme (SAE1/UBA2) was associated with poor overall survival.
Induced resistance to the second generation PI carfilzomib (CFZ) enhanced SUMO pathway
activity. Accordingly, CFZ-pretreated patients showed enhanced SUMO pathway activity
in the MM compartment. Treatment of MM cell lines with subasumstat (also TAK-981),
a novel small-molecule inhibitor targeting the SUMO E1 activating enzyme, showed synergistic
treatment efficacy with CFZ in both PI-sensitive and PI-resistant MM cell lines irrespective
of the TP53 state. Combination therapy was effective in two murine MM xenograft models,
where in vivo growth was significantly inhibited, and in patient-derived primary MM
cells in vitro. Mechanistically, combinatorial treatment of subasumstat and CFZ enhanced
genotoxic and proteotoxic stress and apoptosis.
Summary/Conclusion: Our findings reveal activated SUMOylation as a therapeutic target
in MM and point to combined SUMO/proteasome inhibition as a novel and potent strategy
for the treatment of patients with MM.
P856: A SINGLE-CELL FUNCTIONAL PRECISION MEDICINE LANDSCAPE OF MULTIPLE MYELOMA
K. Kropivsek1,*, P. Kachel2, S. Goetze3,4,5, R. Wegmann1, Y. Severin1, B. D. Hale1,
Y. Festl1, J. Mena1, A. van Drogen3,4,5, N. Dietliker2, J. Tchinda6, B. Wollscheid3,4,5,
M. G. Manz2, B. Snijder1
1Department of Biology, Institute of Molecular Systems Biology, ETH Zurich; 2Department
of Medical Oncology and Hematology, University Hospital Zurich; 3Department of Health
Sciences and Technology, Institute of Translational Medicine; 4Swiss Multi-Omics Center,
PHRT-CPAC, ETH Zurich, Zürich; 5Swiss Institute of Bioinformatics, Lausanne; 6Pediatric
Oncology, Children’s Research Centre, University Children’s Hospital Zurich, Zürich,
Switzerland
Background: Multiple myeloma (MM) is a cancer of plasma cells, defined by complex
genetics and extensive intra- and inter-patient heterogeneity. Despite improved patient
survival driven by a plethora of treatment options, the disease remains incurable.
Molecularly-guided precision medicine to individualize treatment strategies in MM
has had limited success, in part due to the genetic and molecular complexity of the
disease. Functional precision medicine, a complementary approach in which patient
treatment is guided by the ex vivo drug response of patient cells, has not yet been
evaluated for MM systematically.
Aims: Our observational study has two aims: 1) To evaluate single-cell resolved functional
precision medicine (also called Pharmacoscopy) as a way to tailor treatment for individual
MM patients; 2) To identify new treatment options for, and improve our molecular understanding
of, MM.
Methods: In our observational clinical study, image-based ex vivo drug screening (Pharmacoscopy;
PCY) measured the single-cell level responses to drug and immunotherapy combinations
for 138 patient samples from 98 patients covering all stages of the disease. Single-cell
morphologies are analyzed by deep convolutional neural networks, quantifying the cellular
composition and morphological myeloma heterogeneity at time of sampling. PCY data
is integrated with 1) sample-matched proteomics of purified cell subpopulations, 2)
bone marrow serum cytokine profiling, as well as with 3) patient-matched genetics
and clinical data. Lastly, ex vivo drug sensitivities are matched to clinical responses.
Results: Our large-scale single-cell morphological characterization of Multiple Myeloma
samples reveals three reoccurring cell community modes, which we call PhenoGroups
(PGs). These PGs represent different disease stages, with both genetic and inflammatory
associations, independently confirmed by orthogonal data. The ex vivo responses to
61 existing and novel therapeutic strategies (ranging from single drugs to quadruple
drug combinations) reveal considerable differences in patients’ ex vivo responses
to the same treatments. Integration with proteotype data discovers the comprehensive
molecular network underlying variable drug sensitivities, recovering known regulators
of drug response in myeloma and identifying new possible mechanisms of action. Furthermore,
integration with genetics and clinical characteristics across patients recovers known
biomarkers, e.g. proteasome inhibitor resistance for TP53 mutant patient samples,
and proposes novel biomarkers for individualized patient treatments. The strongest
predictor of ex vivo drug sensitivity is the sample composition as captured by the
PhenoGroups, which we find is also reflected in the clinical course. Finally, longitudinal
follow-up and Kaplan-Meier analyses confirm that Pharmacoscopy-measured drug responses
are significantly predictive of the clinical response of patients (Figure 1).
Image:
Summary/Conclusion: Our single-cell functional precision medicine platform (Pharmacoscopy)
combined with patient-matched OMICs provides molecular and clinical insights into
existing and novel treatment strategies for MM patients. Furthermore, Pharmacoscopy
is significantly predictive of clinical response in MM, warranting further clinical
investigation.
P857: BORTEZOMIB- AND CARFILZOMIB-RESISTANT MYELOMA CELLS SHOW INCREASED ACTIVITY
OF ALL THREE ARMS OF UNFOLDED PROTEIN RESPONSE AND ENRICHMENT OF SIRTUIN SIGNALING
PATHWAY
T. Kubicki1,*, K. Bednarek2, M. Kostrzewska-Poczekaj2, M. Łuczak3, Z. Kanduła1, K.
Lewandowski1, L. Gil1, M. Jarmuż-Szymczak2, D. Dytfeld1
1Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical
Sciences; 2Institute of Human Genetics, Polish Academy of Science; 3Institute of Bioorganic
Chemistry, Polish Academy of Science, Poznan, Poland
Background: Proteasome inhibitors (PIs) are among most potent and widely-used class
of drugs in multiple myeloma (MM) treatment. One of the main challenges in MM therapy
is acquired resistance to drugs, PIs are no exemption from this phenomenon. Several
theories were proposed to describe mechanisms responsible for resistance to most commonly
used PIs – bortezomib and carfilzomib. One of the most promising explanations involves
modulation of unfolded protein response (UPR) pathway. The pathway is initiated by
conformational changes in three sensor proteins (PERK, IRE1α, ATF6) in response to
overaccumulation of misfolded proteins. Recent study (Sarin et al, Leukemia 2020)
suggests that MM cell lines used for in vitro experiments differ substantially in
resemblance to patients’ tumors, partially explaining difficulties in translation
of results from laboratory models to MM treatment. Here we evaluated MM1S cells, that
are among best fitted to resemble actual MM patients’ biology.
Aims: The study aimed at describing functional differences between PI-sensitive MM1S
cells (MM1S WT) and their daughter cells, resistant to either bortezomib (MM1S/R BTZ)
or carfilzomib (MM1S/R CFZ), as well as between both resistant cell lines.
Methods: Resistant cell lines were generated by continuous culture with increasing
concentrations of drugs. Subsequently proteomic profiling of sensitive and resistant
cells was performed. To functionally analyze proteomic results proteasome activity
and generation of reactive oxygen species (ROS) were measured. Finally, changes in
UPR activation were assessed by Western blot analysis of key proteins involved in
this pathway.
Results: BTZ- and CFZ-resistant cell lines were successfully generated. IC50 values
were 3-fold higher for the resistant cells (MM1S WT IC50=15.2 nM for BTZ, IC50=8.3 nM
for CFZ; MM1S/R BTZ IC50=44.5 nM; MM1S/R CFZ IC50=23.0 nM). When exposed to different
PI than during resistance generation period MM1S/R BTZ cells were resistant to CFZ
(IC50=43.5 nM) whereas MM1S/R CFZ cells were similarly sensitive to BTZ as MM1S WT
(IC50=24.0 nM). Unsupervised principal component analysis revealed that MM1S/R BTZ
and MM1S/R CFZ differ significantly from MM1S WT cells and also from each other. Canonical
pathway analysis showed similar pathways enriched in both comparisons – MM1S WT vs
MM1S/R CFZ and MM1S WT vs MM1S/R BTZ, however important differences were present in
terms of statistical significances of particular pathways (Figure). Sirtuin signaling
was identified as the most enriched pathway in BTZ-resistant cells (top-4 in CFZ-resistant)
and EIF2 signaling in CFZ-resistant cells. In functional studies, both PIs continued
to block chymotrypsin-like proteasome activity in resistant cells. The relative reduction
in the activity was similar for resistant and sensitive cells – 65% for MM1S/WT in
BTZ, 45% for MM1S/R BTZ, 96% for MM1S/WT in CFZ, 77% for MM1S/R CFZ. Baseline activity
of all 3 catalytical domains of proteasome was significantly higher in the resistant
cells. MM1S/R BTZ cells generated lower amount of ROS in comparison to MM1S WT (64%),
while there were no differences in similar comparison for MM1S/R CFZ cells. Both baseline
and drug-induced activity of UPR was higher in PI-resistant cells then in MM1S WT
and included all three arms of UPR - IRE1α/XBP1s, ATF6 and EIF2α/ATF4 (downstream
effectors of PERK).
Image:
Summary/Conclusion: Contrary to some previous reports, performed mainly on other MM
cell lines, PI-resistant MM1S cells show upregulation of UPR. This reflects heterogeneity
of MM and prompts further studies on UPR role and its interplay with sirtuin signaling.
P858: EPIGENETIC REGULATION OF CD155 ON MULTIPLE MYELOMA CELLS INFLUENCES ANTI-TUMOR
T-CELL CYTOTOXICITY
L. Martinez-Verbo1,*, P. Llinàs-Arias1, L. Villanueva1, C. A. García-Prieto1,2, D.
Piñeyro1, G. Ferrer1,3,4, M. Esteller1,5,6
1Cancer Epigenetics, Josep Carreras Leukaemia Research Institute, Badalona; 2Barcelona
Supercomputing Center, Barcelona, Spain; 3Feinstein Institutes for Medical Research,
Northwell Health, New York, United States of America; 4Centro de investigación Biomédica
en Red Cancer, Madrid; 5Institució Catalana de Recerca i Estudis Avançats; 6Physiological
Sciences Department, School of Medicine and Health Sciences, University of Barcelona,
Barcelona, Spain
Background: DNA methylation remains as one of the key molecular players regulating
gene expression. In multiple myeloma (MM) and thanks to data available, we identified
CD155 as a putative epigenetically regulated gene. Cytotoxic T-cells (CD8+) are crucial
in the clearance of malignant cells and their activation is tightly regulated by immune
checkpoint events (IC). Tumor cells take advantage of this system and escape immune
recognition, avoiding T-cell attacks by inducing exhaustion. CD155 is involved in
the CD8+ T-cell activation IC and it is of interest because could act as an inhibitory
or stimulatory signal. We investigated the role of CD155 in MM and its importance
in T-cell inhibition in-vitro, in relation to its epigenetic state and expression.
Aims: To evaluate the role of CD155 methylation in MM cells and how it affects T-cell
activation and tumor elimination.
Methods: The methylation status of the promoter region of CD155 was studied in several
MM cell lines from COSMIC database and experimentally validated in four (RPMI-8226,
MM.1S, AMO-1 and KMS-12-BM). RPMI-8226 was selected as an unmethylated cell line for
CD155 knock down model (depleted expression) and scramble model by lentiviral transduction.
We analyzed if CD155 has an impact in cell proliferation and cell cycle with MTT assay
and flow cytometry. Co-culture experiments were performed with the CD155 models and
T-cells isolated from healthy donors for 48 hours and studied T-cell cytotoxicity
by analyzing the remaining MM cells with flow cytometry. Finally, we analyzed CD155
expression and survival in MM from CoMMpass project public data.
Results: We validated the in-silico methylation data by bisulfite sequencing and negative
regulation by Western Blot and quantitative PCR in four different cell lines. Depletion
of CD155 did not have a significant impact on cell growth, apoptosis or cell cycle.
For instance, when we co-cultured the CD155 models with pre-activated T-cells, we
detected more cytotoxicity towards CD155 depleted cells, whereas CD155 expressing
cells resisted T-cell activity (p=0.02). In order to determine if the inhibition was
mediated by interaction CD155-TIGIT, we added 10ug/ml of neutralizing αTIGIT and/or
αPD1 antibody to the co-culture systems. While T-cells co-cultured with CD155 depleted
cells in the presence of αTIGIT did not change their levels of cytotoxicity, expressing
cells were eliminated more successfully. In the presence of αPD1, we saw a significant
restoration of T-cell cytotoxicity against both models and antibody combination showed
a synergic effect in expressing models’ co-culture whereas CD155 depleted levels remained
independent of αTIGIT presence. Supporting these results, the data obtained from the
CoMMpass project showed the MM patients (N= 793) with higher expression of CD155 had
shorter overall survival than lower expressing ones (p<0.001).
Summary/Conclusion: In MM cells, the expression of CD155 is regulated by the methylation
status of its promoter region. Unmethylated MM cells expressing CD155 promote T-cell
inhibition and its depletion resulted in an improvement of T-cell cytotoxicity activity
against malignant cells. The addition of anti-TIGIT and anti-PD1 validated that the
CD155 T-cell inhibition is mediated by the interaction with TIGIT, independently of
PD1. These findings indicate that CD155 unmethylated MM cases are at higher risk warranting
further investigation.
P859: ANTI-CD38 NANOBODY JK36 ALLOWS RELIABLE MRD DETECTION IN DARATUMUMAB TREATED
MULTIPLE MYELOMA PATIENTS
E. Meseguer Martinez1,*, J. Marco Buades1, A. García Feria1, P. Ribas García1, M.
J. Fernandez Llavador1, A. López Gabaldon1, S. Broseta Tormos1, E. Francés Aracil1,
O. Cortés Ortega1, E. Donato Martínez1, M. Fernandez Zarzoso1, M. Panero Ruiz1, M.
J. Cejalvo Andújar1, A. Tolosa Muñoz1, M. L. Juan Marco1, D. Ivars Santacreu1, E.
Gómez Beltrán1, M. J. Sayas Lloris1
1Hematología y Hemoterapia, Hospital Universitario Doctor Peset, Valencia, Spain
Background: The introduction of immunotherapies such as daratumumab, a CD38 monoclonal
antibody has improved survival in multiple myeloma (MM), however a few considerations
on minimal residual disease (MRD) assessment by flow cytometry have been raised and
deserve our attention. In flow cytometry CD38 is frequently used as a gating marker
of plasma cells (PCs), but daratumumab can hampering myeloma cell detection. A strategy
to overcome this interference is to staining PCs with anti-CD38 nanobody JK36. Nanobodies
are single variable domain antibody fragments (VHH) that often expose a long complementarity-determining
region 3 (CDR3), feature that allows anti-CD38 nanobody JK36 recognizing a cryptic
epitope not masked by anti-CD38 therapies.
Aims: To compare the median fluorescence intensity (MFI) of CD38 multiepitope (ME)
antibody and anti-CD38 nanobody in bone marrow samples from MM patients not treated
with daratumumab. Secondly, to compare flow cytometric MRD data obtained using CD38-ME
and anti-CD38 nanobody from daratumumab treated MM patients.
Methods: Samples were obtained from 10 MM patients not treated with daratumumab to
compare the MFI of CD38-ME vs CD38-nanobody at baseline and after incubation with
daratumumab at a concentration of 10 nM for 60 min at room temperature. Marrow samples
were stained using panel CD38/CD56/CD19/CD138/CD45 with variable CD38, CD38-ME vs
anti-CD38 nanobody. Data were analyzed using Kaluza 2.1.1 software (Beckman Coulter).
Moreover, 17 bone marrow samples were processed using bulk lysis and subsequently
stained using the MM-MRD EuroFlow 8-color antibody combination panel and tubes 1 and
2 with variable CD38, CD38-ME vs anti-CD38 nanobody. Samples were acquired on CytoFlex
flow cytometer (Beckman Coulter) and data were analyzed using Infinicyt 2.0 software
(Cytognos). Statistical analysis was performed using SPSS version 24, verifying the
assumption of normality of the variable, pairwise comparisons were carried out using
the T-tests for related samples.
Results: The MFI values of CD38 ME on samples from daratumumab not treated patients
was higher than samples after incubation with daratumumab (p=0.003). For CD38 nanobody,
no significant differences were observed for the MFI values of samples, independent
of daratumumab incubation (p=0,139) (figure 1A). The percentage of loss of MFI were
higher in the CD38 ME group (-36,32%) than in the CD38 nanobody group (-0,35%). Regarding
17 patients treated with anti-CD38 therapies, 14 were treated with daratumumab and
3 with isatuximab. Sixteen samples were MRD positive with a sensitivity of 10-5 or
10-6 by both approaches. As shown in figure 1B, CD38-ME and CD38 nanobody both showed
high and comparable MFI on PCs from patients treated with anti-CD38 therapies.
Image:
Summary/Conclusion: The anti-CD38 nanobody JK36 construct has lower coefficient of
variation than CD38 ME antibody. It is a viable alternative to CD38 ME antibody to
gate PCs by flow cytometry allowing reliable MRD detection to identify PCs in the
presence of anti CD38 biologics. Nanobody technology open new avenues in the detection
capability of MRD flow cytometry studies in the era of immunotherapy.
P860: THE DE NOVO DNA METHYLTRANSFERASE DNMT3B PLAYS AN IMPORTANT ROLE IN MM CELL
GROWTH, CLONOGENICITY AND DRUG RESPONSE
C. Muylaert1,*, L. Van Hemelrijck1, P. Vlummens1,2, A. Maes1, K. De Veirman1, E. Menu1,
K. Vanderkerken1, E. De Bruyne1
1Department of Hematology and Immunology-Myeloma Center Brussels, VUB, Brussels; 2Department
of Internal Medicine, Ghent University Hospital, Ghent, Belgium
Background: Multiple myeloma (MM) is an incurable plasma cell malignancy due to the
development of drug resistance (DR). In about half of the MM patients, modifications
are observed in epigenetic modifiers (epiplayers) at the time of diagnosis and this
frequency is further increased at relapse, indicating an important role for epiplayers
in MM cell DR. However, so far, only for two epiplayers, MMSET and EZH2, a clear role
in MM cell DR development has been established. With the aim to identify new, clinically
relevant epiplayers involved in MM progression and relapse, we recently compared the
RNASeq data from matched newly diagnosed and relapsed patients from the MMRF CoMMpass
study. We found that the epiplayer DNMT3B is significantly increased in relapsed patients,
suggesting a possible role for DNMT3B in MM relapse.
Aims: The aim of this study is to explore the role of DNMT3B in MM cell biology and
drug response.
Methods: Publicly available gene expression profiling data of three independent cohorts
of newly diagnosed MM patients (GSE4581, E-MTAB-372, MMRF-COMMPASS), one cohort of
relapsed patients (GSE9782), BM plasma cells (E-MTAB-372), primary MM cells (E- MTAB-372),
and human MM cell lines (HMCLs; E-TABM-1088 and E-TABM-937) was used. DNMT3B specific
targeting was achieved by using the DNMT3B specific inhibitor Nanaomycin A and genetic
inhibition using doxycycline inducible shRNA against DNMT3B. Viability and apoptosis
were assessed using a CellTiter-Glo assay and an AnnexinV/7AAD staining respectively.
In addition, BrdU incorporation and cell cycle analysis was evaluated to assess cell
proliferation. Clonogenic capacity was evaluated by a colony formation assay.
Results: We found that DNMT3B mRNA levels increase during disease progression and
high DNMT3B mRNA expression correlates with a worse disease outcome in both newly
diagnosed and relapsed patients, indicating a role for DNMT3B in MM progression and
DR. In line, Nanaomycin A treatment (up to 72h) led to a significant and dose-dependent
decrease in cell viability and proliferation and increase in apoptosis in the XG-2,
XG-7 and AMO-1 HMCLs. We also validated the anti-MM activity of Nanaomycin A on human
primary MM cells. Since DNMT3B is thought to play a significant role in cancer cell
stemness, we next evaluated the effect on clonogenicity. A significant and dose-dependent
decrease in the number of colonies was observed when low doses of Nanaomycin A were
added to AMO-1 (100 and 200 nM) and XG-2 (30 and 50 nM) cells on the day of plating
(day 0), but not when added 7 days after plating (day 7). In contrast, treatment with
a high Nanaomycin A concentration (800 nM) significantly reduced the number of colonies
at both timepoints, indicating that high Nanaomycin A doses are cytotoxic whereas
low doses impair the proliferation and clonogenicity of the MM cells. The anti-clonogenic
effect of DNMT3B targeting was confirmed by DNMT3B knockdown in AMO-1 cells. Finally,
combining Nanaomycin A (100 nM) with bortezomib (Bz, 4 nM) resulted in a significant
decrease in colony formation compared to either Nanaomycin A or Bz treatment alone.
Summary/Conclusion: Taken together, our findings indicate that DNMT3B is a novel promising
target to overcome or delay relapse in MM. DNMT3B targeting impairs MM cell proliferation
and clonogenicity and sensitizes the cells to the proteasome inhibitor Bz. In the
near future, the anti-myeloma activity of DNMT3B targeting will be validated in vivo
using the 5TMM murine MM models and the underlying mechanisms will be further determined.
P861: SIALOFUCOSYLATED STRUCTURES ENABLE PLATELET BINDING TO MYELOMA CELLS CONFERRING
PROTECTION FROM NK-MEDIATED CYTOTOXICITY
A. Natoni1,*, M. Cerreto1, M. S. De Propris1, M. T. Petrucci1, I. Del Giudice1, S.
Intoppa1, M. L. Milani1, L. Kirkham-McCarthy2, R. Henderson3, D. Swan3, M. O’Dwyer2,
A. Guarini4, R. Foà1
1Hematology, Department of Translational and Precision Medicine, Sapienza University,
Rome, Italy; 2Biomedical Sciences, School of Medicine, National University of Ireland
Galway; 3Department of Haematology, Galway University Hospital, Galway, Ireland; 4Department
of Molecular Medicine, Sapienza University, Rome, Italy
Background: Multiple myeloma (MM) is a plasma cell disorder that develops in the bone
marrow (BM), characterized by an unchecked proliferation and by the ability to disseminate
in different parts of the skeleton. Several studies have highlighted a prominent role
of platelets in the metastatic dissemination of cancer cells. Indeed, by binding to
tumor cells, platelets promote adhesion to target organs and evasion from natural
killer (NK)-mediated cytotoxicity. Importantly, tumor/platelet interactions are mostly
mediated by P-selectin. We have previously shown that MM cells enriched for Sialyl
Lewis X (SLex), a sialofucosylated structure recognized by selectins, efficiently
home into the BM and become resistant to bortezomib in vivo (Natoni et al, 2017).
Aims: We herein hypothesized that sialofucosylation participates in platelet recruitment
on the surface of MM cells via P-selectin, forming a “cloak” that protects tumor cells
from NK-mediated cytotoxicity.
Methods: Platelets were purified from peripheral blood samples obtained from healthy
individuals. MM cell lines and their derivative that have been enriched for SLex,
which strongly bind to selectins, were co-cultured with platelets for 30 min and platelet
binding was assessed by flow cytometry using the CD41/CD61 antibody. In some experiments,
cells were incubated with platelets in the presence of an anti-P-selectin blocker
antibody. To examine the NK-mediated cytotoxicity towards tumor cells, parental and
SLex MM cells were co-cultured with platelets for 24 h and then incubated with autologous
NK cells expanded in vitro for 7 days. NK-mediated cytotoxicity was assessed after
4h by flow cytometry using Annexin V/7-AAD staining. To examine platelet/MM binding
in patients, BM samples from MM patients at different disease stages were analyzed
by flow cytometry using a panel of antibodies to identify platelet binding to malignant
plasma cells.
Results: In MM cell lines, platelets bound exclusively to SLex enriched cells, suggesting
that efficient binding requires sialofucosylation. This binding could be blocked by
a P-selectin antibody, indicating that the interactions between MM cells and platelets
are partially dependent on P-selectin. Platelets significantly decreased NK-mediated
cytotoxicity in the SLex enriched (P=0.0025) but not in the parental MM cells, confirming
the important role of sialofucosylation in platelet binding. Platelets also bound
to malignant cells from primary BM aspirates. Interestingly, the binding to platelets
occurred in SLex positive and negative cells, suggesting that in vivo non-sialofucosylated
ligands may also be involved in platelet binding. Importantly, the SLex CD41/CD61
double positive population seems to accumulate in MM patients at relapse (median diagnosis
vs relapse: 0.86 vs 3.03; P=0.0034).
Summary/Conclusion: We herein report that MM cell lines enriched in SLex interact
with platelets in vitro, and that this interaction partially protects MM cells from
NK-mediated cytotoxicity. These results suggest that in vivo, platelets may promote
immune evasion facilitating the dissemination of MM cells. Moreover, SLex positive
primary malignant BM plasma cells bind platelets, particularly from patients at relapse,
suggesting that in vivo these cells may accumulate as the disease progresses and becomes
resistant to treatment. Finally, the interactions between platelets and MM cells could
be disrupted by a P-selectin antibody, indicating a possible therapeutic target to
restrict metastasis and re-sensitize MM cells to NK cells.
P862: SERUM MASS SPECTROMETRY TO ANALYZE DISEASE RESPONSE IN PATIENTS WITH RELAPSED/REFRACTORY
MULTIPLE MYELOMA RECEIVING ARI0002H, AN ACADEMIC BCMA-DIRECTED CAR T-CELL THERAPY
I. Ortiz De Landazuri1,*, A. Oliver-Caldés2, M. Español-Rego1, M. T. Contreras3, A.
Zabaleta4, C. Agulló3, N. Puig5, V. Cabañas6, V. González-Calle5, R. Jiménez2, S.
Inogés4, P. Rodríguez-Otero4, B. Martin-Antonio7, J. L. Reguera8, A. López-Diaz de
Cerio4, D. Benítez-Ribas1, L. G. Rodríguez-Lobato2, E. A. González1, L. Rosiñol2,
J. Yagüe1, J. M. Moraleda6, Á. Urbano-Ispizua2, M. V. Mateos5, M. Juan1, B. Paiva4,
M. Pascal1, C. Fernández de Larrea2
1Immunology Department; 2Hematology Department, Hospital Clínic de Barcelona, Institut
d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona; 3Clinical Biochemistry
Department, Hospital Universitario de Salamanca, Salamanca; 4Clínica Universidad de
Navarra, Centro de Investigacion Medica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona;
5Hematology Department, Hospital Universitario de Salamanca, Salamanca; 6Hospital
Clinico Universitario Virgen de la Arrixaca., Murcia; 7Instituto de Investigación
Sanitaria, Fundación Jiménez Díaz, Madrid; 8Hospital Universitario Virgen del Rocío,
Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), University of Sevilla, Sevilla,
Spain
Background: ARI0002h is an academic BCMA-directed CAR T-cell that has been reported
as effective in patients (pts) with relapsed/refractory multiple myeloma (RRMM). In
these pts, next generation flow cytometry (NGF) in bone marrow (BM) allows the identification
of deeper responses than serum protein immunofixation (IFE) after treatment.
Aims: Here, we explore EXENT Quantitative Immunoprecipitation Mass Spectrometry (EXENT
QIP-MS) as a highly-sensitive, serum-based, alternative monitoring technique and able
to identify therapeutic monoclonal antibodies (t-mAb) interferences.
Methods: Thirty-three RRMM pts with measurable disease in serum by IFE or free light
chains were included. All of them received ≥2 prior regimens including a proteasome
inhibitor, an immunomodulatory drug and daratumumab, and were refractory to the last
one. These pts were treated with ARI0002h, 30 in the setting of the CARTBCMA-HCB-01
clinical trial (NCT04309981) and 3 as compassionate use. The M-protein (MP) was analyzed
in serum by EXENT QIP-MS using IgG/A/M/κ/λ isotypic beads at three time points: before
ARI0002h infusion, and 28 and 100 days post-infusion. Finally, 98 samples were analyzed
by EXENT QIP-MS. Every patient had received daratumumab previously, but the last infusion
date varied widely. The presence of disease in BM was also investigated by NGF (sensitivity
≥ 2x10-6) at 28 and 100 days post-infusion. The median follow-up of the pts was 15
months (range 5 to 20).
Results: Before ARI0002h infusion, MP could be identified in 33 (100%) pts by EXENT
QIP-MS. After 28 and 100 days post-infusion, MP was also detected by this method in
serum in 24 (72.7%) and 18 (56.3%) pts, respectively.
Investigating the ability of EXENT QIP-MS to identify the patient´s MP, interferences
due to daratumumab and tocilizumab were successfully detected in serum with a IgG-kappa
peak in the mass spectrum of 11693 and 11750 m/z, respectively. Before ARI0002h, daratumumab
was identified in 10 pts (30.3%) and persisted even at day +100 in 5 of them (15.2%).
Tocilizumab was detected at day +28 in 15 (71.4%) pts out of the 21 receiving it,
persisting at day +100 in 3 of them (14.3%).
Then, we compared the results obtained by EXENT QIP-MS and IFE at the two time-points
analyzed (n=63). Results were concordant in 81% of samples and discordant in 19% (p<0.0001).
All discordances were due to EXENT QIP-MS(+)/IFE(-) results, except in one case. In
this patient, the serum was mistakenly considered as IFE(+) (IgG-kappa) owing to the
interference of daratumumab. Discordant results were equally noted at day +28 and
+100 (Fig.1).
From 25 and 26 MRD-evaluable pts at day +28 and +100, 96% and 92% were MRD(-) in BM
by NGF, respectively. NGF was discordant in all cases (EXENT QIP-MS(+)/NGF(-)), except
in 1 patient at +28 days and 3 pts at +100 days, reflecting the MP persistence in
the absence of BM disease. After 16 months, 75% of pts with EXENT QIP-MS(-)/NGF(-)/IFE(-)
at day +28 were alive and without progression, and only 2 pts have relapsed from the
disease, both after 12 months (14 and 16 months, respectively).
Image:
Summary/Conclusion: In this study including pts with RRMM and measurable disease,
serum EXENT QIP-MS allowed the identification of the MP in all cases with high sensitivity.
EXENT QIP-MS was also able to differentiate between the MP and t-mAb, which translated
in a correct labeling of treatment response. As compared to both IFE in serum and
NGF in BM, EXENT QIP-MS was able to identify residual disease in a higher proportion
of cases.
P863: GENERATION OF A FIRST-IN-CLASS INHIBITOR FOR THE MASTER ONCOREGULATOR HNRNP
K IN MULTIPLE MYELOMA
Á. Otero Sobrino1,*, J. Le Coq2, P. Aguilar-Garrido1, M. Á. Navarro Aguadero1, M.
I. Albarrán3, J. Klett3, C. Blanco3, R. Fernández-Leiro4, J. Martínez-López5, M. Velasco1
1H12O-CNIO Haematological Malignancies Clinical Research Unit; 2Electron Microscopy
Unit; 3Biology Section; 4Genome Integrity and Structural Biology Group, Spanish National
Cancer Research Centre (CNIO); 5Grupo de Hematología Traslacional, Hospital 12 de
Octubre de Madrid, Madrid, Spain
Background: Haematological malignancies constitute a plethora of different neoplasms
including leukaemia, lymphomas and myeloma. Despite all of them having robust first-line
treatments and a myriad of therapeutic alternatives, some remain incurable due to
the resistance and refractoriness of tumour cells. The heterogeneous nuclear ribonucleoprotein
K, hnRNP K, is a master oncoregulator that binds C-rich tracks of nucleic acids and
is implicated in multiple biological functions such as splicing, polyadenylation and
translation. Indeed, hnRNP K contributes to treatment resistance and poor outcomes
in haematological malignancies. Compared to other hnRNPs, hnRNP K has the unique structural
characteristic of containing three K-homology (KH) domains that allows it to bind
both ssDNA and RNA sequences, as well as a K-interactive (KI) region that binds to
multiple critical proteins such as Src, Fyn or Lyn amongst others. Interestingly,
hnRNP K regulates the p53/p21 and c-Myc pathways, and our group has characterised
hnRNP K as a driver of leukaemia so far, thus proving its role in haematological neoplasms.
Aims: We aim to develop a first-in-class inhibitor for hnRNP K and test its efficacy
in circumventing resistance of haematological malignancies.
Methods: We have produced full-length (FL) hnRNP K protein in-house and verified that
it was properly folded and could bind RUNX1 RNA and mRNA sequence-based ssDNA, using
Tycho and fluorescence anisotropy technologies. We then carried out a high-throughput
screen of over 7,000 small molecules (both FDA and non-FDA approved) using AlphaLISA
technology and validated both chemically and biologically the inhibition of hnRNP
K with those identified compounds. Lastly, we have performed phenotypical analysis
of the effects of the hnRNP K inhibitors in Multiple Myeloma cell line AMO1 and L363
overexpressing hnRNP K through CRISPR synergistic activation mediator (SAM).
Results: We have successfully produced and purified the FL hnRNP K protein, with stable
inflexion temperatures (Ti) as measured by Tycho. Furthermore, we corroborated that
it maintained its ssDNA and RNA binding abilities through fluorescence anisotropy.
Lastly, we set up the AlphaLISA system to screen a library of over 7,000 small molecules
and identified circa 10 interesting hits, from which we validated the 5 best based
on their inhibition capacity and specificity of hnRNP K. We verified the chemical
properties of these small molecules. Lastly, we confirmed that they had a biological
effect of decreasing the levels of hnRNP K downstream pathways as well as inducing
changes in viability dose-response curves, using Multiple Myeloma cells (AMO1 and
L363) genetically modified with CRISPR/SAM technology to overexpress hnRNP K. Likewise,
we investigated the absence of such biological effect in hnRNP K-Knock Out (KO) AMO1
and L363 cells, obtained by CRISPR-Cas9.
Image:
Summary/Conclusion: We have identified a first-in-class hnRNP K inhibitor that specifically
binds and blocks hnRNP K in modified Multiple Myeloma cell lines. This constitutes
a novel and promising therapeutic approach that could help overcome the current drug
resistance in haematological neoplasms currently found in the clinic.
This work was financially supported by CRIS contra el Cancer Association (NGO) AES
ISCIII (PI18/00295), ISCIII Miguel Servet (CP19/00140) and IF-Marie Sklodowska Curie
Actions grant (MAtChinG – 101027864).
P864: CTPS1 IS A NOVEL THERAPEUTIC TARGET IN MYELOMA - SELECTIVE SMALL MOLECULE INHIBITION
DELIVERS SINGLE AGENT ACTIVITY AND SYNERGISES WITH ATR INHIBITION
C. Pfeiffer1,*, P. Beer2, H. Asnagli2, A. Bolomsky3, A. Grandits4,5, A. Schneller1,
J. Huber1, N. Zojer1, M. Schreder1, A. Parker2, H. Ludwig1
1Department of Medicine I, Klinik Ottakring, Wilhelminen Cancer Research Institute,
Vienna, Austria; 2Step Pharma, Saint-Genis-Pouilly, France; 3National Cancer Institute,
Center for Cancer Research, Lymphoid Malignancies Branch, National Institutes of Health,
Bethesda, United States of America; 4Department of Medicine I, Division of Oncology,
Medical University of Vienna; 5Comprehensive Cancer Center, Vienna, Austria
Background: Recent years have seen major improvements in the treatment of myeloma;
however, relapse occurs in the majority of patients. Novel treatment approaches are
moving away from cytotoxic chemotherapy towards targeted therapies with defined mechanisms
of action. Malignant cells have an increased demand for key cellular components and
a reliance on de novo synthesis pathways. CTP synthetase 1 (CTPS1) plays a pivotal
role in pyrimidine production, by catalysing the rate limiting step in the de novo
synthesis of CTP, which is required for DNA, RNA and phospholipids. Human genetic
studies have identified an essential and non-redundant role for CTPS1 in lymphoid
cell proliferation which is complemented by the homologous CTPS2 isoform outside the
haemopoietic system.
Aims: To evaluate the role of CTPS1 as a novel target in myeloma, elucidate the molecular
consequences of CTPS1 inhibition and test rationally designed combination therapy.
Methods: STP938 is a potent small molecule CTPS1 inhibitor with >1,300-fold selectivity
for CTPS1 over CTPS2. The effects of STP938 on cell proliferation (metabolic activity,
tetrazolium indicator), apoptosis (annexin V) and cell cycle (propidium iodide) were
assessed in vitro using 12 myeloma cell lines. The role of CTPS1 was further assessed
in CRISPR knock-out (KO) experiments. Activation of proteins in the replication stress
and DNA damage response pathways was analysed by western blotting. Additive/synergistic
effects of combining STP938 with two different ATR inhibitors were assessed on cell
line proliferation and enumerated by Bliss score (Synergyfinder 2.0).
Results: STP938 showed single agent activity against 6 of 12 cell lines tested, with
IC50 values for sensitive lines ranging from 19 to 128 nM (Figure A). STP938 showed
cytotoxic activity against sensitive lines, evidenced by induction of apoptosis. KO
experiments confirmed that MM cell lines depend on CTPS1 for proliferation. Exposure
to STP938 was associated with accumulation of cells in S phase, induction of a replication
stress response as evidenced by activation (phosphorylation) of CHEK1, and induction
of the DNA damage response pathway as evidenced by activation of CHEK2 (albeit to
a lesser extent than activation of CHEK1). Importantly, S phase accumulation, induction
of replication stress and induction of DNA damage response was observed in cell lines
where STP938 did not produce significant anti-proliferative activity (Figure B). Given
the ability of STP938 to activate CHEK1, STP938 was tested in combination with an
ATR inhibitor, as ATR is the main upstream activator of CHEK1. The ATR inhibitor ceralasertib
demonstrated additive or synergistic activity when combined with STP938 (Figure C).
Notably, synergy between STP938 and ATR inhibition, along with induction of apoptosis,
was observed in cell lines resistant to single agent STP938. Results were confirmed
with a second ATR inhibitor (VE-821, Figure C).
Image:
Summary/Conclusion: Inhibition of CTPS1 by single agent STP938 showed anti-proliferative
activity against 6 of 12 myeloma cell lines, and induced replication stress in all
cell lines. Combined STP938 and ATR inhibition showed anti-proliferative activity
in all cell lines, including those resistant to single agent STP938. These data suggest
a model whereby myeloma cells with high background replication stress are sensitive
to killing by single agent STP938, whereas cells with lower background replication
stress are sensitised by STP938 to cell death induced by ATR inhibition (Figure D).
STP938 will shortly enter clinical development for patients with late stage lymphoid
neoplasms.
P865: BIOMARKER ANALYSIS TO SUPPORT DOSE OPTIMIZATION OF IBERDOMIDE AS MONOTHERAPY
AND IN COMBINATION WITH STANDARD OF CARE TREATMENTS FOR MULTIPLE MYELOMA FROM A PHASE
1/2 TRIAL
M. Amatangelo1,*, Y. Cheng1, W. Pierceall1, N. W. van de Donk2, S. Lonial3, M. Wang1,
J. Emerson1, K. Hong1, P. Maciag1, T. Peluso4, A. Gandhi1, A. Thakurta1
1Bristol Myers Squibb, Princeton, NJ, United States of America; 2Amsterdam University
Medical Center, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center
Amsterdam, Amsterdam, Netherlands; 3Winship Cancer Institute, Emory University, Atlanta,
GA, United States of America; 4Celgene International Sàrl, a Bristol-Myers Squibb
Company, Boudry, Switzerland
Background: Iberdomide (IBER), a novel cereblon E3 ligase modulator (CELMoD®) compound,
induces ubiquitination and proteasome-dependent degradation of Ikaros and Aiolos proteins,
resulting in tumoricidal and immunomodulatory activity in multiple myeloma (MM). IBER
is being investigated for the treatment of relapsed/refractory MM (RRMM) in a phase
1/2 study (NCT02773030). Due to the recent focus by health authorities on dose selection
for oncology products other than the maximum tolerated dose (MTD), a comprehensive
analysis of pharmacodynamics (PD) and pharmacokinetics (PK) was implemented to support
dose optimization of IBER.
Aims: To inform dose selection of IBER as monotherapy, and in combination with dexamethasone
(DEX; Iber+d) and with DEX and daratumumab (DARA; IberDd).
Methods: PK samples were collected on treatment of cycles (C) 1–4 to estimate IBER
exposure using population PK (area under the concentration curve over 24-h dosing
interval [AUCτ]). Biomarkers included analysis of peripheral blood samples on C1 day
(D)1 and mid-cycle through C4 for immunophenotyping, absolute neutrophil counts (ANC),
and assessment of serum free light chain (sFLC), as a biomarker of tumor burden. Bone
marrow samples were collected at screening and C2D15 for immunohistochemistry.
Results: IBER AUCτ increased in a dose-proportional manner between 0.3 and 1.0 mg
as monotherapy and 0.3 and 1.6 mg in combination with DEX, with moderate variability.
Median time to maximum serum concentration of IBER was 2–4 h post dose with an elimination
half-life of ~12 h. Comparable IBER exposure was observed between monotherapy and
Iber+d. Reductions in Ikaros/Aiolos protein levels in tumor cells were observed at
all dose levels in both cohorts with a >90% reduction at the 0.45-mg dose. However,
a >50% decrease in sFLCs was observed only at doses ≥0.9 mg, and doses of 1.6 mg induced
faster and deeper decreases vs lower doses. In the immune compartment, IBER treatment
induced similar dose-/exposure-dependent PD changes as monotherapy and Iber+d, which
appeared to saturate at higher doses. A >80% reduction in mature B cells and ~2-fold
increase in proliferating/activated T and NK cells were observed at doses ≥1.0 mg.
PD activity of IBER was not attenuated by prior refractoriness to immunomodulatory
drugs (IMiD® agents) or anti-CD38 therapy. Based on these results, doses between 1.0
and 1.6 mg were tested in the IberDd cohort. PK and PD of IBER were similar with concomitant
administration of DARA. In this cohort, a reduction in mature B cells and sFLCs was
more consistently observed at 1.6 mg vs lower doses; however, higher IBER exposures
were associated with more pronounced decreases in ANC when compared with Iber+d. This
was clinically manageable and the MTD of IBER was not determined in any cohort.
Summary/Conclusion: Similar PK and PD results were observed in patients treated with
IBER monotherapy, Iber+d, and IberDd. Across all cohorts, a >20% difference in dose
was needed for meaningful change in IBER exposure, immune PD began to saturate at
doses ≥1.0 mg, and decreases in tumor burden were greatest at the 1.6-mg dose. Based
on these results, IBER doses of 1.0, 1.3, and 1.6 mg were chosen for dose optimization
of IberDd for RRMM. Taken together with data suggesting immune surveillance is a driver
of maintenance therapy efficacy and a desire to increase IBER tolerability with long-term
treatment, doses of 0.75, 1.0, and 1.3 mg were selected for dose optimization of IBER
monotherapy in the newly diagnosed MM maintenance setting.
P866: CORRELATIVE ANALYSIS TO DEFINE PATIENT PROFILES ASSOCIATED WITH MANUFACTURING
AND CLINICAL ENDPOINTS IN RELAPSED/REFRACTORY MULTIPLE MYELOMA PATIENTS TREATED WITH
IDECABTAGENE VICLEUCEL (BB2121)
J. Rytlewski1,*, J. Fuller1, D. R. Mertz1, C. Freeman2, S. Manier3, N. Shah4, T. B.
Campbell1
1Bristol Myers Squibb, Princeton, NJ; 2Moffitt Cancer Center, Tampa, FL, United States
of America; 3CHU de Lille, University of Lille, Lille, France; 4University of California
San Francisco, San Francisco, CA, United States of America
Background: The anti-B-cell maturation antigen (BCMA) autologous chimeric antigen
receptor (CAR) T cell therapy idecabtagene vicleucel (ide-cel; bb2121) has achieved
deep and durable responses in relapsed/refractory multiple myeloma (RRMM) patients
(Munshi, N Engl J Med 2021). Prior studies have described the correlation between
patient characteristics and clinical outcomes from CD19 CAR T cell therapy (Finney,
J Clin Invest 2019; Fraietta, Nat Med 2018).
Aims: We sought to define patient profiles correlated with manufacturing and clinical
endpoints in RRMM patients treated with ide-cel in clinical trials using unsupervised
clustering and multivariate machine learning across multiple key variable domains.
Methods: Clinical and manufacturing data were harmonized across 164 RRMM patients
treated with ide-cel in KarMMa (NCT03361748) and KarMMa-2 cohort 1 (NCT03601078).
Based on individual multivariate importance, 10 selected peripheral blood mononuclear
cell (PBMC), drug product (DP), and in-process cell growth variables were used to
define patient clusters. Random forest classifiers were generated to identify patient
characteristics associated with manufacturing and clinical endpoints. Wilcoxon rank
sum and Kruskal-Wallis tests were used to compare 2 and 3+ categorical groups; Cox
proportional hazards were used to compare groups with time-to-event data.
Results: Using an unsupervised method, 4 patient clusters were identified that represented
10%, 57%, 15%, and 18%, respectively, of the total 164 patients. Cluster 4 was the
most favorable and was characterized by a higher frequency of T cells in PBMCs; increased
T-cell size during manufacturing; higher DP T-cell transduction, potency, and vector
copy number; and ultimately a > 3-fold higher CAR T cell yield compared with the least
favorable cluster 1. Cluster 2 contained most patients and was associated with intermediate
manufacturing endpoints. Patients in cluster 4 had a higher complete response rate,
longer progression-free survival, and were defined by lower tumor burden, higher absolute
lymphocyte count (ALC), and longer washout period after alkylator treatment, among
others (Table).
Image:
Summary/Conclusion: The current study identified patient profiles in RRMM using accessible
laboratory or medical history data that correlated with longitudinal outcomes. These
findings may inform patients likely to achieve improved outcomes with CAR T cell therapy.
P867: TUMOR PROFILING OF IDECABTAGENE VICLEUCEL (IDE-CEL; BB2121) PATIENTS IN KARMMA
SHOWED COMPARABLE RESPONSES IN EXISTING MOLECULAR HIGH-RISK SUBSETS AND PRELIMINARY
GENE SIGNATURE OF DURABLE RESPONSE
N. Martin1,*, A. Xu**1, N. Stong**1, J. Rytlewski1, O. Finney2, T. Campbell1, W. Pierceall1,
E. Flynt1, E. Thompson1, S. Kaiser1
1Bristol Myers Squibb, Princeton, NJ; 22seventy bio, Boston, MA, United States of
America
Background: Multiple myeloma (MM) tumors exhibit increasing prevalence of high-risk/resistance
(HR) features with each successive relapse, leading to poorer outcomes in late-line
patients (pts). This may arise primarily as malignancy heterogeneity increases, and
with selective pressure from successive treatment regimens. Idecabtagene vicleucel
(ide-cel; bb2121) was the first approved anti-BCMA CAR T therapy in late-line relapsed/refractory
MM (RRMM), and CAR Ts represent a novel mechanism of action (MOA) in the MM treatment
landscape. In the KarMMa study (NCT03361748), high incidences of overall responses
(OR) to ide-cel were observed, including in the pt subgroup with HR cytogenetic features.
Aims: Assess baseline multi-omics molecular profiles in KarMMa pt tumors to characterize
known and novel molecular features and ide-cel outcomes. Evaluate post–ide-cel tumor
composition by analyzing paired pt specimens (pretreatment and at relapse).
Methods: Bone marrow aspirates were collected from KarMMa pts at pretreatment (n=97
RNA, n=68 DNA) and progression (n=64 RNA, n=33 DNA). RNA sequencing and whole genome
sequencing was performed on CD138+ cells. Known molecular HR genomic (biallelic p53
inactivation, high cancer clonal fraction del17p, HR t(4,14), and cereblon mutation)
and transcriptomic (MDMS8 gene signature) features were analyzed, and correlations
with OR and progression-free survival (PFS) evaluated. BCMA mutation and copy number
variation were evaluated. Differential gene expression and principal component analyses
explored novel transcriptomic signatures and response.
Results: Molecular HR features were identified in 44% (43/97) of pts at pretreatment
and 48% (31/64) at progression. Some tumors had multiple HR features consistent with
heterogeneity in late-line RRMM. Paired pre/post–CAR T samples did not show dominant
enrichment for ≥ 1 HR features at progression. OR and PFS were similar in those with
vs. without each molecular HR feature analyzed. One principal component (PC4) was
correlated with PFS (p=0.002). The top weighted genes in PC4 may be a novel signature
associated with durable ide-cel responses, and exploratory analyses of this signature
are ongoing. Loss of one copy of BCMA was observed in 4% (3/68) and 12% (4/33) of
pts at pretreatment and progression, respectively. Biallelic loss of BCMA at progression
was observed in 6% (2/33) of pts, one of whom had single copy number loss pretreatment.
Summary/Conclusion: Baseline multi-omic–based tumor HR features were not associated
with OR. This finding suggests ide-cel activation and expansion remains a critical
axis for OR that may not be substantially influenced by tumor intrinsic features.
Biallelic loss of BCMA was not observed pretreatment and infrequently at progression,
consistent with previous reports. Baseline HR features did not correlate with PFS,
and a dominant selection for any one HR feature at progression was not noted. These
observations were consistent with previous reports in non-molecularly defined HR subgroups
from KarMMa and the hypothesis that CAR T MOA may have a broader spectrum of clinical
activity across more diverse molecular subtypes of pts, especially existing HR subsets;
this could be a consideration as CAR Ts are developed in earlier lines of therapy.
A transcriptional signature was associated with more durable ide-cel responses, which
we postulate may outline a distinct suboptimal pretreatment feature in an ide-cel
context; further exploration of this preliminary signal is ongoing.
**Authors contributed equally to this abstract.
P868: A CIRCULATING SERUM MIRNAS-BASED MODEL TO PREDICT EARLY MORTALITY IN MULTIPLE
MYELOMA PATIENTS TREATED WITH BORTEZOMIB-BASED REGIMENS.
A. Puła1,2,*, P. Robak2,3, D. Jarych4, D. Mikulski2,5, I. Dróżdż6, J. Szemraj7, T.
Robak1,2
1Department of Hematolgy, Medical University of Lodz; 2Copernicus Memorial Hospital;
3Department of Experimental Hematology, Medical University of Lodz; 4Laboratory of
Virology, Institute of Medical Biology, Polish Academy of Sciences; 5Department of
Biostatistics and Translational Medicine; 6Department of Clinical Genetics; 7Department
of Medical Biochemistry, Medical University of Lodz, Lodz, Poland
Background: Multiple myeloma (MM) is a hematological malignancy characterized by the
clonal proliferation of plasma cells in the bone marrow. Despite the progress made
in the treatment of MM, some patients die within the first year of diagnosis. Numerous
studies underline the role of miRNAs in the pathogenesis of MM and their potential
role in prognosis.
Aims: The aim of this study was to analyze the expression of selected miRNAs in the
serum of MM patients treated with bortezomib-based regimens and determine their potential
to predict early mortality.
Methods: The study was conducted in prospectively-recruited patients with newly-diagnosed
MM who were qualified for bortezomib-based treatment regimens. All were admitted to
the Department of Hematology, Medical University of Lodz between 2017 and 2021. Venous
blood samples were collected from the patients before treatment initiation, processed,
and stored for further use. Total RNA, including miRNA, was isolated from serum, and
reverse-transcribed to cDNA. The expression of 31 selected miRNAs was determined using
a miRCURY LNA miRNA Custom PCR Panel. The miRNA selection was based on the results
of our previous study (Robak et al., Cancers Cancers (Basel) 2020; 12(9): 2569). Three
miRNAs were used for expression normalization: hsa-miR-23b-3p, hsa-miR-151a-5p and
hsa-miR-152-3p. Clinical data, including patient characteristics on diagnosis, treatment
regimen, response to treatment and follow-up were obtained from hospital records.
Differential expression analysis, univariate and multivariate logistic regression
models were performed using R version 3.6.3.
Results: A total of 69 MM patients were included in the study with a mean age at diagnosis
of 64.9 ± 11.0 years. Among them, 17 patients experienced death within 12 months of
diagnosis. Patients with early mortality were significantly older (72.6 vs. 62.3 years,
p=0.0005), and more frequently men (70.6% vs. 42.3%, p=0.0429). No differences were
observed in R-ISS distribution or CRAB symptoms between the groups, nor were any significant
differences between percentage myeloma infiltration in the bone marrow and various
laboratory findings, including LDH, serum M protein, albumin, CRP, and uric acid.
In the differential expression analysis, two miRNAs were significantly downregulated
in early mortality group- hsa-miR-328-3p (fold change- FC: 0.72, p=0.0342) and hsa-miR-409-3p
(FC: 0.49, p=0.0357). Similarly, hsa-miR-328-3p (OR 0.44, 95%CI: 0.20-0.97, p=0.0415)
and hsa-miR-409-3p (OR 0.69, 95%CI: 0.48-0.98, p=0.0400) were found to have a protective
effect against early mortality in univariate analyses. The multivariate logistic regression
analysis found that miRNAs retained their significance when established clinical prognostic
factors were included. The final model consisted of hsa-miR-409-3p (OR 0.61, 95%CI:
0.37-0.99, p=0.0480), hsa-miR-328-3p (OR 0.33, 95%CI: 0.13-0.87, p=0.0254), age (OR
1.13, 95%CI: 1.03-1.23, p=0.0096) and R-ISS 3 (OR 2.91, 95%CI: 0.63-13.47, p=0.1723).
A receiver operating characteristics (ROC) analysis for the model yielded an area
under the curve (AUC) of 0.863 (95%CI: 0.761-0.965). At the optimal cut-off value
of 0.28, the sensitivity and specificity reached 88.2% and 77.5%, respectively.
Image:
Summary/Conclusion: hsa-miR-409-3p and hsa-miR-328-3p are independent factors related
to early mortality in MM patients. Our results were used to generate a multiple regression
model that may have the potential to predict early mortality. Further external validation
of our model is necessary to confirm its clinical value.
P869: PRODUCTION BY GUT MICROBIOTA AS PROGNOSTIC BIOMARKER IN MM
A. Rodríguez-García1,*, R. Garcia-Vicente1, M. L. Morales1, R. Gómez-Gordo2, P. Justo1,
C. Cuéllar1, J. Sánchez-Pina1, D. Gómez-Garre1, J. Martínez-López1, M. Linares3
1Department of Hematology, Hospital Universitario 12 de Octubre, Hematological Malignancies
Clinical Research Unit H120-CNIO; 2Microbiota and Vascular Biology Laboratory, Hospital
Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC); 3Department
of Biochemistry and Molecular Biology, Universidad Complutense de Madrid, Madrid,
Spain
Background: There is increasing evidence compelling that the microbiota has a strong
impact on cancer. The presence of these microorganisms and their activity could have
an impact on immune cells and the bone marrow, playing an important role in the progression
of multiple myeloma (MM). The short-chain fatty acids (SCFAs) metabolites from gut
microbiota have been linked to beneficial effect in the response of MM to treatment.
Aims: In this work we explore whether microorganisms and metabolites of the gut microbiota
are imbalanced in monoclonal gammopathies and their possible role in the development
of MM.
Methods: We collected 51 patients with monoclonal gammopathy of undetermined significance
(MGUS) (n = 12), smoldering (SMO) (n=7) and MM at diagnosis (MMdx) (n = 11), at relapse
(MMrf) (n=7) and at complete remission (CR) (n = 7, 3 of them paired with their diagnostic
stage). Furthermore, healthy patients (n = 8) were included as controls (C). After
the extraction of the microbial DNA from fecal samples, the ribosomal subunit of the
bacterial 16S gene was sequenced on the Illumina® MiSeq ™ and bioinformatic analysis
and quantification of absolute abundance were carried out. The analysis of metabolites
released by the gut bacteria was performed by the quantification of SCFAs acetate,
butyrate and propionate on serum samples by MRM (LC-QQQ-MS). The effect of SCFAs on
the proliferation of human MM tumor cell lines was evaluated using standard MTT assays.
Results: Patients with MMrf showed the lowest estimated richness with significant
differences in Alpha diversity measured by Pielou’s index (Fig 1A). Beta diversity
clustered separately and was significant in patients with MM at complete remission
compared to MMdx in paired samples (Fig. 1B). Then, we compare the taxonomic composition
of the gut microbiota of patients at different stages of the disease, and we found
significant differences in the relative abundance of some microorganisms. On one hand,
the genus Butyricimonas decreased in MM and its preclinical stages compared to controls
and the genus Blautia was lower at MMdx compared to MMcr patients. Both microorganisms
are butyrate producers which suggests a possible beneficial role of these microorganisms.
In contrast, some genus such as Lachnoclostridium were increased in MMdx (Fig. 1C).
To delve into the functional profiles of the microbial communities, we predicted metabolic
pathways based on taxonomic profiles and we observed an increased in pathways involving
the SCFA propionate in MMcr compared to MMrf (Fig. 1D). The SCFA butyrate was found
decreased in MMdx compared with MGUS patients in serum levels (Fig. 1E). The in vitro
assays with MM cell lines showed that the SCFAs acetate, butyrate and propionate had
an antiproliferative effect (Fig. 1E).
Image:
Summary/Conclusion: Our study showed differences in the abundance levels of gut microbiota
both on the progression and the response of the MM disease. The presence of some microorganisms
and their metabolites, such as SCFAs, suggests a beneficial role which could be exploited
as biomarkers of prognosis.
P870: DEFINING A NOVEL FUNCTION FOR THE POST-TRANSLATIONAL MODIFICATION UFMYLATION
IN THE ADAPTIVE RESPONSE TO ARGININE DEPRIVATION IN MULTIPLE MYELOMA
A. Romano1,*, G. Scandura1, C. Giallongo2, E. La Spina1, A. Barbato1, D. Tibullo3,
K. Todoerti4, A. Neri4, G. A. Palumbo2, F. Di Raimondo2
1Dipartimento di Chirurgia e Specialità Medico Chirurgiche; 2DIPARTIMENTO DI SCIENZE
MEDICHE, CHIRURGICHE E TECNOLOGIE AVANZATE; 3DIPARTIMENTO DI SCIENZE BIOMEDICHE E
BIOTECNOLOGICHE, Università degli Studi di Catania, CATANIA; 4Department of Oncology
and Hemato-oncology, University of Milan, Milano, Italy
Background: Our previous work showed that arginine deprivation is associated to multiple
myeloma (MM) bortezomib refractoriness, but the molecular basis of this adaptive response
is still largely unknown.
Aims: We designed an integrative transcriptional and metabolic study to explore the
contemporary transcriptional and metabolic changes occurring upon arginine deprivation
in four human myeloma cell lines (HMCLs, U266, NCI-H929, OPM2 and MM1.s).
Methods: HMCLs were individually cultured with customized complete with different
Arg concentrations, correspondent respectively to 100%, 25% and 10% of the arginine
concentration in MM bone marrow. After 24-48 hours of culture, the cells were collected
for global metabolomic analysis (Metabolon Inc) and transcriptome profiling by Illumina
platform. Apoptosis and mitochondrial depolarization were measured by FACS. Protein
expression was evaluated by western blot analysis.
Results: In all tested HMCLs the progressive arginine deprivation (range 1124-0 µM)
induced delay in cell cycle until proliferation arrest (in total lack of arginine
in culture media), with increase of G0-G1 length, at specific timepoints for each
cell line tested. However global protein synthesis was not affected and surprisingly,
in all tested cell lines, the monoclonal component secretion in extracellular medium
was preserved after 96 hours of arginine starvation.
At 48 hours of arginine deprivation (range 1124-100 µM) GCN2 pathway was induced in
HMCLs with consequent increase of basal levels of ATF4, p62, GABARAP and autophagy
flux. After 48 hours of arginine starvation, coupled metabolomics and RNA-seq, disclosed
an impairment in the pool of nucleotides and activation of salvage purine pathway,
that could explain changes in cell cycle and proliferation. Arginine-deprived MM cells
had higher levels of choline and phospho-ethanolamine with lower levels of glycerophosphorylcholine
(GPC) compared to controls suggesting an effect of arginine-deprivation on membrane
dynamics, associated to increased production of fatty acids, palmitate (16:0) and
stearate (18:0), required for the generation of new membranes. Culture in arginine
deficient media resulted in complete depletion of reduced glutathione (GSH) and oxidized
glutathione (GSSG), with depletion of gamma-glutamylcysteine, indicating increased
levels of oxidative stress. Gene set enrichment analysis (GSEA) showed deep transcriptome
rearrangements, involving upregulation of genes required for the unfolded protein
response, NF-kB response to TNF, p53 pathway and networks and proteosome degradation,
with a minimal effect on metabolism features, except the upregulation of genes involved
in lactate generation and degradation. In all cell lines, transcripts involved in
the UFMylation pathway were affected, with reduction of UFSP2 and increase of UFMYlation
targets RPL26, PKM2, ENO-1, PGK-1 and SQSTM1/p62. In a large series of primary samples
(9N - 20MGUS - 33SMM - 170MM - 24PPCL - 12SPCL) and 18 MM cell lines, through bioinformatic
analysis of previously published datasets (GENE1.0_BAv20_Neri-Gutierrez), we found
that myeloma progression was associated to increased expression of UFMYlation targets
ENO-1, PGK1 and DLD.
Summary/Conclusion: Taken together, our data clearly indicate how arginine starvation
can induce an heterogeneous adaptation processes occur in individual tumour clones.
The ER stress adaptive response triggered by arginine deprivation promotes UFMylation
activation, due to downregulation of UFSP2 and accumulation of RPN1/RPL26 targets.
P871: ISB 1442, A FIRST-IN-CLASS CD38 AND CD47 BISPECIFIC ANTIBODY INNATE CELL MODULATOR
FOR THE TREATMENT OF CD38 POSITIVE HEMATOLOGICAL MALIGNANCIES
C. Grandclement1, C. Estoppey1, E. Dheilly1, M. Panagopoulou1, E. Martini1, V. Labanca1,
S. De Angelis1, J. Frei1, A. Drake1, A. Rubod1, G. Gudi2, V. Udupa3, J. Koch Olsen4,
R. Giovannini4, M. A. Doucey1, E. Feldman2, C. Konto2, A. Srivastava1, M. Perro1,
S. Sammicheli1,*
1Ichnos Sciences, Epalinges, Switzerland; 2Ichnos Sciences, New York, United States
of America; 3Glenmark Pharmaceuticals Limited, Mahape, India; 4Ichnos Sciences, La
Chaux-de-Fonds, Switzerland
Background: ISB 1442 is a first in class 2 + 1 biparatopic bispecific antibody targeting
CD38 x CD47 using the BEAT® 2.0 (Bispecific Engagement by Antibodies based on the
TCR) platform. The CD38 binding arm consists of bi-paratopic Fabs that strongly bind
to CD38 in two different epitopes on tumor cells and do not functionally compete with
daratumumab. The anti-CD47 arm comprises a single Fab arm designed to block the interaction
between CD47 and the signal-regulatory protein alpha (SIRPα) receptor present on phagocytes
(including macrophages, monocytes and dendritic cells). With this design, the high
affinity anti-CD38 Fab arms preferentially drive binding to tumor cells and enables
blocking of proximal CD47 receptors on the same cell via avidity-induced binding.
The Fc portion of ISB 1442 is engineered to enhance antibody dependent cell phagocytosis
(ADCP), antibody dependent cell cytotoxicity (ADCC) and complement dependent cytotoxicity
(CDC).
Aims: ISB 1442 was developed for the treatment of hematologic malignancies that express
CD38, including multiple myeloma (MM), acute myeloid leukemia (AML), and T-acute lymphoblastic
leukemia (T-ALL), with the intention of overcoming mechanisms of resistance to CD38-targeted
therapies such as daratumumab and isatuximab, and minimizing hemagglutination/hemolysis
on red blood cell (RBC) observed with anti-CD47 monoclonal antibodies (mAb) such as
magrolimab.
Methods: ISB 1442 was tested for its capacity in vitro to induce ADCP, ADCC and CDC
relative to daratumumab and magrolimab across a broad range of MM, AML and T-ALL cell
lines expressing different levels of CD38 and CD47. To assess the complex mechanisms
of action of ISB 1442 in a single system, a multiple mode of action of killing (MMoAK)
assay was established to allow for simultaneous killing by natural killer cells (ADCC),
autologous macrophages (ADCP), and complement from human serum (CDC). In vivo, ISB
1442 was assessed in a therapeutic model of subcutaneously established Raji tumor
xenograft in CB17/SCID mice compared to daratumumab and magrolimab. On-target specificity
was evaluated in vitro in human and monkey whole blood assays.
Results: In vitro, ISB 1442 exhibited higher killing potency compared to daratumumab
across a range of CD38-expressing MM, AML and T-ALL tumor cells. Additionally, ISB
1442 showed in vitro tumor killing potency through phagocytosis comparable to magrolimab,
acting mostly through ADCP. In the CDC, ADCC and MMoAK assays, ISB 1442 exhibited
tumor cell killing that was twice as high as daratumumab in MM cell lines.
In vivo, ISB 1442 induced higher tumor growth inhibition than daratumumab and comparable
tumor regression to magrolimab.
ISB 1442 did not cause any detectable hemolysis, RBC depletion or platelet aggregation
and showed markedly lower hemagglutination relative to magrolimab, suggesting a more
favorable on-target specificity profile in humans. In monkeys, ISB 1442 showed more
pronounced binding on RBC than magrolimab due to higher expression of CD38, suggesting
that monkey is a more sensitive species than human for toxicological evaluation of
CD38-targeted therapies.
Summary/Conclusion: In summary, we report a novel approach for the treatment for CD38
positive hematologic malignancies by co-targeting CD38 and CD47 using a first in class
multispecific antibody. Based on its unique design and multiple mechanisms of action,
ISB 1442 is anticipated to enhance antitumor activity in patients relative to anti-CD38
mAbs by overcoming primary and acquired tumor escape mechanisms of resistance.
P872: DEL(1P32) REMAINS A POWERFUL PROGNOSTIC FACTOR IN A LARGE COHORT OF NDMM PATIENTS:
AN UPDATE
A. Schavgoulidze1,2,*, A. Perrot2,3, T. Cazaubiel4, X. Leleu5, S. Manier6, L. Buisson2,7,
S. Maheo2,7, L. Do Souto Ferreira2,8, R. Lannes7,9, L. Pavageau2,7, C. Hulin4, J.-P.
Marolleau10, L. Voillat11, K. Belhadj12, M. Divoux13, B. Slama14, S. Brechignac15,
M. Macro16, A.-M. Stoppa17, L. Sanhes18, F. Orsini-Piocelle19, J. Fontan20, M.-L.
Chretien21, H. Demarquette22, M. Mohty23, H. Avet-Loiseau2,7, J. Corre2,7
1Unit for Genomics in Myeloma, IUCT-Oncopole; 2CRCT INSERM U1037 and Paul Sabatier
University; 3Hematology Department, University Hospital IUC-Oncopole, Toulouse; 4Hematology
Department, University Hospital, Bordeaux; 5Hematology Department, University Hospital,
Poitiers; 6Hematology Department, University Hospital, Lille; 7Unit for Genomics in
Myeloma, University Hospital IUC-Oncopole; 8Unit for Genomics in Myeloma, University
Hospital, Toulouse, France; 9Dana-Farber Cancer Institute, Harvard Medical School,
Boston, United States of America; 10Hematology Department, University Hospital, Amiens;
11Hematology Department, Hospital, Chalon-sur-Saône; 12Hematology Department, University
Hospital, Créteil; 13Hematology Department, University Hospital, Nancy; 14Hematology
Department, Hospital, Avignon; 15Hematology Department, University Hospital, Bobigny;
16Hematology Department, University Hospital, Caen; 17Hematology Department, Institut
Paoli Calmettes, Marseille; 18Hematology Department, Hospital, Perpignan; 19Hematology
Department, Hospital, Annecy; 20Hematology Department, University Hospital, Besançon;
21Hematology Department, University Hospital, Dijon; 22Hematology Department, Hospital,
Dunkerque; 23Hematology Department, Saint-Antoine University Hospital, Paris, France
Background: Multiple myeloma (MM) is the second hematological malignancy in the Western
countries. Currently, the Revised International Staging System (R-ISS) is widely used
to assess patients’ prognosis. Cytogenetic abnormalities (CA) affecting the chromosome
1, gain 1q and del(1p32), were not included in the new criteria of HR CA despite their
relatively high frequencies (respectively 35% and 11%), due to insufficient data in
the study. However, we have recently confirmed the significant impact on del(1p32)
on myeloma‘s prognosis, being the second most pejorative abnormality, just after del(17p).
Aims: The aim of this study is to update our data about the prognostic impact of del(1p32)
on a large cohort of NDMM patients.
Methods: Clinical data were obtained from 2551 NDMM patients, followed up for ≥ 36
months or having died or progressed within 36 months post treatment. Informed consent
was obtained for all included patients. 1258 patients were treated by an intensive
therapy. Follow-up duration was estimated using reverse Kaplan-Meier method. Overall
survival (OS) and progression free survival (PFS) curves were estimated using the
Kaplan-Meier method and were compared using the log-rank test. Tests were two-sided,
and P < .05 was considered significant. All analyses were performed using R version
4.1.1.
Results: We observed 12.4% of patients displaying del(1p32), which was the expected
proportion. Median follow-up was 67.4 months. The OS of patients harboring del(1p32)
was twice as short as the OS of patients without del(1p32) (60.4 and 123.9 months,
respectively, P < 0.0001) (Figure 1). Likewise, PFS was significantly shorter in del(1p32)
patients (18.1 and 29.1 months, respectively, P < 0.0001). These poorer outcomes were
observed even in patients treated with an intensive therapy (del(1p32) vs. no del(1p32);
PFS 26.5 vs. 37.0 months, P < 0.0001; OS 72.0 vs. 127.4 months, P < 0.0001).
We observed higher frequencies of del(17p) and gain 1q in del(1p32) patients (21.3%
and 53.2% respectively), compared to general MM population. To check if the poor survival
is not only due to these higher levels of association, we have decided to focus on
patients harboring del(1p32) without the main high-risk (HR) CA. HR CA were defined
by the presence of del(17p), t(4;14) and/or gain 1q. Patients without HR CA had a
lower PFS and OS when they carried del(1p32) (del(1p32) vs. no del(1p32); PFS 25.6
vs. 34.8 months, P = 0.0004; OS 83.0 vs. 136.1 months, P = 0.0002).
It is now widely admitted that cumulating HR CA worsen the prognosis. Thus, we have
assessed the effect of additional CA on the prognosis of del(1p32) patients. Additional
CA were CA defined as HR CA above. As expected, the overall survival of del(1p32)
patients significantly decreases when this abnormality was associated with other CA
(OS: del(1p32) alone 83.0 months, del(1p32) with 1 HR CA 45.8 months, del(1p32) with
2 HR CA or more 36.5 months, P < 0.0001).
Image:
Summary/Conclusion: Here we have confirmed the pejorative impact of del(1p32) in multiple
myeloma on the largest cohort of NDMM patients ever evaluated to our knowledge (316
del(1p32) patients). Our results demonstrate the importance of the detection of del(1p32)
at diagnosis because of its huge impact on the prognosis, especially in the era of
risk-adapted treatment strategy. The multivariate analysis is in progress.
P873: CARFILZOMIB RESISTANCE IS ASSOCIATED WITH SIGNIFICANT DEREGULATION OF THE BH3
FAMILY PROTEINS
A. Schneller1,*, A. Bolomsky1, N. Zojer1, M. Schreder1, H. Ludwig1
1Clinic Ottakring, Department of Medcin I, Wilhelminen Krebsforschung, Wien, Austria
Background: Despite the remarkable clinical activity of PIs (Proteasome inhibitors)
in MM (multiple myeloma), most patients ultimately relapse and become PI-resistant.
Deregulation of the apoptosis pathway has been identified as a possible cause of drug
resistance in tumor cells. The intrinsic apoptosis pathway is regulated by the BCL2
protein (BH3) family. Previous studies indicated a substantial role of the BH3 family
in PI resistance and in response to PI. For instance, the sensitivity to the BCL-2
inhibitor venetoclax is increased in PI resistant AMO-1 cells and a deregulation of
BH3 proteins Noxa and MCL1 was noted in MM cell lines exposed to bortezomib. These
data suggest that mechanisms involved in PI resistance are closely interconnected
with the intrinsic apoptosis pathway orchestrated via the BH3 family, revealing novel
opportunities for therapeutic intervention.
Aims: Here, we analyze the interplay between BH3 protein family members in PI resistant
vs. sensitive cell lines aiming to decipher survival strategies of PI resistance.
Methods: MM cell lines were exposed to serially increasing doses (max. 25nM) of carfilzomib
for > 1 year, leading to the outgrowth of clones with acquired carfilzomib resistance.
Analysis of BH3 protein expression and sensitivity to BH3 mimetics in PI resistant
cell lines were performed via western blot and viability assays, respectively. Analysis
of BH3 protein dependency was done using the flow cytometry-based BH3 profiling method.
Thereby, cytochrome c release was measured upon exposure to peptides specifically
binding anti-apoptotic BH3 proteins, allowing to identify the relevance of distinct
anti-apoptotic BH3 proteins in the cell line of interest. Apoptosis induction of the
cells was analyzed by flow cytometry.
Results: Viability assays indicate that all 7 tested carfilzomib resistant MM cell
lines developed a cross resistance to the MCL-1 inhibitors S63845 and AZD-5991. Decreased
sensitivity to the BCL-2 inhibitor venetoclax as well as to the BCL-xL inhibitor A1331852
was observed in resistant KMS12PE cells. As previously shown, these effects are linked
to MDR1 upregulation in carfilzomib resistant cells and reversible by MDR-1 inhibition.
BH3 profiling indicates that under naïve conditions the changes in BH3 protein dependency
could be observed in 3/7 carfilzomib resistant cell lines. Specifically, resistant
KMS12PE cells exhibit a decreased sensitivity to the MCL-1 binding peptide, carfilzomib
resistant OPM-2 cells show an increased sensitivity towards peptides binding to BCL-xL
and A1 and resistant AMO-1 cells demonstrate a reduced sensitivity a peptide binding
to BCL-xL, Bcl-2 and BCL-w. Protein expression analysis of carfilzomib sensitive vs.
resistant cell line variants indicate alterations of BCL2 family members (MCL-1, BCL-xL,
BCL-2, BAD, BID, BAX, BAK, BIM, NOXA and PUMA) in untreated conditions as well as
under carfilzomib treatment (Figure 1). Notably, carfilzomib treatment upregulated
BCL-xL in 5 and BAK in 7 out of 7 carfilzomib resistant cell line variants.
Image:
Summary/Conclusion: Our data show significant deregulation of the BH3 protein family
in carfilzomib resistant cell lines highlighting the importance of the apoptosis signaling
pathway in PI resistance. This is further emphasized by the coherent deregulation
of BCL-xL and BAK throughout carfilzomib exposed carfilzomib resistant cell lines
when compared to their sensitive cell line variants. Moreover, BH3 profiling indicates
a change in dependency on anti-apoptotic BH3 protein in carfilzomib resistant vs.
sensitive MM cell lines.
P874: DECIPHERING PLASMA CELL HETEROGENEITY AND TUMOR MICROENVIRONMENT IN LIGHT-CHAIN
AMYLOIDOSIS USING SINGLE CELL RNA-SEQUENCING
A. Srivastava1,*, R. Kumari1, S. Adhikari1, P. Sergeev1, J. J. Miettinen1, M. H. Suvela1,
K. Flanagan2, A. Slipicevic2, N. N. Nupponen2, F. Lehmann2, C. Heckman1
1Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki,
Finland; 2Oncopeptides AB, Stockholm, Sweden
Background: Immunoglobulin light-chain amyloidosis (AL) is a rare disease caused by
plasma cell secretion of misfolded light chains that assemble as amyloid fibrils,
which deposit on vital organs causing organ dysfunction. Similar to AL, multiple myeloma
(MM) is also caused by atypical plasma cell (PC) expansion in the bone marrow. Although,
much research has been done on MM to understand disease mechanisms and to develop
effective treatments, knowledge related to AL is still limited. Notably, there is
little information available regarding the associated tumor microenvironment (TME)
of AL. As the TME plays an important role in disease development and treatment response,
comprehensive investigation of the AL TME could improve disease diagnosis and treatment.
Furthermore, information distinguishing PCs from AL and MM patients is relatively
sparse, but could help determine if treatments developed for MM would also be applicable
for the treatment of AL.
Aims: The main objective of our study was to decipher the underlying disease mechanisms
of the AL at cellular and molecular level by analyzing AL patient samples using single
cell RNA sequencing (scRNAseq). To achieve this, we aimed to understand heterogeneity
within AL vs MM plasma cells and the TME at the cellular and gene expression level.
Methods: The Finnish Hematology Research Biobank provided 23 viably frozen bone marrow
mononuclear cell (BM-MNC) samples from 22 AL patients and 24 samples from 23 MM patients.
The BM-MNCs were thawed and sorted based on their CD138 expression and cell viability
(7AAD dead cell marker). The viable CD138+ and CD138- cells were mixed in a ratio
where CD138+ were enriched, but not exceeding 50%. The Chromium Single Cell 3’RNAseq
run and library preparation were done using Chromium™ Single Cell 3’ Reagent version
3 chemistry. 10x Genomics Cell Ranger v3.0.1 pipelines were used for the initial data
processing. Further, the quality control and data preprocessing was performed using
Seurat 4.1.0. For cell type annotations to identify the clusters, specific cell type
markers for the immune cells were obtained from sctype database, GSEA-MSigDB and manual
curation.
Results: From the integrated analysis of scRNAseq data from 22 AL patient samples,
we identified a total of 27 distinct clusters of cells belonging to 10 unique cell
types. Out of the 27 clusters, 10 clusters were identified as PC sub-populations.
The proportions of PCs in the samples negatively correlated to the other major cell
types such as NK cells, T cells, gamma delta T cells and monocytes. High transcriptional
variability was observed between the PC clusters of the AL patients indicating a high
degree of heterogeneity among the AL PCs. Analysis of the MM scRNAseq data resulted
in 14 PC clusters, however variability between these clusters was not much distinct
as compared to AL. A comparison between AL and MM PC clusters resulted in overlapping
transcriptional profiles of 2 AL PC clusters with one MM PC cluster. 5 out of 10 clusters
of PCs in AL patients were found to be associated with protein processing in the endoplasmic
reticulum and apoptosis pathway.
Summary/Conclusion: Within the AL TME, we observed negative correlation of major immune
cell types with PCs, suggesting that immune cells may contribute to controlling tumor
cell burden in AL. In addition, PC sub-populations in amyloidosis patients are very
heterogeneous at the inter-individual level compared to MM patients further indicating
that AL patients may benefit from more tailored treatment approaches.
P875: ACTIVATION OF LNCRNA NEAT1 LEADS TO SURVIVAL ADVANTAGE OF MULTIPLE MYELOMA CELLS
BY SUPPORTING A REGULATORY LOOP WITH DNA REPAIR PROTEINS: RATIONAL BASES FOR NEAT1
THERAPEUTIC TARGETING IN THE DISEASE
E. Taiana1,2,*, D. Ronchetti1,2, V. K. Favasuli1,2, I. Silvestris1,2, N. Puccio1,2,
K. Todoerti3,4, C. Bandini5,6, I. Craparotta7, L. Di Rito7, S. Erratico8,9, D. Giannandrea10,
R. Piva5,6, M. Bolis7, Y. Torrente9, R. Chiaramonte10, N. Bolli1,2, L. Baldini1,2,
A. Neri1,2
1Oncology and Hemato-oncology Department, University of Milan; 2Hematology, Fondazione
Cà Granda IRCCS Policlinico; 3Pathology Department; 4Molecular Oncology 1 Department,
IRCCS Istituto Nazionale dei Tumori, Milan; 5Molecular Biotechnology and Health Sciences
Department, University of Turin; 6Città Della Salute e della Scienza Hospital, Turin;
7Oncology Department, Mario Negri IRCCS; 8Novystem Spa; 9Unit of Neurology, Fondazione
Cà Granda IRCCS Policlinico; 10Health Sciences Department, University of Milan, Milan,
Italy
Background: Multiple myeloma (MM) is a fatal malignant proliferation of antibody-secreting
bone marrow plasma cells characterized by marked genomic instability.
Long non-coding RNA (lncRNA) represents the largest class of non-protein coding genes
in the human genome. Their crucial role in solid tumor and hematological malignances,
including MM, is well documented.
NEAT1 is a highly expressed nuclear lncRNA, representing the core structural component
of paraspeckle organelles. We previously demonstrated that NEAT1 silencing negatively
regulates proliferation and viability of MM cells, both in vitro and in vivo, highlighting
its pivotal role in DNA integrity maintenance, by regulating the homologous recombination.
Despite the increasing information obtained by loss-of-function approaches, there
is still a lack of information regarding possible oncogenic benefits acquired by MM
cells upon NEAT1 overexpression.
Aims: Taking advantage of the use of the CRISPR/Cas9 SAM genome editing approach,
the present study aimed to shed light on the biological and molecular implication
of NEAT1 overexpression in MM.
Methods: We adopted the CRISPR/Cas9 Synergistic Activation Mediator editing system
to transactivate NEAT1 in AMO-1 MM cell line. LNA-gapmeR antisense oligonucleotide
technology was used to silence NEAT1 in MM cells by gymnotic delivery. Hypoxia was
induced by culturing the cells into a modular incubator chamber flushed with a mixture
of 1% O2, 5% CO2 and 94%N2 at 37 °C. NEAT1 expression was assessed by qRT-PCR and
RNA-FISH. Cell cycle phases distribution and apoptotic cells percentage were assessed
by flow cytometry. Clonogenic potential was evaluated by methylcellulose assay. Protein
expression was investigated by WB and IF. Cell morphological analysis was performed
after May-Grunwald Giemsa staining. Transcriptomic analysis was performed by RNA sequencing,
following the TruSeq Stranded Total RNA protocol. Libraries with optimal quality and
quantity were run on NextSeq 500. Statistical significance was determined by Student
t test analysis; differences were considered significant when P-value was <0.05.
Results: We performed a transcriptomic analysis of AMO-1 cells either transactivated
or silenced for NEAT1. Our data revealed a NEAT1 pivotal role in the maintenance of
DNA integrity, showing a significant deregulation of almost all the crucial cellular
DNA repair mechanisms for both single and double strand breaks. In particular, we
found that NEAT1 transactivation affects DNA repair mechanisms through the activation
of a molecular axis including two fundamental kinase proteins, i.e. ATM and DNA-PKcs,
and the direct target pRPA32. Furthermore, our data strongly confirmed the activation
of this NEAT1-orchestrated molecular axis whether MM cells are exposed to nutrient
starvation and hypoxia, suggesting its implication in conferring oncogenic and pro-survival
properties to MM cells. The disruption of this oncogenic loop leads to MM cell death
and the loss of pro survival advantages, thus suggesting that NEAT1 should be considered
a crucial factor for MM cells survival.
Summary/Conclusion: Overall, we provided novel important insights into the role of
NEAT1 in MM pathobiology, demonstrating that its deregulation strongly correlates
with adaptation to stress condition, often associated with late stages of the disease.
Taken together, our results suggest that NEAT1 could represent Achilles’ heel for
MM cells and should be considered a potential therapeutic target for MM treatment.
P876: INCIDENTAL DISCOVERY OF PROBABLE PATHOGENIC GERMLINE VARIANTS IN MULTIPLE MYELOMA
PATIENTS UNDERGOING SOMATIC GENOMIC PROFILING
S. Thibaud1,*, T. Brander2, M. Balwani2, T. Mouhieddine1, D. Melnekoff2, O. Van Oekelen2,
A. Lagana2, J. Houldsworth3, A. Chari1, J. Richter1, L. Sanchez1, S. Richard1, C.
Rodriguez1, A. Rossi1, H. J. Cho1, S. Jagannath1, K. Onel4, S. Parekh1
1Hematology & Medical Oncology; 2Genetics and Genomic Sciences; 3Molecular Pathology,
Icahn School of Medicine at Mount Sinai, New York; 4Sema4, Stamford, United States
of America
Background: Next-generation sequencing (NGS) is increasingly used to characterize
the somatic mutational landscape of patients with multiple myeloma (MM) and guide
precision medicine approaches. Somatic genomic profiling (SGP) in cancer patients
can lead to the incidental discovery of pathogenic or likely-pathogenic germline variants
(PGVs) in ~4% of cases (PMID 26787237). Our previous analysis of whole exome germline
data from 895 MM patients in the MMRF CoMMpass dataset showed ~8% of MM patients carry
a PGV in a hereditary cancer gene (HCG), and identified shared characteristics among
PGV carriers including younger age at diagnosis, higher likelihood of having family
history of hematologic malignancy, higher likelihood of t(11;14) MM, and favorable
outcomes (Thibaud et al, ASH 2021).
Aims: The present study aims to assess the rate of detection of probable PGVs (PPGVs)
in MM patients undergoing targeted SGP.
Methods: We retrospectively reviewed results of 1,614 commercial NGS panels performed
by a single genetic testing laboratory on bone marrow aspirates of 1,095 MM patients
treated at our institution since 2015 who were not enrolled in the MMRF CoMMpass study.
We considered individual variants to be PPGVs when all of the following criteria were
met: 1) variant found in a panel of 106 well-established HCGs (same panel used in
our prior study, but excluding TP53 given high rate of somatic mutations in MM); 2)
variant unambiguously listed as pathogenic or likely pathogenic (P/LP) in ClinVar,
or variant classified as “conflicting interpretations of pathogenicity” but reported
by ≥2 CLIA-certified labs as P/LP; 3) variant allele frequency (VAF) >30% (per PMID
33236764); 4) variant detected in all available test reports for a given patient (and
VAF >30% in all available test reports). Well known founder variants (FV) in specific
populations were labeled as PPGV-FVs.
Results: 95 MM patients (8.7%) had a genetic variant meeting criteria to be classified
as a PPGV. Two patients had 2 distinct PPGV. PPGV were most commonly detected in the
following HCGs (Table 1): APC (n=22), CHEK2 (n=20), ATM (n=7), BRCA1 (n=6), MUTYH
(n=6). Of all PPGV carriers, 47 (49%) carried a PPGV-FV. Median VAF for PPGVs was
48.2% (range 30-85%). 34 PPGV carriers had serial NGS tests available (range 2-5 reports);
in these patients, VAF standard deviation across serial measurements was <5% in 89%
of cases. Taken together, these findings indicate a high likelihood that PPGV detected
in these MM patients while undergoing tumor sequencing are indeed incidentally-discovered
PGV warranting referral to a genetic counselor for consideration of confirmatory testing.
Image:
Summary/Conclusion: SGP of bone marrow aspirate in MM patients incidentally detected
PPGV in well-established HCG in 8.7% of cases. These findings are consistent with
those of our prior study, which found an 8.6% prevalence of PGV in newly-diagnosed
MM patients. In MM patients undergoing SGP, detection of a mutation involving a HCG
and with a VAF >30% should prompt referral to genetic counseling for confirmatory
testing, as PGV detection can have clear implications for patients and their families.
P878: HIGH MUTATIONAL BURDEN OF CHROMATIN MODIFYING GENES DEFINES A RELATIVELY INDOLENT
SUBGROUP OF PLASMA CELL DYSCRASIAS
X. Wang1,*, B. Qiao2, Y. Yu1, J. Xu2, L. Zuo2, J. Chen1, H. Cai1, H. Han1, X.-X. Cao1,
C. Sun2, J. Li1
1Department of Hematology, Peking Union Medical College Hospital, Chinese Academy
of Medical Sciences and Peking Union Medical College, Beijing; 2Institute of Hematology,
Union Hospital, Tongji Medical College, Huazhong University of Science and Technology,
Wuhan, China
Background: Plasma cell dyscrasias (PCDs), including multiple myeloma (MM), light
chain amyloidosis (AL), POEMS syndrome, and MGUS, present different clinical features.
But as malignancies of plasma cells, they potentially have common mutational patterns
associated with chemotherapy and prognosis, which haven’t been elucidated.
Aims: We aimed to reveal the clinically relevant subgroups of MM, AL, POEMS, and MGUS
defined by mutational patterns of bone marrow plasma cells (BMPCs).
Methods: We conducted targeted gene sequencing including 370 genes of BMPCs from patients
with MM (n=163), AL (n=121), POEMS (n=67) and MGUS (n=13). All the subjects provided
informed consent.
Results: Principal component analysis of the top 100 variable genes revealed 2 major
subgroups among patients with PCDs (Fig. 1a). Approximately 25% of MM, AL, POEMS and
MGUS, subgroup 1 was featured by the heavily mutated genes enriched in histone modifications
and chromatin organization, such as KMT2D, EP400, RUNX1, SOX9, KDM5A, NCOR2, and ARID1B.
Subgroup2 showed lower total mutational burden and was featured by mutations of proto-oncogene
and B cell differentiation-associated genes, such as NCOR1, APOBEC4, KRAS, TP53, FGFR3,
FAM46C, and MYC (Fig. 1b). The high mutational burden of chromatin-modifying genes
was exclusively observed in subgroup 1 and was comparable among MM, AL, POEMS, and
MGUS (Fig. 1c). Patients in subgroup 1 showed worse responses to the first-line CyBorD
for MM and AL (Fig. 1d), but better responses to the lenalidomide-based VRD for MM
and Rd for POEMS. Overall, patients in subgroup 1 demonstrated longer progression-free
survival (PFS), which was observed in MM, AL (Mayo12, Stage I, II), and POEMS, respectively
(Fig. 1e).
Image:
Summary/Conclusion: These results provide mechanistic insights into a relatively indolent
subgroup among patients with MM, AL, POEMS, and MGUS, which is featured by a high
mutational burden of chromatin-modifying genes. This subgroup can be identified by
detecting the mutations of chromatin-modifying genes in clinical management. Overall
longer PFS and better response to lenalidomide-based first-line therapy can be expected.
P879: EXENT AND FLC MASS SPECTROMETRY FOR SEROLOGICAL IDENTIFICATION AND QUANTIFICATION
OF MONOCLONAL IMMUNOGLOBULINS IN MULTIPLE MYELOMA
S. Huhn1,*, A. M. Poos1, E. K. Mai1, M. Hänel2, H. J. Salwender3, J. Dürig4, I. W.
Blau5, C. Scheid6, K. C. Weisel7, M. Munder8, O. Berlanga9, U. Bertsch1, H. Goldschmidt1,
N. Weinhold1
1Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg; 2Department
of Internal Medicine III, Clinic Chemnitz, Chemnitz; 3Asklepios Tumorzentrum Hamburg,
AK Altona and AK St. Georg, Hamburg; 4Department for Hematology and Stem Cell Transplantation,
University Hospital Essen, Essen; 5Medical Clinic, Charité University Medicine Berlin,
Berlin; 6Department of Internal Medicine I, University of Cologne, Cologne; 7Department
of Oncology, Hematology and BMT, University Medical Center Hamburg-Eppendorf, Hamburg;
8Third Department of Medicine, University Medicine Mainz of Johannes Gutenberg, Mainz,
Germany; 9The Binding Site Group, Ltd, Birmingham, United Kingdom
Background: Mass spectrometry has broad potential for screening and monitoring monoclonal
gammopathies. The technology overcomes some of the limitations inherent to standard
electrophoretic methods for identifying monoclonal immunoglobulins and is amenable
to automation.
Aims: Here we assess the performance of the EXENT assays for the quantification of
M-proteins and whether free light chain evaluation by mass spectrometry (FLC-MS) offers
added sensitivity during multiple myeloma monitoring.
Methods: The MM5 trial compared two regimens of bortezomib-based induction therapy
and of lenalidomide consolidation followed by lenalidomide maintenance in newly diagnosed
multiple myeloma patients. We used mass spectrometry for identifying serum M-proteins
in samples at baseline (n=434), post-induction (n=340), and post-consolidation (n=294).
Briefly, polyclonal antibodies (anti-IgG,-IgA,-IgM,-total κ and -total λ [EXENT, The
Binding Site, UK] and anti-free κ and -free λ [FLC-MS]) covalently attached to paramagnetic
microparticles were separately incubated with serum, washed, treated to elute and
reduce patient immunoglobulins, and spotted onto MALDI plates with HCCA matrix. Spectra
were generated using the MALDI-TOF-MS system. Light chain mass spectra were obtained
using EXENT software to yield immunoglobulin isotype, mass-to-charge ratio (m/z) and
quantity (g/L). Results were compared to serum protein electrophoresis, immunofixation,
and FLC ratios (Freelite®, The Binding Site, UK).
Results: EXENT demonstrated improved sensitivity over immunofixation for identifying
the M-protein at each time-point of the MM5 trial: 99% vs. 95% at baseline; 91% vs
80% post-induction; and 63% vs. 46% post-consolidation. Agreement between methods
was 87%.
The addition of FLC-MS to EXENT further improved sensitivity to 100%, 95.0%, and 72%
at each respective time-point, whereas the combination of immunofixation and FLC ratios
resulted in sensitivities of 100%, 90%, and 54%. Overall concordance between EXENT+FLC-MS
and the combination of immunofixation and FLC ratios was 100% at baseline, 93% post-induction,
and 73% post-consolidation. In all, 158 (11%) samples had an M-protein by EXENT+FLC-MS
only, and 28 (2%) by the combination of immunofixation and FLC ratios.
18 and 92 patients achieved an IMWG complete response after induction and consolidation,
respectively. EXENT identified an M-protein in 5 (28%), and 23 (25%) of these patients,
whereas EXENT+FLC-MS was more sensitive and detected monoclonal proteins in 11 (61%),
and 38 (41%) patients, respectively.
There was acceptable quantitative agreement for M-protein measurements between EXENT
and serum protein electrophoresis (n=631: Passing-Bablok fit: y=0.85x-0.58; coefficient
of determination R2= 0.89; Bland-Altman relative bias (95%CI): -32.62% (-123.79-58.55%)).
However, there was a substantial discordance in 285 samples <10g/L by serum protein
electrophoresis (y=0.58x+0.54; R2=0.28; -44.84% (-179.29 – 84.60%)) compared to 346
samples >10g/L (y=0.85x-0.25; R2=0.82; -19.91% (95%CI: -97.32 – 23.56%)); suggesting
overestimation by serum protein electrophoresis at lower levels.
Summary/Conclusion: EXENT provides superior performance for the identification of
M-proteins throughout monitoring. Sensitivity can be improved by incorporating FLC-MS
assessments, thus providing a means for determining serological residual disease in
patients at complete response by IMWG criteria. Quantification of the M-protein by
mass spectrometry compares well to standard methods and offers accurate results at
levels below the analytical threshold of confidence for electrophoretic approaches.
P880: OVEREXPRESSION OF PFKFB4 PROMOTES ADAPTATION TO HYPOXIC MICROENVIRONMENT IN
MULTIPLE MYELOMA
J. Yang1,*, F. Wang1, B. Chen1
1Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast
University, Nanjing, China
Background: Multiple myeloma (MM) is a hematological malignancy with monoclonal proliferation
of plasma cells, which is closely related to hypoxic bone marrow microenvironment.
Previous studies have found that 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase
4 (PFKFB4) is involved in glycolysis, cell cycle progression, autophagy, and metastasis
in various tumor diseases and overexpression of PFKFB4 is associated with poor prognosis.
However, the underlying mechanism of PFKFB4 in hypoxia adaptation in MM has not been
reported.
Aims: The aim of this study was to explore the role of PFKFB4 in MM cells adaptation
to the hypoxic BM microenvironment and provide potential therapeutic target for clinical
treatment.
Methods: We analyzed expression profile GSE80140 and GSE80545 downloaded from National
Center for Biotechnology Information-Gene Expression Omnibus (NCBI-GEO) database,
and conducted cell transcriptome sequencing. 5MPN, a specific inhibitor of PFKFB4,
was employed to evaluate the effect of PFKFB4 in hypoxia adaptation in MM by cell
proliferation, apoptosis, and cycle assays, F-2,6-BP measurement, and Western blot.
Results: MM cell lines (RPMI-8226 and U266) was found to express higher level of PFKFB4
under hypoxia (1% O2) compared to normoxia (20% O2). 5MPN inhibited MM cells proliferation,
suppressed intracellular F-2,6-BP concentration, and induced cell cycle arrest in
G1 phase. Inhibition of PFKFB4 function had no significant effect on inducing cell
apoptosis. Western blot detected the expression of proliferation, apoptosis and cell
cycle related proteins, and the results were consistent.
Summary/Conclusion: Overexpression of PFKFB4 promotes MM proliferation and cell cycle
progression. 5MPN effectively reserves the process, suggesting that PFKFB4 is a potential
therapeutic target in MM.
P881: CDK7 CONTRIBUTES TO METABOLIC REPROGRAMMING IN MM CELLS THROUGH C-MYC MEDIATED
TRANSCRIPTIONAL CONTROL OF GLYCOLYTIC GENES
Y. Yao1,2, J. Fong Ng1, W. D. Park3, D. Gramegna1, A. Samur1, M. Samur1, E. Morelli1,
M. Chesi4, C. Mitsiades1, K. C. Anderson1, C. Lin3, N. Munshi1, M. Fulciniti1,*
1Dana Farber Cancer Institute, Boston, United States of America; 2Xuzhou Medical University,
Xuzhou, China; 3Baylor College of Medicine, Houston; 4Mayo Clinic, Scottsdale, United
States of America
Background: We have recently elucidated the biological role of CDK7 and explore the
functional consequence of its inhibition in MM using chemical and genetic approaches.
We reported a specific vulnerability in MM cells upon disruption of the CDK7-driven
molecular programs, supporting a model where cell cycle and transcriptional deregulation
are counteracted by inhibition of CDK7 activity leading to cell cycle arrest and subsequent
apoptosis in MM. Importantly, we established that CDK7 positively correlates with
c-MYC and E2F1 transcriptional outputs in primary cells from MM patients, suggesting
its regulatory role for both transcription factors.
Aims: Bioinformatics analyses of RNA-seq data revealed that CDK7 kinase activity is
required for the expression of many components of the glycolytic cascade; in contrast,
most genes for OXPHOS were only weakly modulated by YKL-5-124. Interestingly, recent
studies have identified CDK7 as an activator of glucose consumption in lung cancer
cells, and autosomal dominant polycystic kidney disease. These observations prompted
us to delve into the potential role of CDK7 in regulating glucose metabolism in MM
cells.
Methods: CDK7 inhibition was achieved with 1. selective covalent inhibitor YKL-5-124;
2. protein degradation dTAG system; 3. inducible KD/KO systems. The transcriptional
networks were evaluated by integrated ChIP-seq and RNA-seq data analysis. Metabolic
changes were evaluated by ECAR and OCR assessment using Seahorse analyzer.
Results: To evaluate if CDK7 inhibition suppresses glycolysis we assessed control
and treated cells during a glycolysis stress test with Seahorse analyzer using extracellular
acidification rate (ECAR) as a measure of glycolytic activity. Compared to control,
treated cells displayed a significant defect in glycolysis, glycolytic capacity, and
glycolytic reserve in a c-MYC dependent manner. These differences in glycolytic activity
upon YKL-5-124 treatment were not accompanied by changes in basal oxygen consumption
rate (OCR). Similarly to the drug compound, CDK7 protein degradation using dTAG system
diminished glycolytic activity, suggesting that CDK7 regulates glycolysis in MM model
systems. Evaluation of tumors retrieved from mice after treatment revealed significant
reduction in glycolysis rate. Cancer cells catabolize glucose to lactate in aerobic
conditions: a sharp time dependent increase in extracellular lactate release was observed
in MM cells over time which was suppressed by YKL-5-124 treatment via regulation of
lactate dehydrogenase A (LDHA) expression and enzymatic activity. Together with impairment
of aerobic glycolysis, YKL-5-124 treatment resulted in increased ROS levels and induction
of apoptotic cell death. The cellular level of ROS is an important oxidative molecule
causing endogenous DNA damage. We indeed observed induction of DNA damage as measured
by investigation of γH2AX levels after YKL-5-124 treatment. To explore whether YKL-5-124
induced cell death by ROS-mediated mechanisms, we examined the effects of the thiol
antioxidant N-acetyl-L-cysteine (NAC) on MM cells upon YKL-5-124 treatment. NAC treatment
prevented ROS production, cell death, apoptosis and DNA damage accumulation induced
by CDK7i, indicating a causative link between increased ROS levels upon CDK7i and
tumor cell death.
Summary/Conclusion: Our data demonstrate that CDK7 through its ability to link cell
cycle, transcription and cellular metabolism affects MM cells at several levels, representing
an attractive and therapeutically actionable molecular vulnerability in MM.
P882: IDENTIFICATION OF M7G RNA METHYLATION REGULATORS AND M7G-RELATED GENES IN MULTIPLE
MYELOMA
M. Yu1,*, C. Li2, J. Xu3, M. Liu2, X. Cui2
1Shandong University of traditional Chinese Medicine; 2The Second Affiliated Hospital
of Shandong University of traditional Chinese medicine; 3Affiliated Hospital of Shandong
University of traditional Chinese medicine, Jinan, China
Background: N7-methylguanosine (m7G) is a positively charged, essential modification
at the 5’ cap of eukaryotic mRNA, impacting RNA processing and function, especially
mRNA sublocation, translation, and splicing, which is crucial for the regulation of
tumor progression. However, the m7G modification pattern of multiple myeloma (MM)
is still unclear.
Aims: To explore the impact of m7G regulators on MM prognosis and further conducted
a comprehensive investigation on the differences in m7G-related genes among different
m7G modification patterns.
Methods: We analyzed the mRNA expression of m7G regulators in healthy donors (n =
9) and MM (n = 174) bone marrow plasma cells, investigated their prognostic values,
and comprehensively evaluated the m7G modification patterns of 559 MM bone marrow
samples. These data are from Gene Expression Omnibus (GEO) datasets. ConsensusClusterPlus
R package (1.56.0) was used to group patients.
Results: Compared with healthy donors, the expression of three m7G writers (METTL1,
RNMT, and WBSCR22) was significantly upregulated in MM patients (Fig. 1A). The Kaplan-Meier
(K-M) survival curve showed that three m7G regulators (METTL1, WDR4 and RNMT) were
significantly correlated with survival, and their high expression was associated with
shorter overall survival (OS) (Fig. 1B). Based on the expression levels of m7G regulators,
MM patients with qualitatively distinct m7G modification patterns were classified
into three different groups. The K-M curve analysis showed a significant survival
difference among the subgroups, of which cluster A showed a worse outcome than clusters
B and C (Fig. 1C), and the expression of m7G regulators was higher in cluster A (Fig.
1D). To further investigate the biological behaviors of these distinct m7G modification
patterns, gene set variation analysis (GSVA) was performed. Compared with clusters
B and C, cluster A was markedly enriched in cell proliferation-related pathways. Recent
studies have found that the m7G cap in eukaryotic mRNAs is necessary to regulate sublocation,
translation, and splicing. Therefore, we used the Pearson correlation coefficient
to determine the potential m7G-related mRNAs. As a result, 88 interactions and 88
m7G-related mRNAs (absolute correlation coefficient >0.3, P <0.05) were identified.
Further univariate Cox analysis revealed that 27 mRNAs were independent prognostic
factors for MM patients, 20 of which were high-risk genes, with varying expression
levels in each group (Fig. 1E and 1F). Among them, CDK4, a high-risk gene highly expressed
in cluster A (Fig. 1G), is closely associated with cell proliferation by regulating
the cell cycle, which is consistent with the GSVA results. As with other cancers,
cell cycle dysregulation leads to the progression of MM, where CDK4/6 disorders are
common.
Image:
Summary/Conclusion: This work showed that the high expression of m7G regulators was
correlated with unfavorable prognostic outcomes in MM. The difference in m7G modification
patterns may be an important factor in determining the prognosis of MM patients via
causing disorders of the cell cycle.
P883: TARGETING CALCINEURIN/NFATC1 SIGNALING SENSITIZES PROTEASOME INHIBITION TREATMENT
IN MULTIPLE MYELOMA
J. Zhang1,*, X. Luo1, T. Xia1, K. C. Cheng1, M. H. Ng1
1Blood Cancer Cytogenetics and Genomics Laboratory, Anatomical and Cellular Pathology,
The Chinese University of Hong Kong, Hong Kong, China
Background: Proteasome inhibitors (PIs) (e.g., Bortezomib) are key treatments to improve
the clinical outcome of multiple myeloma (MM). However, disease relapse from the emergence
of PI-resistant clones is the main problem. Insights into the mechanisms underlying
PI-resistance will provide novel druggable targets to prevent relapse. It was well-recognized
that PIs induce endoplasmic reticulum (ER) stress in MM cells. ER stress is associated
with the Calcineurin/nuclear-factor-of-activated-T-cells (NFAT) signaling. Accumulating
studies indicated the importance of Calcineurin/NFAT signaling in tumorigenesis and
drug resistance. However, the involvement of Calcineurin/NFAT signaling in PI-resistance
in MM has not yet been investigated.
Aims: To dissect the role of the Calcineurin/NFAT axis in Bortezomib (BTZ) treatment
response and explore the therapeutic potential by combining Bortezomib and Calcineurin/NFAT
inhibitors in MM.
Methods: We studied the expression pattern of PPP3CA and NFATC1 from MM datasets and
MM cell lines and their association with BTZ responses in MM. The expression of the
four principal NFAT members was detected by expression microarray and their activation
status by western blotting in MM cell lines. We identified the most prominent NFAT
member and assessed its activation under BTZ treatment. A lentivirus-transduction
system was used for overexpressing genes, and siRNA transfection was applied to induce
NFATC1 knockdown in a BTZ-resistant cell line ARH77. Flow cytometry and western blotting
assessed the combinational effect of Calcineurin/NFAT inhibitors and BTZ on MM cells.
Results: The expression level of PPP3CA, which encodes Calcineurin Catalytic subunit
A (CnA), was correlated to the poor clinical outcome of recurrent MM patients (MMRF
cohort). Calcineurin was highly expressed in ARH77, which is less sensitive to BTZ
treatment than other MM cell lines. Among the four NFAT members, we found that NFATC1
and NFATC3 were the most highly expressed members in MM cell lines. However, NFATc1
was the only member suppressed by CsA but activated by the calcium channel agonist
Ionomycin in ARH77. Consistent with Calcineurin, NFATc1 was highly expressed in ARH77,
with reference to a confirmed NFATc1-positive cell line Raji and the nuclear-NFATc1
overexpressing HEK-293T. Additionally, BTZ activated CnA and NFATc1 time-dependently
in ARH77 but failed to activate CnA and NFATc1 in the BTZ-sensitive cell line NCI-H929.
Immunofluorescence showed that BTZ triggered the nuclear translocation of NFATc1.
From GEO datasets, NFATC1 was overexpressed in patients less responsive to BTZ, but
was unchanged in patients subjected to conventional therapies. The Calcineurin/NFATc1
was thus a potential mechanism of BTZ-resistance in MM. Depleting NFATc1 enhanced
BTZ-induced DNA damage-related apoptosis in ARH77. The combinational treatment of
Calcineurin/NFAT inhibitors (CsA and FK506) and BTZ decreased the nuclear/cytosolic
ratio of NFATc1 and promoted apoptosis in two MM cell lines compared to the treatment
of BTZ alone.
Image:
Summary/Conclusion: The activation of the Calcineurin/NFATc1 axis is an important
mechanism that contributes to BTZ-resistance in MM. Targeting this axis confers translational
potential for improving BTZ-based therapies.
P884: POTENTIAL BENEFIT OF POST-TRANSPLANT DOXYCYCLINE IN AL ACHIEVING HEMATOLOGICAL
RESPONSE AFTER AUTOLOGOUS STEM CELL TRANSPLANT – LONG TERM FOLLOW UP
N. Abdallah1,*, A. Dispenzieri1, E. Muchtar1, F. Buadi1, P. Kapoor1, M. Lacy1, Y.
Hwa1, A. Fonder1, M. Hobbs1, S. Hayman1, N. Leung1, D. Dingli1, R. Go1, Y. Lin1, W.
Gonsalves1, M. Binder1, T. Kourelis1, R. Warsame1, R. Kyle1, S. Rajkumar1, M. Gertz1,
S. Kumar1
1Hematology, Mayo Clinic, Rochester, United States of America
Background: Systemic light chain (AL) amyloidosis is a clonal plasma cell disorder
characterized by multiorgan deposition of fibrils composed of misfolded immunoglobulin
light chains. Current treatment strategies targeting the plasma cell clone do not
result in immediate organ recovery, and thus effective fibril-directed therapies are
needed. Doxycycline has been shown to disrupt amyloid fibril formation in preclinical
models. In 2012, we reported initial results on the impact of posttransplant prophylaxis
with doxycycline using data from 445 AL amyloidosis patients; prophylaxis with doxycycline
was associated with improved survival compared to penicillin in those achieving hematologic
response. Since then, few studies have evaluated doxycycline use in AL amyloidosis,
mainly in combination with induction chemotherapy, with mixed results. We provide
here updated results on outcomes of AL amyloidosis patients who received doxycycline
prophylaxis post autologous stem cell transplantation (ASCT) in our institution.
Aims: To evaluate the impact of doxycycline use for post-ASCT antimicrobial prophylaxis
on survival in AL amyloidosis patients.
Methods: We included 553 patients who underwent ASCT from July 1996 to June 2014.
We excluded patients who died within 100 days of ASCT. Doxycycline 100 mg daily was
substituted for penicillin prophylaxis in patients with penicillin allergy; since
2013, doxycycline has become the standard for prophylaxis in AL amyloidosis at our
institution. Prophylaxis was typically continued for a year after ASCT. Antibiotic
type was determined by review of medical records. The Kaplan-Meier method was used
to estimate time to next treatment (TTNT) and overall survival (OS), both measured
from the time of transplant; curves were compared using the log-rank test.
Results: Median age was 59 and 60% were male. The median time from diagnosis to ASCT
was 4 months; 90% had early ASCT (within 1 year of diagnosis) and 57% received upfront
ASCT without prior induction. Penicillin was used for prophylaxis in 67% (372 patients);
the rest received doxycycline. Hematologic and organ response rates were similar in
the 2 groups. Median follow up from ASCT was 10.5 vs. 13.6 years in the doxycycline
and penicillin groups, respectively. Among patients with early ASCT, TTNT was 5.5
(95%CI: 4.2-8.1) vs. 5.8 (95%CI: 4.7-6.8) years, respectively (P=0.95). Median OS
was 12.0 (95%CI: 11.0-19.6) vs. 11.0 (95%CI: 9.6-12.7) years, respectively (P=0.17);
5- and 10-year OS rates were 81% vs. 75% and 62% vs. 54% in the doxycycline and penicillin
groups, respectively. Among patients with day 100 hematologic response, there was
a non-statistically significant trend towards improved OS with doxycycline, but no
difference in those without hematologic response: median OS with doxycycline vs. penicillin
was 21.1 vs. 14.6 years in patients with day 100 dFLC <4 mg/dL (P=0.18) Figure 1a.
In patients with normal day 100 FLCr, median OS was NR vs. 12.8 years, respectively
(P=0.10) Figure 1b. There was no difference between the 2 groups when OS was stratified
by Mayo 2012 stage (1-2 vs 3-4), number of involved organs (1 vs. >1), ASCT period
(before or after 2012), or pre-ASCT induction (Yes vs. No).
Image:
Summary/Conclusion: After 13 years of follow up we observed a trend towards improved
OS with doxycycline as post-ASCT prophylaxis in patients who achieve a hematological
response. Prospective studies are needed to elucidate whether doxycycline improves
outcomes in AL amyloidosis and identify the optimal dose, duration, timing, and the
subset of patients likely to benefit.
P885: PROSPECTIVE ASSESSMENT OF MYELOMA TUMOUR BURDEN AND BONE DISEASE USING DW-MRI
AND EXPLORATORY BONE BIOMARKERS
G. Agarwal1,*, G. Nador1, S. Varghese1, H. Getu1, C. Palmer2, C. Rodgers3, C. Pereira2,
G. Sallemi2, K. Partington4, N. Patel4, R. Soundarajan5, R. Mills5, R. Brouwer5, A.
Shah6, D. Peppercorn6, C. Edwards2, M. Javaid2, S. Gooding1, K. Ramasamy1
1Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust;
2The Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences,
University of Oxford, Oxford; 3Department of Clinical Neurosciences, University of
Cambridge, Cambridge; 4Department of Radiology, Oxford University Hospitals NHS Foundation
Trust; 5University of Oxford, Oxford; 6Department of Radiology, Hampshire Hospitals
NHS Foundation Trust, Hampshire, United Kingdom
Background: Key clinical priorities for multiple myeloma (MM) are to reduce tumour
burden and complications, of which lytic bone disease is the major cause of morbidity.
To guide treatment, there is a pressing need for biomarkers that can accurately quantify
MM tumour burden and associated bone disease. To this end, Diffusion-Weighted Magnetic
Resonance Imaging (DW-MRI) and bone turnover markers are promising radiological and
serum metrics, respectively, although their clinical value in prospective assessment
is unclear.
Aims: LOOMIS was a single-centre observational cohort study that evaluated the relative
merits of DW-MRI and bone turnover markers in prospectively assessing tumour burden
and bone disease when added to standard clinical assessment, in patients with MM and
monoclonal gammopathy of undetermined significance (MGUS).
Methods: A total of 67 patients were enrolled (14 newly diagnosed MM, 12 relapsed
MM, 15 smouldering MM, 14 MGUS and 12 healthy volunteers) between March 2018 and March
2020. At baseline (and 6-month follow-up for MM/MGUS), participants had a DW-MRI scan
and serum measurements of established (P1NP, CTX-1, ALP) and exploratory (DKK1, sclerostin,
RANKL:OPG ratio) bone turnover markers. MM/MGUS patients additionally had standard
myeloma bloods and Dual-Energy X-ray Absorptiometry (DXA) at each visit, as correlates
of tumour burden and bone loss. DW-MRI scans were double reported by expert Radiologists
for Apparent Diffusion Coefficient (ADC) measurements of lytic bone lesion(s), and
Myeloma Response Assessment and Diagnosis System (MY-RADS) Response Assessment Category
(RAC) scoring. Patients were classified by International Myeloma Working Group (IMWG)
response criteria as a clinical correlate of therapy response.
Results: On assessment of baseline tumour burden, there was no correlation between
single lesion DW-MRI ADC and serum paraprotein [p>0.05]; however, there was moderate
positive correlation between serum DKK1 and serum paraprotein [r=0.39, p=0.04]. At
follow-up, radiological MY-RADS RAC scoring correlated with conventional IMWG response
criteria [p=0.015]; additionally, longitudinal relative change in serum DKK1 differed
between IMWG-defined therapy responders (37% decrease) and non-responders (15% increase)
[p<0.01]. On baseline assessment of bone loss, there was moderate positive correlation
between serum sclerostin and DXA bone mineral density at femoral neck [r=0.40, p<0.01]
and lumbar spine [r=0.54, p<0.001]. At follow-up, there was a moderate negative correlation
between longitudinal % change in the RANKL:OPG ratio and % change in DXA bone mineral
density at femoral neck [r= -0.45, p<0.01].
Summary/Conclusion: Our prospective trial validates DW-MRI-based MY-RADS RAC scoring
as a qualitative radiological tool to assess therapy response. Whilst previous work
has supported single lytic lesion ADC measurements as a correlate of tumour volume,
we did not find this; small sample size and prior chemotherapy in our study may have
limited our ability to detect this. In comparison, serum bone turnover markers such
as DKK1 may provide a more global measure to quantify myeloma burden both at baseline
diagnosis and longitudinally with therapy. Additionally, our data highlight serum
sclerostin and RANKL:OPG ratio as potential biomarkers to assess and monitor bone
loss. Overall, our study highlights emerging radiological and serum biomarkers of
tumour burden and associated bone loss in MM and MGUS, which merit further exploration
alongside uniformly treated cohorts, to better understand their clinical utility.
P886: AFRICAN AMERICAN PATIENTS WITH SMOLDERING MULTIPLE MYELOMA MAY HAVE A LOWER
RISK OF PROGRESSION COMPARED TO WHITE PATIENTS
T. Akhlaghi1,*, K. Maclachlan2, N. Korde2, S. Mailankody2, A. Lesokhin2, H. Hassoun2,
S. X. Lu2, D. Patel2, U. Shah2, C. Tan2, A. Derkach3, O. Lahoud4, H. J. Landau4, G.
L. Shah4, M. Scordo4, D. J. Chung4, S. A. Giralt4, S. Z. Usmani2, O. Landgren5, M.
Hultcrantz2
1Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside
and West; 2Myeloma Service, Department of Medicine; 3Department of Epidemiology and
Biostatistics; 4Adult Bone Marrow Transplant Service, Department of Medicine, Memorial
Sloan Kettering Cancer Center, New York; 5Myeloma Service, Sylvester Comprehensive
Cancer Center, University of Miami, Miami, United States of America
Background: The incidence of multiple myeloma (MM) and its precursor stages is two
to threefold higher in African Americans (AAs) compared to whites when adjusted for
socioeconomic status, age, and sex. However, there is limited information on whether
racial background affects the risk of progression from smoldering MM (SMM) to MM.
Aims: To assess the effect of race on the progression from SMM to MM.
Methods: Patients with SMM presenting to Memorial Sloan Kettering Cancer Center between
the years 2000 and 2019 and who identified as either AA or white were included in
this retrospective study. Baseline patient and disease characteristics were collected
at the time of diagnosis including laboratory, imaging, and pathology reports. Differences
in distributions of continuous and discrete characteristics were assessed by Kruskal-Wallis
and chi-square tests. Time to progression (TTP) was assessed using the Kaplan-Meier
method with log-rank test for comparisons. Univariate and multivariate Cox proportional
hazard models were used to estimate effects of risk factors on TTP with hazard ratios
(HR) and 95% confidence intervals (CI).
Results: A total of 576 patients were included (70 were AA, 12%). Median follow-up
time was 3 years in AAs and 4 years in whites. Differences in baseline characteristics
between AAs and whites included median age (60 years in AAs [IQR 51-67] vs 64 years
in whites [IQR 56-72], p = 0.01), median hemoglobin level (12.3g/dL in AA [IQR 11.8-13]
vs 12.8g/dL in whites [IQR 11.8-13.9], p = 0.02), and immunoparesis including 1 or
2 uninvolved immunoglobulins (31% and 10% in AAs vs 56% and 27% in whites, p = 0.002).
There was no difference in bone marrow plasma cell percentage (BMPC), M-spike, free
light chain ratio, or Mayo-2018 SMM risk score. AA race was associated with a significantly
decreased risk of progression in the univariate model (HR 0.57, CI 0.34-0.94). In
the multivariate model adjusting for age, sex, and variables associated with an increased
risk of progression in the univariate model (BMPC, M-spike, free light chain ratio,
immunoparesis and low albumin), AA race remained associated with a decreased risk
of progression (HR 0.39, CI 0.16-0.95). Overall, AA patients with SMM had a significantly
(p = 0.027) longer median TTP (9.7 vs 6.2 years), and a lower 2-year (12.6% vs 20.1%)
and 5-year (34% vs 44.6%) progression rate than whites. Because AA patients were younger
at diagnosis, we stratified patients by age group, < 65 vs ≥65 years. In patients
< 65 years, there was no difference in progression rate. In patients aged ≥65 years,
AA patients continued to have a longer TTP than whites (9.8 vs 5.2 years, p = 0.02).
Image:
Summary/Conclusion: In our retrospective single institution experience, AA patients
with SMM had a lower risk of progression to MM compared to whites. Both groups had
similar Mayo-2018 risk scores, however, AA patients had a lower degree of immunoparesis
at baseline. Future studies are needed to better understand if these differences are
explained by differences in disease biology including genomic mechanisms, immune microenvironment,
and systemic immune response.
P887: IN-HOUSE ANTI BCMA CAR-T THERAPY FOR THE TREATMENT OF RELAPSED REFRACTORY MULTIPLE
MYELOMA: PHASE 1 RESULTS.
N. Asherie1,*, S. Kfir-Erenfeld1, B. Avni1, S. Grisariu1, M. Assayag1, T. Sharon1,
E. Lebel2, C. J. Cohen3, M. E. Gatt2, P. Stepensky1
1Bone Marrow Transplantation; 2Hematology, Hadassah Medical Center, Jerusalem; 3The
Laboratory of Tumor Immunology and Immunotherapy, Bar-Ilan University, Ramat Gan,
Israel
Background: B-Cell Maturation Antigen (BCMA) targeted CAR-T cell therapy is a treatment
strategy showing great promise. Recently, anti-BCMA CAR-T cell therapy has been approved
by the FDA for treatment of relapsed/refractory multiple myeloma. However, the major
drawbacks of this treatment are the length of the manufacturing process, limited access
and the excessive cost. Thus, there is an urgent need for development of in-house
manufactured CAR-T cell treatments. Based on favorable pre-clinical results using
our novel CAR-T-anti BCMA construct, in February 2021 we have launched a BCMA-targeted
CAR-T cell based clinical trial treating relapsed refractory (R/R) multipe myeloma
(MM) patients (NCT04720313).
Aims: This phase I-II clinical trial is a dose escalation study. The first goal was
to explore the safety of our product. The phase I first part of the trial consisted
of three escalating cell doses ranging between 150- and 800x10^6 CAR+ cells.
Methods: We enrolled 20 R/R MM patients having failed more than three lines of previous
therapies, to receive our CAR-T-anti BCMA product at escalating doses of 150 x 106
(cohort 1, 6 patients), 450 x 106 (cohort 2, 7 patients) and 800 x 106 (cohort 3,
7 patients) cells. The median age was 62 (range: 44-75) years, with a median time
of 55 months from initial disease diagnosis (range, 8-241) and a median number of
5.5 previous treatment lines (range, 3-13). Eighty percent had bone marrow (BM) involvement
and the majority were categorized as ISS/R-ISS II. The vast majority of patients underwent
previous autologous BM transplantation and were refractory to proteasome inhibitors,
immunomodulatory agents (IMiD) and anti-CD38 antibody, while only 10 patients (50%)
were previously exposed to an anti-BCMA agent (belantamab). Patients in cohort 1 had
a higher rate of extramedullary disease, high LDH and massive BM involvement (>50%
plasma cells in BM biopsy), while patients in cohort 3 had a higher ECOG performance
score and a lower rate of cytogenetic high-risk disease.
Results: At a median follow up of 156 days, overall response was 75% for the entire
cohort with 2/6,1/7 and 1/7 patients not responding in cohorts 1, 2 and 3, respectively.
Six patients achieved MRD negative CR (4 of them in cohort 3), while 3 achieved MRD
positive (or MRD not evaluated) CR and 6 had achieved VGPR (4 of them in cohort 2).
The median PFS for the entire study group was 166 days (range, 24-289); 75 days (range,
24-289), 181 days (range, 61-209) and not reached (range, 20-124) for cohort 1,2 and
3, respectively. The median duration of response (DOR) for the entire study group
was 151 days (range, 32-259). The progression rate in patients exposed to belantamab,
was higher (7/9, 78%) compared to the non-exposed patients (4/11, 36%). On the day
of last follow up, 75% of patients were still alive. Eleven (55%) patients progressed.
Four (25%) patients died due to disease progression. Ninety percent of patients developed
cytokine release syndrome (CRS), mostly on the day of cell infusion (median day 0,
range 0-21), while none suffered from grade 3-4 CRS. None of the patients developed
immune effector cell-associated neurotoxicity syndrome (ICANS).
Summary/Conclusion: The anti-BCMA CART - cell therapy developed at Hadassah Medical
Center proves safe at the three cohorts, with manageable toxicities and evidence of
initial short term favorable responses attesting to its efficacy in heavily pretreated
R/R MM patients. These data are very encouraging and demonstrate the capability of
single center local medical institution to develop, manufacture and deliver in-house
CART.
P888: REPLACING STEROIDS IN TRANSPLANT-INELIGIBLE MULTIPLE MYELOMA: THE PHASE 2 ISATUXIMAB-BORTEZOMIB-LENALIDOMIDE-DEXAMETHASONE
REST STUDY
F. B. Askeland1,2,*, E. Haukås3, T. S. Slørdahl4, D. Schjøll5, A. Lysen1, E. Hermansen6,
F. Schjesvold1,2
1Oslo Myeloma Center, Department of Hematology; 2KB Jebsen Center for B Cell Malignancies,
Oslo University Hospital, Oslo; 3Department of Cancer and Blood Diseases, Stavanger
University Hospital, Stavanger; 4Department of Hematology, St. Olavs Hospital, Trondheim,
Norway; 5Oslo Myeloma Center, Department of Hematology, Oslo University Hospital,
Oslo; 6Department of Hematology, Zealand Iniversity Hospital, Roskilde, Denmark
Background: Standard therapies for newly diagnosed multiple myeloma (NDMM) patient’s
ineligible for autologous stem-cell transplantation (ASCT) have been lenalidomide-dexamethasone
(Rd), bortezomib-lenalidomide-dexamethasone (VRd) or bortezomib-melphalan-prednisone
(VMP). The benefit of adding an anti-CD 38 monoclonal antibody (mAb) to the standard
treatments of VMP and Rd has been demonstrated in two large phase 3 studies (Facon,
2019. Mateos, 2018). Isatuximab is an anti-CD38 mAb approved in combination with pomalidomide-dexamethasone
and carfilzomib-dexamethasone to treat relapsed/refractory MM, and has also shown
clinical responses as combination therapy to treat NDMM (Trudel, 2019).
Corticosteroids have been the backbone of most myeloma targeted therapies since the
discovery of their effectiveness. Due to the continuous treatment paradigm, the cumulative
dose of corticosteroids in patients is substantial. Steroids inflict reduction in
both short- and long-term quality of life and increase patients’ receptiveness for
infections. Infections are one of the main reasons for complications and death during
active first line treatment, and the rate rises with age. Both steroid-free regimens
and regimens with reduced corticosteroids have demonstrated improved safety and similar
efficacy as regimens containing steroids.
Aims: The study will evaluate isatuximab (Isa) in combination with bortezomib (V)
and lenalidomide (R) with minimal dexamethasone (d) as first-line treatment in transplant-ineligible
patients. The primary endpoint is the number of patients who achieve measurable residual
disease negative (Euroflow NGF 10-5) complete response during and/or after 18 cycles
of study treatment. Secondary endpoints include progression free survival, overall
survival, overall response rate (ORR), safety evaluations and patient-reported outcome.
Methods: The REST study is an academic, single arm, open-label, phase 2 study of NDMM
patients ineligible for ASCT. 50 patients will be included and receive Isa-VRd (Isa:10 mg/kg
IV Days 1, 8, 15, 22 during cycle 1, QW cycle 2-18; V: 1,3 mg/m2 SC Days 1,8,15 during
cycle 1-8; R: 25 mg PO Days 1-21 during all cycles; d: 20 mg PO Days 1,8,15,22 only
for the first 2 cycles), all 28-day cycles.
Results: As of January 31, 2022, 17 patients have been enrolled. Baseline characteristics
can be found in Table 1. At data-cutoff, the median number of cycles started by patients
was 3.6 (range 1-8) and the median duration of exposure was 16 (range 3-28) weeks.
8 patients developed 14 grade ≥3 non-hematological adverse eventd (AE), including
infections (n=7), fever (n=1), skeletal pain (n=2), acute renal failure (n=1), peripheral
sensory neuropathy (n=1), diarrhea (n=1) edema (n=1). There were no AEs leading to
definitive treatment discontinuation. At a follow up of 6 months, the ORR was 100%
(15/15) and the ≥very good partial response rate was 53.3% (8/15 patients).
Image:
Summary/Conclusion: Isa-VRd in the transplant ineligible population has a tolerable
safety profile. Isa-VRd is showing encouraging preliminary efficacy in NDMM ineligible
for transplant. Enrollment and follow-up is ongoing.
Acknowledgements: Sanofi sponsors the REST study.
P889: WILL OUTCOME IMPROVE BY TREATING MULTIPLE MYELOMA PATIENTS AT MRD RELAPSE? THE
REMNANT STUDY (RELAPSE FROM MRD NEGATIVITY AS INDICATION FOR TREATMENT)
F. B. Askeland1,2,*, A.-M. Rasmussen1,2, A. Lysen1, E. Haukås3, A. Eilertsen4, M.
Moksnes5, G. Tsykunova6, A. Vik7,8, B. D. Eiken9, J. H. Sørbø10, J. Rolke11, K. O.
Sand12,13,14, R. F. Hallstensen15, L. Myrseth1, T. S. Slørdahl16,17, F. Schjesvold1,2
1Oslo Myeloma Center, Department of Hematology; 2KG Jebsen Center for B Cell Malignancies,
Oslo University Hospital, Oslo; 3Department of Hematology and Oncology, Stavanger
University Hospital, Stavanger; 4Department of Hematology, Akershus University Hospital,
Oslo; 5Cancer and Hematology Center, Vestfold Hospital, Tønsberg; 6Department of Medicine,
Haukeland University Hospital, Bergen; 7Department of Hematology, University Hospital
of North Norway; 8Institute of Clinical Medicine, UiT, The Artic University of Norway,
Tromsø; 9Department of Oncology, Hospital Østfold, Kalnes; 10Medical Department, Levanger
Hospital, Levanger; 11Department of Hematology, Sørlandet Hospital, Kristiansand;
12Department of Medicine, Ålesund Hospital; 13Department of Reasearch and Innovation,
Møre og Romsdal Hospital Trust; 14Department of Health Sciences in Ålesund, Norwegian
University of Science and Technology, Ålesund; 15Department of Hematology, Nordland
Hospital, Bodø; 16Department of Hematology, St. Olavs Hospital; 17Norwegian University
of Science and Technology (NTNU), Trondheim, Norway
Background: Although many new treatment modalities have become available, multiple
myeloma (MM) is nevertheless considered an incurable disease.
In recent years, the importance of achieving measurable residual disease (MRD) negativity
is becoming more clearly defined in patients with MM. There is solid evidence that
bone marrow MRD negativity is one of the strongest prognostic factors, and deeper
responses correlate with increasingly favorable outcomes. The deepest and most durable
responses are achieved in the first line (1.L) of treatment.
Aims: The REMNANT study will evaluate if treating MRD relapse after 1.L therapy prolongs
progression free survival (PFS) and overall survival (OS) compared to starting treatment
at biochemical relapse/progressive disease (PD) in MM patients.
Methods: The REMNANT study is an academic, multicenter, open-label, randomized phase
II/III study of newly diagnosed (ND) MM patients eligible for autologous stem cell
transplant (ASCT). Few exclusion criteria apply and patients with kidney failure and
amyloidosis can be enrolled.
391 NDMM patients (age 18-75 years) eligible for ASCT will be enrolled in the phase
II part of the study and receive standard of care (SOC) 1.L treatment according to
Norwegian national guidelines; four pre-transplant induction and four post-transplant
consolidation cycles of bortezomib, lenalidomide and dexamethasone (VRd). After induction,
patients will undergo tandem or single transplant, depending on toxicity and response
to first transplant.
Following 1.L treatment 176 patients who are ≥ complete response (CR) and MRD negative
(NGF Euroflow) will be enrolled in part 2 of the study. Patients will be randomized
in a 1:1 ratio to receive second line treatment at MRD relapse in arm A or at PD in
arm B. Randomization will be stratified by R-ISS stage at diagnosis and single versus
tandem transplant. At loss of MRD negativity in arm A or at PD in arm B, second line
treatment will be carfilzomib, dexamethasone and daratumumab until progressive disease.
In part 2 we will compare different methods for measuring MRD; NGF Euroflow, Next
Generation Sequencing (NGS), mass spectrometry and PET-CT. We will evaluate how the
different methods compare when it comes to sensitivity and how they correlate to outcome
(PFS and OS). The microbiota composition has shown to impact outcome in myeloma patients.
Therefore, we will investigate how the microbiota at diagnosis differs in patients
who achieve MRD negativity and those who still are MRD positive after treatment.
Results: We will present updated results from part 1 of the study. As of February
16th 2022, 136 patients have been enrolled in part 1.
Twenty-six patients have been evaluated for MRD status post consolidation. Thirteen
patients (50%) were ≥CR and MRD negative and have been enrolled in part 2 of the study.
In Table 1 baseline characteristics and results are presented.
Image:
Summary/Conclusion: Part 1 of the REMNANT study will clarify the overall response
rate and depth of response measured by MRD, after SOC Norwegian 1.L treatment
Part 2 will determine if treating patients at MRD relapse, when tumor burden is lower,
will improve outcome.
P890: THE OUTCOME OF SECOND PRIMARY MALIGNANCIES DEVELOPING IN MM PATIENTS
I. Avivi1,*, D. H vesole2, J. Dávila Valls3, L. Usnarska-Zubkiewicz4, V. Milunovic5,
B. B. Bogumiła Osękowska6, A. Kopińska7, M. Gentile8, B. P. MARTÍNEZ9, P. Robak10,
E. crusoe11, R. LUIS GERARDO12, M. Gajewska13, V. Gergely14, M. Delforge15, Y. Cohen16,
G. Alessandro17, C. peña18, C. Shustik19, G. Mikala20, K. Žalac21, A. H. Denis22,
B. Peter23, K. Weisel24, J. martinez lopez25, A. Waszczuk-Gajda26, M. Krzystanski27,
A. Jurczyszyn28
1Head of Hematology Division, Tel Aviv Medical Center, Tel AVIV, Israel; 2John Theurer
Cancer Center, Hackensack University Medical Center, Hackensack, United States of
America; 3Complejo Asistencial de Avila, Complejo Asistencial de Avila, Avila, Spain;
4Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw,
Poland; 5Division of Hematology, Clinical Hospital Merkur, Zagreb, Croatia; 6Pomeranian
Medical University in Szczecin, Pomeranian Medical University in Szczecin, Szczecin;
7Department of Haematology, Oncology and Internal Diseases, University Clinical Centre,
Medical University of Warsaw, Warsaw, Poland; 8Hematology Unit, AO Cosenza, Cosenza,
Italy; 9(University Hospital of Salamanca, Cancer Research Center-IBMCC, Salamanca,
Spain; 10Medical University of Lódź, Medical University of Lódź, Lódź, Poland; 11:
Federal University Of Bahia, University Hospital and Rede D’or Oncologia, Bahina,
Brazil; 12Clinic Barcelona Hospital Universitari, Clinic Barcelona Hospital Universitari,
Barcelona, Spain; 13Department of Hematology and Bone Marrow Transplantation, Silesian
Medical University, Katowice, Poland, Warsaw, Poland; 14Department of Internal Medicine
and Haematology, Semmelweis University, Budapest, Hungary; 15Department of Hematology,
UZ Leuven, Leuven, Belgium; 16Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv,
Israel; 17Hematology, Department of Medical Science, Surgery and Neuroscience, University
of Siena, Siena, Italy; 18Sección Hematología, Hospital del Salvador, Santiago, Chile;
19Hematology Department, MUHC - McGill University Health Centre, montreal, Canada;
20Department of Hematology and Stem Cell Transplantation, South-Pest Central Hospital,
Budapest, Hungary; 21Department of hematology of Clinics for Internal Medicine, University
Hospital Center ‘‘Sestre Milosrdnice’’, Zagreb, Croatia; 22N Ireland Centre for Stratified
Medicine, Ulster University, Londonderry, Ireland; 23Department of Medicine, Warren
Alpert Medical School, Brown University, Rhode Island, United States of America; 24:
University Medical-Center Hamburg-Eppendorf, Hamburg, Germany; 25Nieves Lopez-Muñoz
Hospital, Madrid, Spain; 26Department of Hematology, Oncology and Internal Diseases,
Warsaw Medical University, Warsaw; 27Department of Hematology; 28Department of Hematology,
Jagiellonian University Medical College, crakow, Poland
Background: The continuing improvement in response rates and overall survival (OS)
of multiple myeloma (MM) patients exposed to the cumulative effect of multiple anti-MM
agents including IMiDs, PIs, mAbs and cytotoxic chemotherapy, has resulted in an increased
risk for second primary malignancies (SMPs).
There is minimal detailed information regarding the clinical characteristics of patients
who develop SPMs, risk factors predicting development, time to onset, SPM’s management
and outcomes in patients treated with novel agents outside clinical trials.
Aims: To investigate the characteristics and outcome of SPMs reported in MM patients
and define impact of SPM diagnosis on MM management and outcome.
Methods: A multicenter international ‘real-world’ retrospective analysis, reviewing
MM databases of 25 participating centers and analyzing the characteristics and outcome
of SPM reported in patients that were treated with novel-based induction. In addition,
the impact of SPM diagnosis on MM management and outcome are reported.
Results: Two hundred and one consecutive MM patients, experiencing SPM ≥ 6 months
since initiation of first line therapy were included in the analysis. Clinical characteristics
included the following: 64% were male, median age at 66±10.57 years, 10% with a prior
history of cancer. 36% and 25% of evaluable cases presented with high ISS-3 and high-risk
cytogenetics. 87(43%) underwent an autologous hematopoietic cell transplantation and
51(27%) received maintenance therapy. Median number of lines of therapy prior to SPM
defection was 2 (1-7).
With a median follow-up of 50± 45 months since MM diagnosis, 115 patients (57%) were
diagnosed with solid cancers: colorectal cancer (n=20, 17.4%), lung (n=18, 15.6%),
breast (n=14, 12%), prostate (n=8, 7%), melanoma (n=10, 9%), pancreas (n=10, 9%),
bladder (n=7, 6%) and gastric (n=7, 6%), skin cancer (n= 34, 16.9%) and cancer of
unknown origin (n=9, 4.5%). 46 patients (22.9%) developed hematological cancers, including
lymphoma and MDS/AML; reported in 7(6%) and 27 (13.4%) cases respectively. 79% (n=158)
were actively treated for SPM and 39.5% responded to therapy.
Forty Eight percent (n=98) of the patients were concurrently receiving an anti-MM
therapy at the time of SPM diagnosis. MM treatment remained unchanged in 27 patients,
discontinued and not substituted with any other anti-MM therapy until MM progression
in 59 patients and changed to a new anti-MM therapy in 12 patients.
Within a median follow-up period of 112 months since MM diagnosis and 31 months since
SPM detection, 50.5% (n-101) have died; 32 due to solid cancer (32/101, 28%), 11 due
to hematological cancer (11/46, 24%) and 13 (6%) due to MM progression. Cause of death
was not available for 45 cases. Median OS since MM diagnosis and since SPM detection
were 63 months and 8.5 months, respectively (Figure 1). (Detailed analysis regarding
causes of death and risk factors to be presented at the meeting).
Image:
Summary/Conclusion: With the continuing improvement in OS, a higher proportion of
MM patients are likely to develop SPM. OS since SPM diagnosis is shortened, approaching
8.5 months only, suggesting that the SPMs, originating in this scenario, are generally
aggressive. Surveillance for early detection and treatment of SPMs are warranted.
Further studies are necessary to determine the factors associated with the development
of SPM to avoid precipitating agents especially in patients with lower risk disease
since these individuals should have extended survival that may be substantially shortened
by the development of treatment-related SPM.
P891: IXAZOMIB, POMALIDOMIDE AND DEXAMETHASONE (IXPD) IN RELAPSED OR REFRACTORY MULTIPLE
MYELOMA (RRMM) CHARACTERIZED WITH HIGH-RISK CYTOGENETICS. IFM 2014-01
A. Bobin1, S. Manier2, J. De Keizer1, L. Karlin3, A. Perrot4, C. Hulin5, D. Caillot6,
C. Mariette7, P. Lenain8, V. Richez9, M. Tiab10, C. Touzeau11, A. Jaccard12, O. Decaux13,
C. Araujo14, K. Belhadj15, L. Benboubker16, C. Déal17, M. Macro18, L. Vincent19, B.
Arnulf20, B. Bareau21, T. Braun22, C. Calmettes23, D. mamoun24, H. Zerazhi25, H. Demarquette26,
P. Feugier27, C. Fohrer-sonntag28, S. Godet29, M.-O. Petillon2, H. Avet-Loiseau4,
X. Leleu1,*
1CHU de Poitiers, Poitiers; 2chu de lille, lille; 3CHU de Lyon, Lyon; 4CHU de Toulouse,
Toulouse; 5CHU de Bordeaux, Bordeaux; 6CHU de Dijon, Dijon; 7CHU de Grenoble, Grenoble;
8CHU de Rouen, Rouen; 9CHU de Nice, Nice; 10CH la Roche sur Yon, La Roche Sur Yon;
11CHU de Nantes, Nantes; 12CHU de Limoges, Limoges; 13CHU de Rennes, Rennes; 14CH
de Bayonne, Bayonne; 15CHU de Créteil-APHP, Créteil; 16CHU de Tours, Tours; 17Intergroupe
Francophone du Myélome, Paris; 18CHU de Caen, Caen; 19CHU de Montpellier, Montpellier;
20Hopital Saint Louis, Paris; 21Hopital Privé Sevigne, Cesson Sevigne; 22Hopital Avicenne,
Bobigny; 23CH de Périgueux, Périgueux; 24CHU de Angers, Angers; 25CH de Avignon, Avignon;
26CH de Dunkerque, Dunkerque; 27CHRU de Nancy, Nancy; 28CHU de Strasbourg, Strasbourg;
29CHU de Reims, Reims, France
Background: High risk (HR) cytogenetics remains of poor prognosis, particularly in
the RRMM setting. IFM 2010-02, Pd in advanced HR RRMM, had demonstrated limited activity
with no addition of a proteasome inhibitor (PI), and a good safety profile. We hypothesized
that addition of Ixazomib (oral PI at increased dose density) to Pd (IxPd) in HR RRMM
would improve convenience, thus adherence to treatment, and in parallel improve efficacy
with no increased toxicity.
Aims: We aimed to determine the efficacy and safety profile of the association ixazomib,
pomalidomide and dexamethasone HR RRMM.
Methods: Eligible patients had a RRMM in L2 refractory to lenalidomide but not to
pomalidomide and ixazomib. HR was defined by presence of either del(17p) and/or t(4;14)
at diagnosis or study entry. Patients received 17 induction cycles, 21-days long,
consisting of ixazomib 3mg/day (d 1, 4, 8 and 11), pomalidomide 4mg/day (d1 to 14)
and weekly dexamethasone, followed by a maintenance phase of 28-day cycles with ixazomib
4mg/day (d 1, 8 and 15) and pomalidomide 4mg/day (d 1 to 21), until progression. The
primary endpoint was time to progression (TTP). The number of patients to be recruited
was initially calculated based on an expected doubling of the median TTP obtained
in IFM 2010-02 with Pd in either 2 HR RRMM population, but the number was recalculated
due to difficulties in recruitment to demonstrate an expected doubling of the median
TTP from 3 months, as a whole. No statistical comparison can be done across HR groups
given that the study was powered to analyze the 2 groups as a whole and not separately
with 26 patients.
Results: Twenty-six patients were enrolled in the study. Median age was 72 years (Interquartile
range: 67-78). Ten patients presented with del(17p) and twelve patients with t(4;14).
All patients were refractory to lenalidomide.
With a median follow-up time of 9.8 months, the median TTP was 9.7 months (95% confidence
interval (CI) 4.4;-). The median OS was 12.2 months (CI95% 11.1;-). The ORR and ≥VGPR
rate was 15 (58%) pts and 7 pts (27%) in the study as a whole. The ≥VGPR rate was
30% and 33% in del(17p) and t(4;14), respectively. No CR was observed with IxPd in
our series.
The most common adverse events (≥10% occurrence) were neutropenia (n=13), thrombocytopenia
(n=12), asthenia (n=9), anemia (n=7), rash (n=4), diarrhea (n=3), dizziness (n=3),
general physical health deterioration (n=3), muscle spasms (n=3), peripheral oedema
(n=3), pyrexia (n=3). Ten SUSARs were declared with no modification to the safety
profile of ixazomib and pomalidomide according to the independent data monitoring
committee.
Summary/Conclusion: The study IFM 2014-01/IxPd in HR L2 RRMM met its primary end point
objective since the median TTP was superior to 3 months, the median TTP reported in
IFM2010-02 as a whole, with the addition of Ixazomib to Pomalidomide and dexamethasone
in this population characterized with a very poor outcome. This data confirms the
importance of proteasome inhibitors in HR RRMM. This phase 2 study needs confirmation
in a larger cohort.
P893: CARFILZOMIB IN COMBINATION WITH LENALIDOMIDE AND DEXAMETHASONE (KRD) AS SALVAGE
THERAPY FOR MULTIPLE MYELOMA PATIENTS: ITALIAN, MULTICENTER, RETROSPECTIVE EXPERIENCE
OUTSIDE OF CLINICAL TRIALS
E. A. Martino1,*, C. Conticello2, E. Zamagni3, V. Pavone4, S. Palmieri5, M. Musso6,
P. Tacchetti7, A. Mele4, L. Catlano8, E. Vigna1, A. Bruzzese9, F. Mendicino1, C. Botta10,
D. Vincelli11, G. Farina12, M. Barone13, C. Cangialosi14, K. Mancuso15, I. Rizziello15,
S. Rocchi15, A. P. Falcone16, G. Mele17, G. Reddiconto18, B. Garibaldi19, E. Iaccino20,
G. Tripepi21, F. Di Raimondo22, P. Musto23, A. Neri24, M. Cavo3, F. Morabito25, M.
Gentile1
1Azienda Ospedaliera di Cosenza, COSENZA; 2University of Catania, Catania; 3Seràgnoli
Institute of Hematology and Medical Oncology, University of Bologna, Bologna; 4Hospital
Card. G. Panico, Tricase; 5Ospedale Cardarelli, Napoli; 6Dipartimento Oncologico,
La Maddalena, Palermo; 7Istituto di Ematologia “Seràgnoli”, Bologna; 8AUOP “Federico
II”, Napoli; 9Azienda Ospedaliera of Cosenza, Cosenza; 10Internal Medicine and Medical
Specialties, University of Palermo, Palermo; 11Great Metropolitan Hospital “Bianchi-Melacrino-Morelli”,
Reggio Calabria; 12AORN “Sant’Anna e San Sebastiano”, Caserta; 13”Tortora” Hospital,
Pagani; 14A. O. Ospedali Riuniti Villa Sofia-Cervello, Palermo; 15IRCCS Azienda Ospedaliero-Universitaria
di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna; 16IRCCS Casa Sollievo della
Sofferenza, San Giovanni Rotondo; 17Hematology Unit, Brindisi; 18Hospital Vito Fazzi,
Lecce; 19Azienda Policlinico-OVE, University of Catania, Catania; 20University “Magna
Graecia” of Catanzaro, Catanzaro; 21National Research Institute of Biomedicine and
Molecular Immunology, Reggio Calabria; 22Hematology Section, University of Catania,
Catania; 23”Aldo Moro” University School of Medicine, Bari; 24Fondazione IRCCS Ca’
Granda, Ospedale Maggiore Policlinico, Milano; 25Biotechnology Research Unit, AO di
Cosenza, Cosenza, Italy
Background: Carfilzomib in combination with lenalidomide, and dexamethasone (KRd)
have been approved for the treatment of relapsed and refractory multiple myeloma (RRMM)
based on ASPIRE clinical trial. Efficacy and safety of the triplet outside clinical
trial are still the object of investigation by many groups to confirm ASPIRE results
in the setting of RRMM treated in real-life who don’t meet clinical trial restrictive
inclusion criteria.
Aims: The aim of the present study is to investigate the efficacy and safety of KRd
regimen in the setting of RRMM treated in real-life.
Methods: Here we report a retrospective multicenter analysis of 600 RRMM patients
treated with KRd between December 2015 and December 2018
Results: The median age at KRd start was 64 years (range 33-85) and the median number
of previous therapies was 2 (range 1-11). After a median number of 11 KRd cycles,
the ORR was 79.9%. The median PFS was 22 months and the 2-year probability of PFS
was 47.6%. At multivariate analysis advanced ISS (III), and high-risk cytogenetic
were significantly associated with shorter PFS. The median OS was 34.8 months; the
2-year probability of OS was 63.5% At multivariate analysis creatinine clearance<30 ml/min,
more than 1 line of previous therapy, and high-risk FISH were significantly associated
with poor prognosis. After a median follow-up of 16 months (range 1-50), 259 withdrew
from therapy. The main reason for discontinuation was progressive disease (81.8%).
Seventy-four patients (12.3%) discontinued therapy for toxicity. The most frequent
side effects were hematological (anemia 49.3%, neutropenia 42.7%, thrombocytopenia
42.5%) and cardiovascular (hypertension 14.5%, heart failure 2.5%, arrhythmias 3.6%).
Summary/Conclusion: Our study confirms the safety and efficacy of KRd in the real-life
setting of RRMM patients and encourages its use in clinical practice.
P894: A PHASE II TRIAL TO EVALUATE THE EFFICACY OF DARATUMUMAB WITH DCEP IN RELAPSED/REFRACTORY
MULTIPLE MYELOMA PATIENTS WITH EXTRAMEDULLARY DISEASE AFTER BORTEZOMIB BASED TREATMENT
J. M. Byun1,*, C.-K. Min2, K. Kim3, S.-M. Bang4, J.-J. Lee5, J. S. Kim6, S.-S. Yoon1,
Y. Koh1
1Seoul National University Hospital; 2Seoul St Mary’s Hematology Hospital; 3Samsung
Medical Center, Seoul; 4Seoul National University Bundang Hospital, Seongnam; 5Chonnam
National University Hwasun Hospital, Hwasun; 6Yonsei University College of Medicine,
Seoul, South Korea
Background: Extramedullary multiple myeloma (EMM) is an aggressive subentity of multiple
myeloma (MM), characterized by the ability of a subclone to thrive and grow independent
of the bone marrow microenvironment, resulting in a high-risk state associated with
increased proliferation, evasion of apoptosis and treatment resistance. Despite improvement
in survival for most patients with MM over recent decades, outcomes are generally
poor when EMM develops. Thus, better understanding of the disease and more innovative
therapeutic approaches are needed.
Aims: To study the efficacy and safety of daratumumab in combination with dexamethasone,
cyclophosphamide, etoposide and cisplatin (DCEP)
Methods: This was a multi-center, prospective phase II study (ClinicalTrials.gov identifier:
NCT04065308). A total of 33 patients older than 19 years with multiple myeloma according
to IMWG, relapsed/refractory to bortezomib, and with EMM (short-axis ≥1cm by CT or
PET-CT) were enrolled. As shown in the Figure, patients received 3 cycles of daratumumab
in combination with DCEP, followed by daratumumab maintenance. The primary objective
was complete response rate after 3 cycles of daratumumab with DCEP. The secondary
objectives included: 1) overall response rates; 2) progression free survival (PFS);
3) overall survival; and 4) safety and toxicity profiles. Here we present the interim
data of 24 patients.
Results: The median age at MM diagnosis was 57 years (range 34-71) and 79.2% patients
had EMM at diagnosis. There were 10 patients with t(4;14), 4 patients with del(17p)
and 6 patients with t(14;16). There were 6 patients (25%) with ISS I disease, 10 (41.7%)
with ISS II disease, and 8 (33.3%) with ISS III disease. The median number of prior
lines of treatment was 3 (range 1-5), with all patients being exposed to bortezomib
prior to enrollment. Also, 70.8% patients had prior exposure to carfilzomib, 87.5%
to lenalidomide, and 50% to pomalidomide.
There were 17/24 (70.8%) patients who completed 3 cycles: 3 showed CR; 2 showed VGPR;
11 showed PR and 7 with progressive disease as shown in the Figure. Six patients were
able to complete the study protocol. For all patients the median PFS was 4 months,
but for those who completed the study protocol the median PFS was not reached and
all but 1 remained in remission until the data cut-off in 2022-Jan-31. Patients achieving
PR or better response after cycle 3 showed significantly better PFS (6 vs 2 months,
p<0.001) compared to those who didn’t.
Most patients (22/24) underwent cyclophosphamide/etoposide/cisplatin dose reduction
by 30% at cycle 1. One patient required further dose reduction at cycle 2. There was
1 patient who went off trial at D15 of cycle 1 according to attending physician’s
decision and 2 patients who had to skip D22 daratumumab during cycle 1 due to thrombocytopenia.
No patients required dexamethasone dose adjustments. During cycle 1, 12.5% showed
anemia ≥grade 3, 25% showed thrombocytopenia ≥grade 3, 4.2% showed lymphopenia ≥grade
3, and 4 (16.7%) events of febrile neutropenia. During cycle 3, 5.3% showed anemia
≥grade 3, 10.5% showed thrombocytopenia ≥grade 3, while 15.8% showed lymphopenia ≥grade
3. The most common non-hematological adverse events was nausea (20.8%).
Image:
Summary/Conclusion: In conclusion, daratumumab in combination with DCEP showed overall
response of 66.7% (CR 25%), and durable remission in 20.8% of the enrolled patients.
P895: DARATUMUMAB, BORTEZOMIB, AND DEXAMETHASONE (D-VD) VERSUS BORTEZOMIB AND DEXAMETHASONE
(VD) ALONE IN CHINESE PATIENTS WITH RELAPSED OR REFRACTORY MULTIPLE MYELOMA (RRMM):
UPDATED ANALYSIS OF LEPUS
W. Fu1,*, W. Li2, J. Hu3, G. An4, Y. Wang5, C. Fu6, L. Chen7, J. Jin8, X. Cen9, Z.
Ge10, Z. Cai8, T. Niu11, M. Qi12, X. Gai13, Q. Li13, W. Liu14, W. Liu13, X. Yang14,
X. Chen14, J. Lu15,16
1Shanghai Changzheng Hospital, Shanghai; 2The First Hospital of Jilin University,
Jilin; 3Fujian Medical University Union Hospital, Fujian Institute of Hematology,
Fujian Provincial Key Laboratory of Hematology, Fujian; 4State Key Laboratory of Experimental
Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of
Medical Sciences and Peking Union Medical College; 5Department of Hematology, Tianjin
Medical University Cancer Institute and Hospital, National Clinical Research Center
for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research
Center for Cancer, Tianjin; 6The First Affiliated Hospital of Soochow University;
7The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital,
Jiangsu; 8The First Affiliated Hospital of Zhejiang University, College of Medicine,
Zhejiang; 9Peking University First Hospital, Beijing; 10Zhongda Hospital Southeast
University, Jiangsu; 11Department of Hematology, West China Hospital Sichuan University,
Sichuan, China; 12Janssen Research & Development, LLC, Spring House, PA, United States
of America; 13Janssen Research & Development, LLC, Beijing; 14Janssen Research & Development,
LLC, Shanghai; 15Peking University People’s Hospital, National Clinical Research Center
for Hematologic Disease, Beijing; 16Collaborative Innovation Center of Hematology,
Soochow, China
Background: D-Vd prolonged progression-free survival (PFS) and induced deeper and
more durable responses versus Vd alone in patients with RRMM in the global phase 3
CASTOR study (Weisel K, et al. ASH 2019. Abstract 3192). Results from a prespecified
interim analysis of the phase 3 LEPUS study (median follow-up, 8.2 months) showed
significant clinical benefit with D-Vd versus Vd in Chinese patients with RRMM and
a safety profile that was generally consistent with that of CASTOR (Lu J, et al. Clin
Lymphoma Myeloma Leuk 2021. 21[9]:e699-e709).
Aims: To examine updated efficacy of D-Vd versus Vd in LEPUS at a median follow-up
of 25.1 months.
Methods: In the randomized, multicenter, phase 3 LEPUS study (NCT03234972), Chinese
patients with RRMM and ≥1 prior line of therapy were randomized 2:1 to 8 cycles (21
days/cycle) of Vd (bortezomib 1.3 mg/m2 SC on Days 1, 4, 8, and 11; dexamethasone
20 mg PO/IV on Days 1, 2, 4, 5, 8, 9, 11, and 12) ± daratumumab (16 mg/kg IV once
weekly in Cycles 1-3, every 3 weeks in Cycles 4-8, and every 4 weeks thereafter until
disease progression). PFS was the primary endpoint.
Results: A total of 211 patients were randomized (D-Vd, n = 141; Vd, n = 70), with
a median age of 61 (range, 28-82) years. Patients received a median of 2 (range, 1-11)
prior lines of therapy; 79.1% of patients previously received bortezomib, 26.5% were
refractory to lenalidomide, and 64.0% were refractory to their last prior line of
therapy. At a median follow-up of 25.1 months, D-Vd prolonged PFS versus Vd alone
(median: 14.8 vs 6.3 months; hazard ratio [HR], 0.35; 95% confidence interval [CI],
0.24-0.51; P <0.00001). The PFS benefit of D-Vd versus Vd was maintained across subgroups,
including patients with prior bortezomib (HR, 0.36; 95% CI, 0.25-0.53), patients with
high-risk cytogenetics (HR, 0.41; 95% CI, 0.23-0.71), and patients who were refractory
to last prior line of therapy (HR, 0.42; 95% CI, 0.27-0.65). Overall response rate
(ORR; 84.7% vs 66.7%; P = 0.00314) and rates of very good partial response or better
(71.5% vs 34.9%; P <0.00001) and complete response or better (40.1% vs 14.3%; P =
0.00016) were higher with D-Vd versus Vd. Median time to first response was numerically
shorter (0.79 months vs 1.23 months) and median duration of response was numerically
longer (16.8 months vs 9.0 months) with D-Vd versus Vd. The estimated 18-month overall
survival rate was 78.8% in the D-Vd group and 65.8% in the Vd group.
Summary/Conclusion: In this updated analysis of LEPUS, D-Vd continued to demonstrate
substantial efficacy benefits versus Vd in terms of PFS, ORR, and depth of response.
These data further support the use of D-Vd as a standard of care in Chinese patients
with RRMM.
P896: DEVELOPMENT AND VALIDATION OF A FRAILTY PREDICTION MODEL MORE SUITABLE FOR MULTIPLE
MYELOMA IN CHINESE PATIENTS: THE TM FRAILTY SCORE.
Y. Chen1, J. Gu1, B. Huang1, J. Liu1, X. Li1, J. Li1,*
1Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University,
Guangzhou, China
Background: Frailty is associated with adverse clinical outcomes in patients with
multiple myeloma (MM). Accurate evaluation of frailty is critical when choosing appropriate
chemotherapy regimens for elderly patients. Currently, the IMWG geriatric assessment
(GA) is widely used to assess tolerance to chemotherapy for elderly patients. However,
the applicability of the IMWG GA in the Chinese MM population is limited.
Aims: To developed a new frailty model that is more suitable for Chinese patients
with MM.
Methods: The applicability of the IMWG GA was evaluated in 167 consecutive patients
diagnosed with MM from June 2019 to September 2021 at the First Affiliated Hospital
of Sun Yat-sen University. Eight comprehensive geriatric assessment (CGA) domains
were analyzed in 135 of these patients to screen items most closely associated with
the outcomes, including grade ≥ 3 adverse events (AEs), treatment discontinuation,
TTP, and OS. A new frailty prediction model was developed, and its fitness and predictive
performance were compared to those of the IMWG GA.
Results: The patients in the IMWG GA Frail group had a higher rate of grade ≥3 AEs,
a higher rate of treatment discontinuation, and a greater risk of death than those
in the Fit group (P<0.05). No significant differences were observed in patients in
the Int-fit group who experienced chemotherapy AEs, treatment discontinuation, and
TTP compared with the Fit group. By screening items in all 8 CGA domains, multivariate
analysis confirmed that the Timed Up and Go test(TUG) and the Mini Nutritional Assessment
Short-Form(MNA-SF) were independent prognostic factors for grade ≥3 AEs and OS. We
found that the TUG and MNA-SF scores could stratify the risk of grade ≥ 3 AEs in the
IMWG GA Int-fit group (P < 0.05). After MNA-SF was added to construct the IMWG GA
Plus model, Harrell’s concordance index (C-index) of the IMWG GA Plus model for the
prediction of grade ≥3 AEs increased from 0.662 to 0.701. The C-index for treatment
discontinuation rose from 0.636 to 0.669. We further combined the TUG test and the
MNA-SF to construct the TM frailty scoring system. The KM curve analysis showed that
the TM frailty score could well distinguish the risk of AEs between the Fit and Frail
groups (P<0.05). The C-index of the TM frailty score was 0.741, 0.690, and 0.702 for
grade ≥ 3 AEs, treatment discontinuation, and OS, respectively, suggesting good predictive
ability, which was higher than that of the IWMG GA (C-index=0.662, 0.636 and 0.631,
respectively) and the IMWG GA Plus model (C-index=0.701, 0.656 and 0.618, respectively).
Image:
Summary/Conclusion: A novel scoring system—the TM frailty score—was developed by combining
the TUG (T) and MNA-SF (M). The TM frailty score is more appropriate than the IMWG
GA for evaluating frailty in the Chinese population.
P897: UPDATED RESULTS FROM THE ONGOING PHASE 1 STUDY OF ELRANATAMAB, A BCMA TARGETED
T-CELL REDIRECTING IMMUNOTHERAPY, FOR PATIENTS WITH RELAPSED OR REFRACTORY MULTIPLE
MYELOMA
A. Dalovisio1,*, N. Bahlis2, N. Raje3, C. Costello4, B. Dholaria5, M. Solh6, M. Levy7,
M. Tomasson8, H. Dube9, M. Damore9, S. Jiang10, C. Basu10, A. Skoura11, E. Chan12,
S. Trudel13, A. Jakubowiak14, M. Chu15, C. Gasparetto16, M. Sebag17, A. Lesokhin18
1Department of Hematology and Oncology, Ochsner Health, Jefferson, United States of
America; 2Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada;
3Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston; 4Moores
Cancer Center, University of California San Diego, La Jolla; 5Vanderbilt University
Medical Center, Vanderbilt-Ingram Cancer Center, Nashville; 6Blood and Marrow Transplant
Group of Georgia, Northside Hospital, Atlanta; 7Department of Medical Oncology, Baylor
Scott and White Health, Dallas; 8Holden Comprehensive Cancer Center, University of
Iowa, Iowa City; 9Oncology Research and Development; 10Early Clinical Development,
Pfizer, San Diego; 11Early Clinical Development, Pfizer, Collegeville; 12Oncology
Research and Development, Pfizer, South San Francisco, United States of America; 13Princess
Margaret Cancer Centre, University Health Network, Toronto, Canada; 14Department of
Medicine, University of Chicago Medical Center, Chicago, United States of America;
15Cross Cancer Institute, Edmonton, Canada; 16Department of Medicine, Duke University
Cancer Institute, Durham, United States of America; 17Cedars Cancer Center, McGill
University Health Center, Montreal, Canada; 18Division of Hematology and Oncology,
Memorial Sloan Kettering Cancer Center/Weill Cornell Medical College, New York, United
States of America
Background: Elranatamab (PF-06863135), a bispecific molecule that engages B-cell maturation
antigen (BCMA) on multiple myeloma (MM) and CD3 on T-cells, induces targeted proliferation
and activation of T cells to redirect the immune response against MM.
Aims: The Phase 1 study, MagnetisMM-1 (NCT03269136), aims to characterize the safety,
pharmacokinetics (PK), pharmacodynamics and efficacy of elranatamab as a single agent
or in combination with immunomodulatory agents for patients (pts) with relapsed or
refractory MM.
Methods: After informed consent, elranatamab was given subcutaneously weekly or every
2 weeks (Q2W) at doses from 80 to 1000µg/kg. A subset of pts received a single priming
dose (600µg/kg or 44mg equivalent) followed 1 week later by the recommended Phase
2 dose (RP2D; 1000µg/kg or 76mg equivalent) thereafter. Treatment-emergent adverse
events (TEAEs) were graded by Common Terminology Criteria for Adverse Events (v4.03)
and cytokine release syndrome (CRS) by American Society for Transplantation and Cellular
Therapy criteria. PK, cytokine and soluble BCMA profiling, and lymphocyte subset analyses
were performed. Response was evaluated by International Myeloma Working Group (IMWG)
criteria. Minimal residual disease (MRD) was assessed by next generation sequencing
at a sensitivity of 1×10-5 in accordance with IMWG criteria.
Results: A total of 55 pts received elranatamab monotherapy at doses ≥215µg/kg as
of 1-Nov-2021. Median age was 64 years (range 42-80), 27% of pts were Black/African
American or Asian, and 27% had high risk cytogenetics at baseline. Median number of
prior lines of therapy was 6 (range 2-15), 91% were triple-class refractory, 22% received
prior BCMA-targeted therapy, and 56% had prior stem cell transplant. The most common
TEAEs (all causality) were CRS, neutropenia, anemia, injection site reaction, and
lymphopenia. With a single priming dose and premedication, the incidence of CRS at
the RP2D was 67% and divided equally between Grade 1 and 2, with no events greater
than Grade 2. PK exposure was dose dependent, and elranatamab 1000µg/kg Q2W achieved
exposure associated with anti-myeloma activity. Elranatamab therapy induced peripheral
T-cell proliferation with a median time to response of 36 days (range 7-73), and the
level of soluble BCMA decreased with disease response. With a median follow up of
8.1 months (range 0.3-21) and including only IMWG confirmed responses, overall response
rate (ORR) was 64% (95% CI 50-75%), and 31% of pts achieved complete response or better.
For responders (n=35), the probability of being event free at 6 months was 91% (95%
CI 73-97%). Single-agent elranatamab induced durable clinical and molecular responses,
and updated results including serial MRD assessment will be presented.
Summary/Conclusion: Elranatamab demonstrates a manageable safety profile and achieves
durable clinical and molecular responses for pts with relapsed or refractory MM.
P898: COMPARISON OF TECLISTAMAB WITH BELANTAMAB MAFODOTIN IN PATIENTS WITH TRIPLE-CLASS
EXPOSED RELAPSED/REFRACTORY MULTIPLE MYELOMA USING MATCHING-ADJUSTED INDIRECT TREATMENT
COMPARISON
M. Delforge1,*, S. Z. Usmani2, N. W. van de Donk3, A. L. Garfall4, P. Moreau5, A.
Oriol6, A. K. Nooka7, L. Rosinol8, N. Bahlis9, P. Rodriguez-Otero10, T. Martin11,
J. Diels12, S. Van Sanden12, L. Pei13, E. Ammann14, R. Kobos13, M. Slavcev14, J. Smit15,
A. Londhe16, A. Krishnan17
1University of Leuven, Leuven, Belgium; 2Memorial Sloan Kettering Cancer Center, New
York, NY, United States of America; 3Amsterdam University Medical Center, Vrije Universiteit
Amsterdam, Amsterdam, Netherlands; 4Abramson Cancer Center, Perelman School of Medicine,
University of Pennsylvania, Philadelphia, PA, United States of America; 5University
Hospital Hôtel-Dieu, Nantes, France; 6Institut Català d’Oncologia and Institut Josep
Carreras, Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain; 7Winship Cancer
Institute, Emory University, Atlanta, GA, United States of America; 8Hospital Clínic,
IDIBAPS, University of Barcelona, Barcelona, Spain; 9Arnie Charbonneau Cancer Institute,
University of Calgary, Calgary, AB, Canada; 10Clínica Universidad de Navarra, CIMA,
CIBERONC, IDISNA, Pamplona, Spain; 11University of California, San Francisco, San
Francisco, CA, United States of America; 12Janssen Pharmaceutica NV, Beerse, Belgium;
13Janssen Research & Development; 14Janssen Global Services, Raritan, NJ; 15Janssen
Research & Development, Spring House, PA; 16Janssen Research & Development, Titusville,
NJ; 17City of Hope Comprehensive Cancer Center, Duarte, CA, United States of America
Background: Treatment options are limited for patients with relapsed/refractory multiple
myeloma (RRMM) who are triple-class exposed (TCE) to immunomodulatory drugs, proteasome
inhibitors, and anti-CD38 antibodies. While there is no standard of care for treatment
of patients with TCE RRMM, belantamab mafodotin (belamaf) is a recently approved,
novel therapeutic option. Teclistamab (tec; JNJ-64007957) is a B-cell maturation antigen
× CD3 bispecific antibody currently being evaluated in the single-arm, phase 1/2 MajesTEC-1
study (NCT04557098) in patients with TCE RRMM who received ≥3 prior lines of therapy.
Aims: Given the absence of a control arm in MajesTEC-1, the efficacy outcomes of patients
treated with tec at the recommended phase 2 dose in MajesTEC-1 were compared with
those treated with belamaf in the phase 2 DREAMM-2 study (NCT03525678).
Methods: An unanchored matching-adjusted indirect comparison was conducted using individual
patient-level data (IPD) from 150 patients treated with tec 1.5 mg/kg weekly in MajesTEC-1
(clinical cutoff of Sep 7, 2021), and published summary-level data from 97 patients
treated with the approved dose of belamaf (2.5 mg/kg every 3 weeks) in DREAMM-2. After
applying the DREAMM-2 eligibility criteria to patients from the intent-to-treat population
of MajesTEC-1, IPD from MajesTEC-1 were weighted to match the aggregated DREAMM-2
baseline patient characteristics. Baseline characteristics of prognostic significance
(such as cytogenetic profile, International Staging System stage, presence of extramedullary
disease, number of prior lines of therapy, and refractory status) were adjusted for
the analysis. Comparative efficacy of tec vs belamaf was estimated for overall response
rate (ORR), complete response or better (≥CR) rate, duration of response (DOR), overall
survival (OS), and progression-free survival (PFS). An odds ratio (OR) and 95% confidence
interval (CI) derived from a weighted logistic regression analysis was used to compare
the relative effects of tec vs belamaf for binary outcomes (ORR and ≥CR rate). A weighted
Cox proportional hazards model was used to estimate time-to-event endpoints (DOR,
OS, and PFS).
Results: After adjustment, the effective sample size of the MajesTEC-1 cohort was
33. Baseline characteristics were balanced between MajesTEC-1 and DREAMM-2 after reweighting
the MajesTEC-1 cohort. Tec-treated patients had improved outcomes compared with patients
treated with belamaf: ORR (OR 2.05; 95% CI 0.92–4.57; P=0.0786), ≥CR rate (OR 2.13;
95% CI 0.80–5.65; P=0.1283), DOR (hazard ratio [HR] 0.19; 95% CI 0.05–0.73; P=0.0149),
OS (HR 0.95; 95% CI 0.47–1.92; P=0.8897), and PFS (HR 0.63; 95% CI 0.34–1.15; P=0.1338).
For most outcomes, the lack of statistical significance may be due to the reduced
effective sample size following adjustment.
Summary/Conclusion: In this comparative analysis, tec showed statistically improved
DOR when compared with belamaf and numerically favorable results for other outcomes.
This highlights the potential of tec as a treatment option for patients with TCE RRMM
who received ≥3 prior lines of therapy.
P899: REAL-WORLD ASSESSMENT OF TREATMENT PATTERNS AND OUTCOMES IN PATIENTS WITH LENALIDOMIDE-REFRACTORY
RELAPSED/REFRACTORY MULTIPLE MYELOMA FROM THE OPTUM DATABASE
B. Dhakal1,*, H. Einsele2, R. Potluri3, J. Schecter4, W. Deraedt5, N. Lendvai4, A.
Slaughter6, C. Lonardi7, S. Nair5, J. He8, N. Joseph9, P. Cost8, S. Valluri8, F. Yalniz10,
L. Pacaud10, K. Yong11
1Medical College of Wisconsin, Milwaukee, United States of America; 2Universitätsklinikum
Würzburg, Medizinische Klinik und Poliklinik II, Wuerzburg, Germany; 3SmartAnalyst
Inc, New York; 4Janssen R&D, Raritan, United States of America; 5Janssen Pharmaceutica
NV, Beerse, Belgium; 6Cilag GmbH International, Zug, Switzerland; 7Janssen, Buenos
Aires, Argentina; 8Janssen Global Services, LLC, Raritan; 9Janssen Scientific Affairs,
LLC, Horsham; 10Legend Biotech USA, Piscataway, United States of America; 11University
College London Cancer Institute, London, United Kingdom
Background: New treatment combinations using proteasome inhibitors (PIs), immunomodulatory
drugs (IMiDs), and anti-CD38 monoclonal antibodies have significantly improved survival
outcomes in patients with multiple myeloma (MM). However, for patients who relapse
and/or become refractory after exposure to PIs and IMiDs, selecting the next regimen
remains a challenge. There are limited data characterizing treatments and outcomes
in this difficult-to-treat population.
Aims: To characterize treatment patterns and outcomes in difficult-to-treat patients
with MM, stratified by their number of prior lines of therapy (PL).
Methods: Data were derived from the Optum deidentified US claims and electronic health
record (EHR) database. Patients with MM (index diagnosis in or after Jan 2011) were
selected if they received 1-3 PL, including with a PI and an IMiD, and were refractory
to lenalidomide. Subsequent treatments beginning in or after Jan 2016 were included
to focus on contemporary therapies. Time zero (T0) was the date when the first subsequent
therapy started after patient met inclusion criteria. Patient characteristics were
assessed at T0. Time-to-event analyses were estimated using the Kaplan-Meier method
(starting at T0) for overall survival (OS) and time to next treatment (TTNT). The
log-rank test for trend was applied to compare Kaplan-Meier curves for OS by line
of therapy.
Results: The database included a total of 13,615 (claims) and 22,626 (EHR) patients
with an index diagnosis of MM and ≥1 line of therapy. 1,028 (claims) and 1,416 (EHR)
patients met inclusion criteria. Median age was 72 (claims) and 68 (EHR) years; 53.4%
(claims) and 52.1% (EHR) were male. 12.2% (claims) and 4.9% (EHR) of patients received
stem cell transplant prior to T0. Patients in both cohorts had significant comorbidities
with mean Charlson Comorbidity Index 3.9 (claims) and 3.2 (EHR). Hypertension and
renal failure were among the most prominent comorbidities. The most common subsequent
treatment regimens based on hierarchy were daratumumab (22.1% claims; 17.4% EHR),
carfilzomib (10.6% claims; 18.4% EHR), and pomalidomide based (12.3% claims; 8.7%
EHR). Daratumumab/pomalidomide/dexamethasone was the most frequently used regimen
(5.4% claims; 3.8% EHR). For claims data, median OS (months) was 36.7 (95% CI: 31.7-41.2).
Median OS was not reached (42.3-NE) for patients with 1 PL (n=546), was 26.2 (21.7-35.7)
for patients with 2 PL (n=380) and was 12.1 (7.6-25.0) for patients with 3 PL (n=102).
Median TTNT (months) was 5.3 (4.9-5.9) overall, including 4.6 (4.3-5.1) for patients
with 1 PL, 6.4 (5.3-7.6) for patients with 2 PL, and 6.0 (4.9-9.4) for patients with
3 PL. For EHR data, median OS was 34.0 (28.2-42.8), with median OS of 46.9 (42.8-NE)
for patients with 1 PL (n=587), 28.2 (23.8-41.7) for patients with 2 PL (n=584), and
20.8 (14.9-29.8) for patients with 3 PL (n=245). Median TTNT was 5.8 (5.1-6.5) overall,
with values of 5.1 (4.7-6.5) for patients with 1 PL, 6.2 (5.3-7.6) for patients with
2 PL, and 5.8 (4.4-8.2) for patients with 3 PL.
Image:
Summary/Conclusion: PI-exposed, lenalidomide-refractory patients with 1-3 PL were
treated with various regimens. These patients have poor OS and progress quickly through
current therapies, suggesting poor progression-free survival. OS is reduced with each
successive line of therapy. This analysis highlights the need for new effective regimens
for this patient population.
P900: SALVAGE AUTOLOGOUS STEM CELL TRANSPLANT IN RELAPSED MULTIPLE MYELOMA: A SINGLE
CENTRE EXPERIENCE
S. Duarte1,*, A. Roque1,2, D. Mota1, J. Carda1,2, C. Geraldes1,2
1Clinical Hematology Department, University Hospital Center of Coimbra; 2Faculty of
Medicine, University of Coimbra, Coimbra, Portugal
Background: Multiple myeloma (MM) is an incurable disease and most patients (pts)
will relapse during their lifetime. Although salvage therapy has improved with the
advent of new drugs, salvage autologous stem cell (ASC) transplant (SAT) remains appropriate
for pts relapsing after primary therapy that includes ASCT with initial remission
duration >18 months (mo).
Aims: Evaluate the outcome of SAT in MM in the new drugs era and efficacy and toxicity
associated to this treatment.
Methods: Single centre retrospective analysis of MM pts submitted to SAT between 2007-2021.
Tandem ASCT excluded. Engraftment defined according to EBMT criteria.
Results: Fifty-three pts submitted to SAT, 58.5% male, median age of 60 yo (37-71),
27.8% with international staging system score III. Median number of prior lines of
therapy before SAT was 2 (2-4), 51 (96.2%) including proteasome inhibitors (PI), 38
(71.7%) immunomodulators (IMiD) and 13 (24.5%) daratumumab. Twenty-five (50%) pts
submitted to maintenance therapy with thalidomide (Thal) after ASCT and 15 (30.6%)
with lenalidomide (Len) after SAT.
Pre-SAT response to therapy was complete remission (CR) in 9.6%, very-good partial
response (VGPR) in 50% and PR in 40.4%. Most pts received conditioning with melphalan
200mg/m2 for both transplants (1st: 100%, 2nd: 96.2%).
After SAT, median time to neutrophil engraftment was 12 days (6-21) and for platelet
recovery was 13 days (3-26). Grade 3 oral and gastrointestinal mucositis observed
in 14% and 6%, respectively, and febrile neutropenia in 91% of pts. SAT response evaluation
showed CR in 28.3%, VGPR in 32.1% and PR in 37.7%. One patient showed progressive
disease. No treatment related mortality (TRM) at 100 days post-SAT.
Median PFS after 1st ASCT was 32 mo. Median time between 1st and 2nd ASCT was 46 mo
(22-140). Median PFS and OS after SAT was 28 and 78 mo, respectively. Comparison between
PFS post-ASCT and post-SAT was not significant (P=0.09).
ISS stage with no impact on PFS (HR 1.99; p=0.263) and OS (HR 2.03; p=0.398), after
SAT. No statistically significant difference in post-SAT PFS (HR 1.23 p=0.592) and
OS (HR 1.84, p=0.312) between patients submitted to transplant before and after 2016,
despite more intensive therapeutic protocols (100% triplets and 24.5% anti-CD38-based
regimens in former period compared with duplets in the first). PFS of pts who reached
CR post-SAT was significantly higher compared to pts in VGPR/PR (NR vs 22 mo, HR 2.97,
p=0.044). However, CR post-SAT with no impact on OS (NR vs 118 mo, HR 1.27 p=0.71).
Maintenance therapy with (Thal) post-ASCT with no impact on PFS (33 vs 32 mo, HR 1.24,
p=0.47). Similarly, the impact of post-SAT maintenance with Len on PFS (51 vs 28 mo,
HR 1.35, p=0.49) and OS (NR vs 73 mo, HR 4.40, p=0.157) was not significant.
Pts with remission time duration after 1st ASCT ≥36 mo had median PFS and OS after
SAT significantly longer (51 vs 22 mo, HR 2.41, p=0.024, and NR vs 71 mo, HR 6.24,
p=0.015, respectively), compared to pts with post-ASCT remissions <36 mo.
Univariate analysis failed to show other clinical and analytical variables with impact
on PFS and OS.
Summary/Conclusion: In our cohort, median PFS and OS after SAT was 78 mo and 28 mo,
respectively. Pts with remissions after 1st ASCT ≥36 mo showed significantly longer
PFS and OS. PFS was significantly improved in pts reaching CR after SAT. Post-SAT
Len maintenance improved PFS and OS, however with no statistical significance. Severe
toxicities were rare and never resulted in TRM. Overall, our centre experience shows
that SAT is an effective and safe treatment in MM in the new drugs era.
P901: POMALIDOMIDE, BORTEZOMIB, AND DEXAMETHASONE IN LENALIDOMIDE–PRETREATED MULTIPLE
MYELOMA: A SUBANALYSIS OF OPTIMISMM BY FRAILTY
A. Oriol1,*, M. Dimopoulos2, F. Schjesvold3,4, M. Beksac5, M. Yagci6, A. Larocca7,
S. Guo8, Y. Mu8, K. Hong9, S. Dhanasiri10, P. Richardson11, K. Weisel12
1Hematology Department, Institut Català d’Oncologia, Barcelona, Spain; 2National and
Kapodistrian University, Athens, Greece; 3Oslo Myeloma Center, Department of Hematology,
Oslo University Hospital; 4KG Jebsen Center for B Cell Malignancies, University of
Oslo, Oslo, Norway; 5Ankara Üniversitesi Tip Fakültes; 6Gazi University Medical Faculty,
Ankara, Turkey; 7SSD Clinical trials in onco-ematologia e mieloma multiplo, Division
of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della
Salute e della Scienza di Torino, Torino, Italy; 8Evidera, Waltham; 9Bristol Myers
Squibb, Princeton, United States of America; 10Celgene International Sàrl, a Bristol-Myers
Squibb Company, Boudry, Switzerland; 11Dana-Farber Cancer Institute, Boston, United
States of America; 12University Medical Center Hamburg-Eppendorf, Hamburg, Germany
Background: Patients (pts) with multiple myeloma (MM) are likely to be older adults,
and advanced age is associated with lower survival rates, in part due to comorbidities
and frailty. Results from the phase 3 OPTIMISMM trial (NCT01734928) demonstrated that
pomalidomide (P) in combination with bortezomib + dexamethasone (Vd) significantly
improved progression-free survival (PFS) in lenalidomide (LEN)–pretreated pts with
relapsed/refractory MM (RRMM) vs Vd, irrespective of age and Eastern Cooperative Oncology
Group performance status (ECOG PS).
Aims: Here, we report results from a post hoc analysis assessing outcomes of PVd vs
Vd in pts with RRMM by frailty status.
Methods: Pts with MM and 1–3 prior lines of therapy (including a LEN-containing regimen)
who had been randomized 1:1 to PVd or Vd were assessed for frailty. Frailty scores
were calculated using age (≤ 75 yr = 0; 76–80 yr = 1; > 80 yr = 2), the Charlson Comorbidity
Index (≤ 1 = 0; > 1 = 1), and ECOG PS (0 = 0; 1 = 1; ≥ 2 = 2). Pts were classified
as non-frail (NF; combined score: 0 or 1) or frail (F; combined score: ≥ 2). PFS,
overall response rate (ORR), and safety outcomes were assessed by treatment group
and frailty status.
Results: In the intent-to-treat population (N = 559) (data cut-off Oct 26, 2017),
93/281 (33.1%) pts who received PVd and 93/278 (33.5%) who received Vd were frail.
Baseline characteristics were similar between treatment groups within each frailty
subgroup. Median PFS was longer with PVd vs Vd in NF (P = 0.001) and F (P = 0.006)
subgroups (Table). ORR was higher with PVd vs Vd in NF (P < 0.001) and F (P < 0.001)
subgroups. Incidence of grade ≥ 3 (G3) treatment-emergent adverse events (TEAEs) was
higher with PVd vs Vd in NF (88.1 vs 61.3%) and F (96.8 vs 87.9%) subgroups; peripheral
neuropathy, acute renal failure, and hypertension were the most common. Treatment
discontinuation due to G3 TEAEs was greater with PVd vs Vd in NF (19.2 vs 18.5%) and
F (30.1 vs 20.9%) subgroups. PVd had a longer median treatment duration vs Vd in NF
(8.8 vs 5.7 mo) and F (8.9 vs 4.3 mo) subgroups.
Image:
Summary/Conclusion: Frail pts with RRMM who received PVd had longer PFS and higher
ORR than pts who received Vd, consistent with the overall OPTIMISMM results. Frail
pts experienced more G3 TEAEs and treatment discontinuations with PVd vs Vd, but treatment
duration was longer with PVd vs Vd.
P902: RELAPSED/REFRACTORY MULTIPLE MYELOMA PATIENTS. A MULTICENTER RETROSPECTIVE ANALYSIS
OF ELIGIBILITY CRITERIA FOR CAR-T CELL THERAPY
F. Fazio1,*, A. Di Rocco1, M. T. Giaimo2, T. Za3, N. Ciccone4, M. Sessa4, B. Gamberi5,
V. Tomarchio6, L. De Padua7, V. Bongarzoni8, S. Mariani9, A. Rago10, A. Piciocchi11,
F. Merli5, A. Cuneo4, V. De Stefano3, R. Foà1, M. Martelli1, M. T. Petrucci1
1Hematology - Azienda Policlinico Umberto I, Department of Traslational and Precision
Medicine Sapienza University of Rome, Roma; 2Hematology - Azienda Ospedaliera Parma,
University of Parma, Parma; 3Hematology - Fondazione Policlinico A. Gemelli IRCCS,
Department of Radiological and Hematological Sciences, Catholic University, Roma;
4Hematology - Ospedale Sant’Anna, University of Ferrara, Ferrara; 5Hematology, AUSLL/IRCCS
Santa Maria Nuova Hospital, Reggio Emilia; 6Hematology - Stem Cell Transplantation,
University Campus Bio Medico, Roma; 7Hematology - Ospedale Fabrizio Spaziani, Frosinone;
8Hematology - Azienda Ospedaliera San Giovanni; 9Hematology - Ospedale Sant’Andrea,
Sapienza University of Rome; 10UOSD Hematology ASLRoma 1; 11Italian Group For Adult
Hematologic Diseases (GIMEMA), Roma, Italy
Background: The overall survival (OS) of multiple myeloma (MM) patients (pts) has
improved over the years due to the introduction of novel drugs, such as proteosome
inhibitors (PI), immunomodulatory drugs (IMiDs) and anti- CD38 monoclonal antibodies
(moAb). Nevertheless, the majority of pts continues to relapse and MM remains an incurable
disease. No standard of care has been established for relapsed/refractory (RR) MM
pts who have been exposed to the main anti-myeloma drugs. The outcome of pts failing
standard of care regimens, which is now defined as triple-refractory, is poor, with
a median progression-free survival (PFS) of 3-4 months and OS of 8-9 months. Novel
therapeutic strategies are warranted to overcome the natural occurrence of relapse
or therapy resistance in RRMM pts. Chimeric antigen receptor (CAR)-modified T cells
are a promising new therapy approach for triple refractory RRMM. Specific CAR-T targets
are being studied, but BCMA-directed CAR-T cells have so far provided the most convincing
evidence of activity, with one product (idecabtagene vicleucel) recently approved
by FDA and EMA.
Aims: The primary endpoint of this observational and retrospective study was to define
the clinical characteristics and outcome of a cohort of RRMM pts potentially eligible
to CAR-T cell treatment according to the KarMMa trial criteria. Secondary endpoints
were aimed at defining specific factors influencing CAR-T cell therapy eligibility
and at identifying a real-life estimate of RRMM pts truly eligible for CAR-T cells.
Methods: This is a cohort analysis on RRMM pts managed between January 2018 and July
2021 at 10 Italian hematology centers. At the time of data collection, 108 RRMM pts
had underwent at least 3 prior therapy regimens; they had received a previous PI,
IMiDs and a moAb, and were considered refractory to the last regimen.
Results: Median age was 68 years (38-86); 63 (59%) pts were >65 years; 57 (53%) were
male. Of 108 pts, 87 (80%) were ECOG 0-1 and 33 (35%) were ISS III. The majority of
pts, 72 (67%), had undergone an autologous stem cell transplantation; 93 pts received
>3 prior lines of therapy. Sixty-seven (62%) were triple-refractory and 41 (38%) were
penta-refractory. Based on the KarMMa trial criteria, 49/108 pts (45%) would be defined
as eligible and 59 (55%) not eligible for CAR-T cell therapy. Organ dysfunction such
as impaired renal function, anemia, thrombocytopenia, neutropenia and a FEV <45% was
the most common criteria for ineligibility (86%). Twenty-one pts (19%) were not eligible
because of an ECOG >2. Of the 59 pts considered ineligible for CAR-T cell therapy,
46 (62%) presented ≥2 ineligibility criteria.
After a median follow-up of 27 months (mo) (8-40.5), the median OS for the entire
cohort was 21.1 mo (Fig. 1A). The median OS was 33 mo in eligible pts vs 8.3 mo in
non-eligible pts (p=0.002) (Fig. 1B). The median PFS of the entire cohort was 8.7
mo (Fig. 1C) and the median PFS was 19.4 mo in eligible pts vs 6 mo (p=0.001) in non-eligible
pts (Fig. 1D).
Image:
Summary/Conclusion: Despite the limits of a retrospective study and a limited cohort,
our real-life analysis shows that heavily treated pts with RRMM are less likely to
be eligible for CAR-T cell therapy. Considering the emergent role of quadruplet combined
approaches for first- line therapy and given the therapeutic relevance of CAR-T cells
for the management of RRMM pts previously exposed to PI, IMiDs and moAb, our data
could help to better define pts who could benefit from CAR-T cells under the current
indications, while waiting for an extension of this approach to earlier disease stages.
P903: DARATUMUMAB PLUS BORTEZOMIB AND DEXAMETHASONE IN NEWLY DIAGNOSED PATIENTS WITH
MAYO 2004 STAGE 3 LIGHT-CHAIN AMYLOIDOSIS: A PROSPECTIVE PHASE 2 STUDY
Y.-J. Gao1,*, K.-N. Shen1, L. Chang1, J. Feng1, L. Zhang1, Y.-Y. Mao1, X.-X. Cao1,
D.-B. Zhou1, J. Li1
1Department of Hematology, Peking Union Medical College Hospital, Beijing, China
Background: Patients with systemic light-chain (AL) amyloidosis at the advanced cardiac
stage exhibit extremely poor survival. Although daratumumab has shown superior outcome
in treatment of AL amyloidosis according to the result of ANDROMEDA trial, whether
these feeble patients can benefit from daratumumab therapy need further investigation.
Aims: To prospectively explore the value of daratumumab plus bortezomib and dexamethasone
in patients with AL amyloidosis at the advanced heart-stage.
Methods: This is a phase 2, open-label, single center clinical trial planning to include
40 newly diagnosed patients with Mayo 2004 stage 3a and 3b AL amyloidosis at Peking
Union Medical College Hospital (Beijing, China). Eligible patients should have measurable
hematological disease (baseline dFLC >50mg/L). Initiation treatment includes daratumumab
(intravenously at 16 mg/kg weekly during cycles 1-2, once every two weeks during cycles
3-6 and once every 4 weeks thereafter for up to 12 cycles), bortezomib (at 1.3mg/m2
subcutaneously weekly during cycles 1-6) and dexamethasone (20mg weekly during cycles
1-6). Each cycle consists of 4 weeks. Treatment responses are evaluated every week
during the first cycle and at the end of each cycle since cycle 2. Termination of
the therapy is considered when patients have progressed disease, serious side effects
related to treatment or initiation of the second-line treatment. (ClinicalTrials.gov
identifier: NCT04474938)
Results: From 28th May, 2021 to 28th January, 2022, 38 patients were enrolled. Twenty-nine
(76.3%) patients were male and the median age was 59 years (range 41-77). The median
NT-proBNP was 10665 pg/ml (range 803 ~ >35000) and the median cTnI was 0.17ug/L (range
0.07-3.07). Twenty-one patients (55.3%) were stage 3b. Median dFLC was 265 mg/L (range
72-2966). Twenty-four patients (63.2%) had NYHA class III or IV heart function. The
median number of organs involved was 2 (range 0-4). Fourteen (36.8%) patients had
kidney involvement and nine (23.7%) patients had liver involvement.
The median number of treatment cycles was 3.25 (range 0.25-9). For the best hematologic
response, 34 of 38 patients (89.5%) reached ≥PR, including 18 patients (47.4%) with
CR and 8 (21.1%) patients with VGPR. At the end of the first cycle, 8 patients (22.9%)
achieved CR; 12 patients (34.3%) reached VGPR; 9 patients (25.7%) had PR and 6 patients
(17.1%) were NR. The hematological ORR was 74.1% at 3 months (40.7% with CR and 18.5%
with VGPR). The median time to the first hematologic remission was 7 days (range 7-21)
and the median time to ≥VGPR was 14 days (range 7 days – 3 months). The cardiac response
rate was 18.5% at 3 months and 23.5% at 6 months. After the median follow-up time
of 4.5 months (range 0.7-8.6), the median OS was not reached. The 6-month survival
rate of all patients was 76.6%, with 84.7% for stage 3a and 69.0% for stage 3b.
The most common grade 3 or 4 adverse events were infection (n=8, 21.1%). Infusion
reaction was recorded in 5 patients (13.2%) and only 1 patient was categorized as
grade 3 reaction. All of them occurred during the first time of infusion. Other serious
adverse effects included grade 3 diarrhea (n=3), pneumothorax (n=1), bone fracture
(n=1), deep venous thrombosis (n=1), hematuresis (n=1), gastrointestinal bleeding
(n=1), ischemic stroke (n=1) and intestinal obstruction (n=1). No patients withdrew
due to adverse events.
Image:
Summary/Conclusion: Our results showed that daratumumab plus bortezomib and dexamethasone
had favorable safety and potential advantage among patients with AL amyloidosis presenting
severe cardiac involvement.
P904: CILTACABTAGENE AUTOLEUCEL VS TREATMENTS FROM REAL-WORLD CLINICAL PRACTICE FOR
TRIPLE CLASS EXPOSED PATIENTS WITH MULTIPLE MYELOMA: ADJUSTED COMPARISONS BASED ON
CARTITUDE-1 AND THE EMMY FRENCH COHORT
O. Decaux1,*, C. Hulin2, A. Perrot3, M. Macro4, L. Frenzel5, J. Diels6, N. J. Perualila6,
F. Ghilotti7, B. Haefliger8, E. Goldsztajn9, S. Vernet10, J. Thevenon10, M. Willaime11,
N. Texier11, J. M. Schecter12, D. Madduri12, C. Jackson12, S. Valluri13, P. Moreau14
1Internal medicine and clinical immunology, CHU Rennes, Rennes; 2Hematology and cell
therapy, CHU Bordeaux, Bordeaux; 3Hematology, Oncopole, Toulouse; 4Hematology, CHU
Caen, Caen; 5Hematology, Necker-enfants malades hospital, Paris, France; 6Health Economics,
Market Access & Reimbursement, Janssen Pharmaceutica NV, Beerse, Belgium; 7Health
Economics, Market Access & Reimbursement, Janssen Pharmaceutica NV, Cologno Monzese,
Italy; 8Health Economics, Market Access & Reimbursement, Cilag GmbH International,
Zug, Switzerland; 9Market Access & Reimbursement; 10Medical affairs, Janssen, Issy-les-Moulineaux;
11Biometry, Kappa Sante, Paris, France; 12Research and Development, Janssen R&D; 13Health
Economics, Market Access & Reimbursement, Janssen, Raritan, United States of America;
14Clinical hematology, University Hospital Hotel-Dieu, Nantes, France
Background: The prognosis of patients with triple-class (TC) exposed, relapsed and
refractory multiple myeloma (RRMM) is poor. CARTITUDE-1 is an open-label, single arm,
phase 1b/2 clinical trial performed to assess the safety and efficacy of ciltacabtagene
autoleucel (cilta-cel), a novel chimeric antigen receptor T-cell therapy. Adjusted
comparisons to other therapies used in real-world clinical practice (RWCP) help inform
the relative efficacy of cilta-cel in TC exposed patients. The Epidemiology of the
therapeutic management of Multiple MYeloma in France (EMMY) study is a retrospective
study capturing information on the therapeutic management of patients from 71 centers
in France, and can serve as an external control arm for CARTITUDE-1.
Aims: To compare cilta-cel with treatments from RWCP in terms of overall response
rate (ORR), very good partial response or better rate (≥VGPR), progression-free survival
(PFS), and overall survival (OS).
Methods: Adjusted comparisons were estimated using individual patient data (IPD) from
CARTITUDE-1 (cut-off date July 2021) and reconstructed IPD from EMMY (cut-off date
March 2021). Within EMMY, all treatment lines initiated after a patient met eligibility
criteria (TC exposed patients in L4+ and ECOG<2) were used for analysis, so long as
eligibility remained intact. Inverse probability weighting (IPW) was used to balance
the cilta-cel and RWCP groups on prognostic baseline variables including refractory
status, time to progression on prior line of treatment (LOT), number of prior LOTs,
ECOG performance status, age, sex, years since MM diagnosis, average duration of prior
LOTs, MM type, and prior transplant. Weights derived from propensity scores estimated
with a multivariable logistic regression and were assigned to EMMY patients, such
that the weighted EMMY cohort reflected the CARTITUDE-1 population. Balance was assessed
using standardized mean differences (SMD). The relative efficacy of cilta-cel versus
RWCP was assessed using relative response rates (RR) for binary outcomes and hazard
ratios (HR) for time-to-event outcomes, using weighted logistic regression and proportional
hazards modeling, respectively. Main analyses were performed in the population of
infused patients in the cilta-cel group and an aligned population of EMMY RWCP patients
that included those still progression-free after 52 days (mean time between apheresis
and cilta-cel infusion). Similar analyses were performed for all enrolled patients.
Results: 113 patients were enrolled in CARTITUDE-1 and 97 received cilta-cel infusions.
437 LOTs (238 patients) from EMMY were included in the RWCP external control arm and
309 LOTs (227 patients) were alive and progression-free after 52 days. More than 100
treatment regimens within the RWCP group were used. After reweighting, baseline traits
were balanced between groups, with all SMD <0.20. ORR (RR 5.1, 95% CI: 3.7;7.1) and
≥VGPR (RR 14.5 [8.0; 26.1] were superior with cilta-cel vs. RWCP, as were both PFS
(HR 0.15 [0.10; 0.22]) and OS (HR 0.21 [0.12; 0.37]). Results for the enrolled populations
were consistent (see Table).
Image:
Summary/Conclusion: Adjusted comparisons between cilta-cel and RWCP in France showed
superior efficacy on all outcomes for cilta-cel vs. RWCP, with 5.1 and 14.5-fold higher
response rates for ORR and ≥VGPR, respectively, and reduced risks of disease progression/death
and death by 85% and 79% with cilta-cel versus RWCP. These findings align with results
from similar comparisons in other countries. Cilta-cel offers substantial clinical
benefits for patients with triple class exposed RRMM.
P905: IXAZOMIB-THALIDOMIDE-DEXAMETHASONE INDUCTION FOLLOWED BY IXAZOMIB OR PLACEBO
MAINTENANCE IN NON-TRANSPLANT ELIGIBLE NEWLY DIAGNOSED MULTIPLE MYELOMA PATIENTS;
LONG-TERM RESULTS OF HOVON-126/NMSG 21.13
K. Groen1,*, M. R. Seefat1, B. van der Holt2, F. H. Schjesvold3,4, C. A. Stege1, M.-D.
Levin5, M. Hansson6,7, R. B. Leys8, J. Regelink9, A. Waage10, D. Szatkowski11, P.
Axelsson12, T. H. Do13, A. Svirskaite14, E. van der Spek15, E. Haukas16, D. Knut-Bojanowska17,
P. F. Ypma18, C. Blimark19, U.-H. Mellqvist20, N. W. van de Donk1, P. Sonneveld21,
A. Klostergaard22, A. J. Vangsted23, N. Abdilgaard24,25, S. Zweegman1
1Hematology, Amsterdam UMC, Amsterdam; 2HOVON Data Center, Department of Hematology,
Erasmus MC Cancer Institute, Rotterdam, Netherlands; 3Hematology, Oslo Myeloma Center;
4KG Jebsen center for B Cell malignancies, University of Oslo, Oslo, Norway; 5Department
of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, Netherlands; 6Skane University
Hospital, Lund; 7Sahlgrenska Academy, Göteborg, Sweden; 8Department of Internal Medicine,
Maasstad Ziekenhuis, Rotterdam; 9Department of Internal Medicine, Meander Medisch
Centrum, Amersfoort, Netherlands; 10IKOM, Norwegian University of Science and Technology,
Trondheim; 11Førde Central Hospital, Førde, Norway; 12Skanes University Hospital,
Lund, Sweden; 13Herlev Hospital, Herlev; 14Aalborg Hospital, Aalborg, Denmark; 15Department
of Internal Medicine, Rijnstate, Arnhem, Netherlands; 16Stavanger University Hospital-Rogaland
Hospital, Stavanger, Norway; 17NU-Hospital - Uddevalla Hospital, Uddevalla, Sweden;
18Department of Hematology, Haga Ziekenhuis, Den Haag, Netherlands; 19Sahlgrenska
University Hospital, Gothenburg; 20Sodra Alvsborgs Sjukhus Boras, Boras, Sweden; 21Erasmus
Medical Center Cancer Institute, Rotterdam, Netherlands; 22Aarhus University Hospital,
Aarhus; 23Department of Hematology, Rigshospitalet, Copenhagen; 24Department of Hematology;
25Academy or Geriatric Cancer Research, Odense University Hospital, Odense, Denmark
Background: In the HOVON 126/NMSG 21.13 trial non-transplant eligible newly diagnosed
multiple myeloma (NTE-NDMM) patients were treated with 9 induction cycles of ixazomib,
thalidomide and dexamethasone (ITd), followed by randomization between either ixazomib
or placebo until progression or unacceptable toxicity. The overall response rate and
PFS data have been previously published.
Aims: We here present the long-term PFS2 and overall survival data.
Methods: Patients were treated with 9 induction cycles (28 days) of ixazomib (4mg
on day 1, 8 and 15), thalidomide (100mg on day 1-28) and dexamethasone (40mg on day
1, 8, 15 and 22), followed by maintenance with either ixazomib or placebo (4mg, both
on day 1, 8 and 15, every 28 days). Patients were classified as fit, intermediate
fit or frail, based on a modified IMWG frailty index which incorporated age, the Charlson
Comorbidity Index (CCI) and the WHO performance as a proxy for (instrumental) Activities
of Daily Living (iADL) (scoring WHO 0 as 0 points, WHO 1 as 1 point, and WHO 2-3 as
2 points).
Results:
From registration: 143 eligible patients were included in the study. After a median
follow-up (FU) of 67.4 months (m), the median PFS was 14.3m (95% CI 11.5-16.8), median
PFS2 was 34.6m (30.7-41.5) and median OS was 58.3m (50.5-65.0). There was no difference
in PFS between frailty subgroups. In contrast, median PFS2 and OS were longer in fit
patients (PFS2: 49.1m (34.6-74.1), OS: NR (66.6-NR)) versus intermediate-fit (30.1m
(25.1-39.0); 51.2m (32.3-63.9) resp.) and frail patients (30.9m (24.0-42.3); 50.5m
(32.9-59.4) resp.).
From randomization: 78 (55%) patients were randomized, 39 patients in each arm. After
a median FU of 60 months from randomization, there was no difference in PFS between
the ixazomib-arm (median 9.5m; 95% CI 5.5-14.8) and the placebo-arm (8.4m; 3.0-13.8).
Median PFS2 was 39.8m (28.8-60.0) for patients on ixazomib, as compared to 28.7m (22.8-43.2)
for patients in the placebo arm, although this difference was not statistically significant.
Median OS was not reached for the ixazomib arm and was 50.7m (41.3-58.1) for the placebo
arm (HR 0.39; 95% CI 0.19-0.78, p=0.008).
In both arms 32 (82%) patients received 2nd line treatment. With the caveat of low
numbers and heterogeneous treatment regimens, more patients in the ixazomib arm received
daratumumab-lenalidomide-dexamethasone (4 patients, 13%) and panobinostat-bortezomib-dexamethasone
(6 patients, 19%), compared to the placebo arm (2 patients (6%) and 3 patients (9%)
respectively).
In order to explain the difference in OS, subsequent lines of therapy are currently
being investigated.
Image:
Summary/Conclusion: With longer FU, we here confirm that ixazomib maintenance therapy
did not improve PFS, compared to placebo. However, PFS2 tends to be longer and OS
was superior in patients treated with ITd followed by maintenance with ixazomib versus
placebo.
P906: IXAZOMIB, DARATUMUMAB AND LOW DOSE DEXAMETHASONE IN FRAIL PATIENTS WITH NEWLY
DIAGNOSED MULTIPLE MYELOMA (NDMM): RESULTS OF THE MAINTENANCE TREATMENT OF THE PHASE
II HOVON 143 STUDY
K. Groen1,*, M. Seefat1, K. Nasserinejad2, C. A. Stege1, E. van der Spek3, Y. M. Bilgin4,
A. Kentos5, M. Sohne6, R. J. van Kampen7, I. Ludwig8, N. Thielen9, N. Durdu-Rayman10,
N. C. de Graauw11, N. W. van de Donk1, E. G. de Waal12, M.-C. Vekemans13, G. J. Timmers14,
M. van der Klift15, S. Soechit16, P. A. Geerts17,18, M. H. Silbermann19, M. Oosterveld20,
I. Nijhof1,21, P. Sonneveld22, S. K. Klein23, M.-D. Levin24, S. Zweegman1
1Hematology, Amsterdam UMC, Amsterdam; 2HOVON Data Center, Department of Hematology,
Erasmus MC Cancer Institute, Rotterdam; 3Department of Internal Medicine, Rijnstate
Hospital, Arnhem; 4Department of Internal Medicine, Admiraal de Ruijter Hospital,
Goes, Netherlands; 5Department of Hematology, Centre Hospitalier Jolimont, Haine-Saint-Paul,
Belgium; 6Department of Internal Medicine/Hematology, St Antonius Hospital, Nieuwegein;
7Department of Internal Medicine, Zuyderland Medical Center, Sittard-Geleen; 8Department
of Hematology, Bernhoven Hospital, Uden; 9Department of Internal Medicine, Diakonessenhuis,
Utrecht; 10Department of Internal Medicine, Franciscus Hospital, location Vlietland,
Schiedam; 11Department of Internal Medicine, Bravis ziekenhuis, Roosendaal; 12Department
of Internal Medicine, Medisch Centrum Leeuwarden, Leeuwarden, Netherlands; 13Department
of Hematology, St Luc Hospital, Bruxelles, Belgium; 14Department of Internal Medicine,
Amstelland Hospital, Amstelveen; 15Department of Internal Medicine, Amphia Hospital,
Breda; 16Department of Hematology, Reinier de Graaf Groep, Delft; 17Department of
Internal Medicine, Deventer Hospital, Deventer; 18currently Isala, Zwolle; 19Department
of Internal Medicine, Tergooi Hospital, Hilversum; 20Department of Internal Medicine,
Canisius-Wilhelmina Hospital, Nijmegen; 21currently St Antonius Hospital, Nieuwegein;
22Department of Hematology, Erasmus MC Cancer Institute, Rotterdam; 23Department of
Internal Medicine, Meander Medical Center, Amersfoort; 24Department of Internal Medicine,
Albert Schweitzer Hospital, Dordrecht, Netherlands
Background: Frail patients with newly diagnosed multiple myeloma (NDMM) have an inferior
PFS and OS, a higher treatment discontinuation rate and more grade ≥3 non-hematologic
toxicity, compared to fit patients. In order to improve their outcome we investigated
a three-drug regimen with a presumed low-toxicity profile; ixazomib, daratumumab and
low-dose-dexamethasone (Ixa-Dara-dex).
This trial is registered at www.trialregister.nl as NTR6297.
Aims: To present the long term follow up outcome, with an emphasis on the maintenance
phase.
Methods: NDDM patients, who were frail according to the IMWG-Frailty Index, were included.
Patients defined frail based on age only (frail-age) were compared to frail patients
based on other reasons (impairments (i)ADL and/or CCI≥2; frail-other) and patients
frail based on age as well as other reasons (frail-both).
After nine cycles of Ixazomib (4mg; days 1, 8, 15), daratumumab (16mg/kg iv; cycles
1-2: days 1, 8, 15, 22; cycles 3-6: days 1, 15; cycles 7-9: day 1) and low dose dexamethasone
(on the days daratumumab was administered; cycle 1-2: 20mg; subsequent cycles 10mg),
patients without progressive disease or excessive toxicity continued with maintenance
treatment, consisting of 8-week cycles with ixazomib (4mg orally on days 1, 8, 15,
29, 36, 43), daratumumab (16mg/kg iv or 1800mg subcutaneously on day 1) with dexamethasone
(10mg intravenously on day 1), until progression, for a maximum of two years.
Results: Sixty-five frail patients were included. After a median follow-up of 39 months,
the median progression free survival was 13.8m (95%CI: 9.2-17.7). The median PFS2
for all patients was 30.7m (22.2-39.1), 39.1 months in patients classified as frail-age,
24.5 months in frail-other patients and 26.6 months in frail-both patient (log-rank
p=0.30). Median OS for all patients was 34.0m (24.0-41.2), not reached (frail age),
28.1m (frail-other) and 30.7 months (frail-both) (log-rank p=0.29) (Table 1).
Thirty-two patients (49%) proceeded to the maintenance phase. During maintenance treatment,
6 patients (19%) had improvement of response: 1 SD to MR, 3 PR to VGPR, 1 VGPR to
CR and 1 VGPR to sCR. The rate of VGPR or better improved from 41% to 50% during maintenance.
During maintenance, 21/32 (66%) patients discontinued therapy, because of progressive
disease (14/21; 67%), toxicity (2/21; 10%; infection and intracranial hemorrhage),
non-compliance (1/21; 5%), intercurrent death (1/21; 5%) and other reasons (3/21;
14%; physician’s choice, dementia and patient condition).
Hematologic ≥3 grade adverse events (AEs) during maintenance were limited: neutropenia
0%, anemia 3% and thrombocytopenia 9%. Non-hematologic ≥3 grade AEs occurred in 18
(56%) patients. Most common AEs were infections (9%), nervous system disorders (9%;
cognitive disturbance, stroke and syncope) and gastro-intestinal complaints (6%).
There were 2 (6%) second primary malignancies and one patient (3%) experienced grade
3 neuropathy.
Image:
Summary/Conclusion: Ixa-dara-dex maintenance treatment in frail patients was safe,
and resulted in an improvement in response rate in 19% of patients. Patients frail
based on age, had higher PFS, PFS2 and OS, as compared to other frail subgroups.
P907: CARFILZOMIB, DEXAMETHASONE, AND DARATUMUMAB (KDD) VS CARFILZOMIB AND DEXAMETHASONE
(KD) IN RELAPSED/REFRACTORY MULTIPLE MYELOMA (RRMM): FRAILTY SUBGROUP ANALYSIS OF
THE CANDOR STUDY
H. Quach1,*, X. Leleu2, M.-V. Mateos3, S. Z. Usmani4, A. K. Nooka5, A. Goldrick6,
R. Najdi6, N. Shu7, T. Facon8
1University of Melbourne, St Vincent’s Hospital, Melbourne, VIC, Australia; 2CHU de
Poitiers, La Miletrie/INSERM CIC 1402, Poitiers, France; 3University Hospital Salamanca/ISAL,
Salamanca, Spain; 4Memorial Sloan Kettering Cancer Center, New York; 5Winship Cancer
Institute, Emory University, Atlanta; 6Amgen, Thousand Oaks, United States of America;
7Parexel, Chengdu, China; 8Hôpital Claude Huriez, Lille, France
Background: Frailty scores based on age, comorbidities, and functional status can
help identify frail patients (pts) at risk of higher rates of treatment-related toxicity
or poor outcomes. An analysis of carfilzomib-treated pts in ASPIRE, ENDEAVOR, and
ARROW found that efficacy and safety of carfilzomib regimens is consistent regardless
of frailty status (Facon Blood Adv 2020).
Aims: To compare efficacy and safety across frailty subgroups in pts with RRMM treated
with KdD vs Kd in the phase 3 CANDOR study.
Methods: This post hoc analysis used a planned interim readout (June 15, 2020) of
CANDOR (NCT03158688). Pts were categorized as fit, intermediate (int), or frail based
on a frailty score of 0, 1, or ≥2. Scoring used an algorithm based on the International
Myeloma Working Group (IMWG) frailty index: the final score is based on age (0 if
≤75 years, 1 if 76-80 years, 2 if >80 years), modified Charlson Comorbidity Index
(CCI [Facon Blood Adv 2020]) (0 if CCI ≤1, 1 if CCI >1), and Eastern Cooperative Oncology
Group performance status (ECOG PS) (0 if ECOG PS=0, 1 if ECOG PS=1, and 2 if ECOG
PS=2). Progression-free survival (PFS) was summarized descriptively, with hazard ratio
(HR) and 95% CI estimated by stratified Cox proportional hazards model. Overall response
rate (ORR) was summarized descriptively, with odds ratio (OR) and 95% CI estimated
by Mantel-Haenszel method. Response and progression were assessed by the Onyx Response
Computer Algorithm using IMWG criteria.
Results: Frailty status was generally proportional between arms, with 27% and 35%
classified as fit, 43% and 36% int, 25% and 27% frail, and 5% and 2% unknown for KdD
and Kd, respectively. Although baseline characteristics of the overall population
were generally balanced, more pts had prior transplant in the KdD vs Kd arm for int
(65% vs 36%) and frail (41% vs 27%) subgroups, and in the frail subgroup fewer pts
were lenalidomide exposed (37% vs 61%)/refractory (25% vs 46%) in the KdD vs Kd arm.
Median duration of treatment for KdD vs Kd was 99 weeks (wks) vs 68 wks for fit pts,
82 wks vs 31 wks for int pts, and 51 wks vs 21 wks for frail pts. Median PFS in KdD
vs Kd arms was not reached (NR) vs 17.6 months (HR 0.64, 95% CI 0.38–1.07) for fit,
NR vs 11.1 months (HR 0.44, 95% CI 0.28–0.69) for int, and 18.5 vs 9.3 months (HR
0.66, 95% CI 0.38–1.14) for frail pts. ORR for KdD vs Kd arms was 89% vs 89% (OR 1.09,
95% CI 0.35–3.38) for fit pts, 87% vs 70% (OR 2.95, 95% CI 1.31–6.62) for int pts,
and 75% vs 54% (OR 2.39, 95% CI 1.09–5.22) for frail pts (Figure). Any grade treatment-emergent
adverse events (TEAEs) occurred in >95% of pts across arms and subgroups. Grade ≥3
TEAEs occurred in 87% (KdD) and 70% (Kd) of fit pts, 84% and 71% of int pts, and 91%
and 90% of frail pts. Fatal TEAEs occurred in 4% and 2% of fit pts, 11% and 9% of
int pts, and 16% and 8% of frail pts in the KdD and Kd arms. Carfilzomib was discontinued
due to TEAEs in 25% vs 17% of fit pts, 22% vs 29% of int pts, and 35% vs 23% of frail
pts. Among TEAEs of interest, acute renal failure occurred in 0 fit pts, 3% vs 14%
of int pts, and 8% vs 5% of frail pts, and infusion reactions occurred in 16% vs 2%
of fit pts, 13% vs 4% of int pts, and 14% vs 13% of frail pts in the KdD vs Kd arms
(Table).
Image:
Summary/Conclusion: Consistent with previous findings of the efficacy and safety benefits
of KdD, a PFS benefit with KdD vs Kd was observed across frailty subgroups, without
increased toxicity. The clinically meaningful ORR benefit in the frail subgroup may
help physicians evaluate treatment options in this challenging group.
P908: FACTORS ASSOCIATED WITH THE CLINICAL PROGNOSTIC PERFORMANCE OF MINIMAL RESIDUAL
DISEASE ASSESSMENT BY NEXT GENERATION FLOW CYTOMETRY IN MULTIPLE MYELOMA
B. Nandakumar1, A. Baranwal1, M. Ebraheem1, H. Olteanu2, M. Shi2, S. Kumar1, W. Gonsalves1,
D. Jevremovic2,*
1Hematology; 2Laboratory Medicine and Pathology, Mayo Clinic, Rochester, United States
of America
Background: Minimal Residual Disease (MRD) negativity (-) using next generation flow
(NGF) cytometry with a minimum sensitivity of 10-5 is strongly associated with improved
progression-free survival (PFS) and overall survival (OS). However, clinicopathologic
factors affecting the prognostic performance of this assay are not clear at this time,
and despite the achievement of MRD (-) status, disease progression is still readily
observed in multiple myeloma (MM).
Aims: We investigated quantitative non-clonal plasma cell factors measurable in the
bone marrow sample that could predict disease progression in MRD (-) MM patients.
Methods: We retrospectively reviewed all patients with MM who underwent a bone marrow
biopsy at the Mayo Clinic, Rochester, USA from July 2017 to December 2020. MRD testing
was done on bone marrow using the established Euroflow protocol with analytic sensitivity
between 10-5 and 2x10-6. In addition to quantitative measurements of the number of
clonal plasma cells, the number of hematogones, polyclonal plasma cells and mast cells
were collected.
The time to next therapy (TTNT) was defined as the time from the date of MRD testing
to the date of starting a new treatment regimen due to disease progression. Patients
who did not require change in treatment for MM were censored at the last known follow-up.
Kaplan-Meier curves and log-rank method were used to compare TTNT.
Results: A total of 1,142 NGF assessments, obtained in 783 different patients with
MM, were found to have an MRD (-) result. The median age of patients at the time of
collection was 63 years. The median absolute number of non-aggregate events captured
was 8,586,360 (range: 840,904 - 9,975,075). For the entire sample cohort, the median
number of polyclonal plasma cells were 1,874 (range: 0 - 121,221), the median number
of hematogones were 70,307 (range: 0 - 6,486,739) and the median number of mast cells
were 734 (range: 0 - 116,834).
Of the 1,142 NGF assessments, follow-up data was available on 675 samples, of which
204 (30%) were associated with disease progression requiring a change in therapy.
Number of polyclonal plasma cells and mast cells greater than their respective medians
were considered elevated and were associated with disease progression (Χ2
p < 0.001 for polyclonal plasma cells and Χ2
p = 0.046 for mast cells).
Patients with higher polyclonal plasma cell events were found to have significantly
higher TTNT compared to those with lower plasma cell events (median NA vs. 31 months,
p < 0.001). The median TTNT for patients with higher mast cell events was also significantly
higher compared to those with lower mast cell events (median NA vs. 36 months, p =
0.002).
Summary/Conclusion: The number of polyclonal plasma cells and mast cells in the bone
marrow sample could be useful in predicting myeloma progression in patients with a
MRD (-) bone marrow assessment by NGF. This observation may be related to the quality
of the specimen (hemodilution), true biologic activity of polytypic plasma cells and
mast cells, or both.
P909: RENAL RESPONSE OF POMALIDOMIDE WITH BORTEZOMIB AND DEXAMETHASONE IN NEWLY DIAGNOSED
MULTIPLE MYELOMA PATIENTS WITH RENAL IMPAIRMENT
Y. Jian1, L. Chang2, M. Shi3, Y. Sun4, X. Chu5, H. Xue6, X. Shen7, J. Ma8, G. Jia9,
Y. Feng10, Z. Xi11, Y. Zhao12, Y. Ma13, J. Xiao14, G. Ma15, Q. Wang16, W. Huang17,
L. Bao18, Y. Dong19, H. Zhou20, C. Sun21, G. Su22, Y. Yan23, Q. Saiyin24, L. Su25,
S. Gao26, W. Tian27, X. Sun28, H. Jing29, D. Gao30, W. Chen1, J. Li2, W. Gao1,*
1Beijing Chaoyang Hospital, Capital Medical University; 2Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing;
3The First Affiliated Hospital of Kunming Medical University, Hematology Research
Center of Yunnan Province, Kunming; 4Chifeng Municipal Hospital, Chifeng; 5The Affiliated
Yantai Yuhuangding Hospital of Qingdao University, Qingdao; 6Affiliated Hospital of
Hebei University, Baoding; 7Heping Hospital Affiliated To Changzhi Medical College,
Changzhi; 8The First Affiliated Hospital of Zhengzhou University, Zhengzhou; 9The
First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of
Secience and Technology, Baotou; 10The Third People’s Hospital Of Datong, Datong;
11Linfen People’s Hospital, Linfen; 12The First Affiliated Hospital, Harbin Medical
University, Harbin; 13Second hospital of Shanxi Medical University, Taiyuan; 14Yantaishan
Hospital Affiliated to Binzhou Medical University, Yantai; 15The Forth Hospital of
Hebei Medical University, Shijiazhuang; 16The Second Affiliated Hospital of Nanchang
University, Nanchang; 17The Fifth Medical Center of People’s Liberation Army (PLA)
General Hospital; 18Beijing Jishuitan Hospital; 19Peking University First Hospital;
20Beijing Luhe Hospital, Capital Medical University, Beijing; 21Union Hospital, Tongji
Medical College, Huazhong University of Science and Technology, Wuhan; 22Cangzhou
Central Hospital, Cangzhou; 23Bayannur Hospital, Bayannur; 24Ordos Central Hospital,
Ordos; 25Shanxi Cancer Hospital, Taiyuan; 26The First Hospital of Jilin University,
Changchun; 27Shanxi Bethune Hospital, Taiyuan; 28The First Affiliated Hospital of
Dalian Medical University, Dalian; 29Peking University Third Hospital, Beijing; 30The
Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
Background: Renal impairment (RI) is a common complication of multiple myeloma (MM),
usually related to poor outcomes. Bortezomib-based regimens have been recommended
as first-line therapy of MM with RI. Recently, pomalidomide also has shown significant
efficacy and an acceptable safety profile in MM patients with RI. However, few prospective
studies evaluated the value of combination of pomalidomide with bortezomib and dexamethasone
(PVD) in these special patients.
Aims: To evaluate the renal response, hematological response and safety of pomalidomide
in combination with bortezomib and dexamethasone (PVD) as first-line therapy in newly
diagnosed MM (NDMM) with RI.
Methods: This study is an ongoing, prospective, open-label, multicenter, phase 2 study
(ChiCTR2100043748). Eligible adult NDMM patients with MM-related RI (defined as estimated
glomerular filtration rate (eGFR) < 40ml/min) who had measurable disease are enrolled
in the study. Exclusion criteria included RI caused by other reasons than tubular
nephropathy, such as renal amyloidosis, light chain deposition disease, etc. Patients
received a maximum of 9 cycles of PVD therapy. For transplant-eligible patients, autologous
stem cell transplantation (ASCT) was administrated after 3 to 6 PVD cycles. Primary
endpoint was renal overall response rate (ORR) at 3 months. Secondary endpoints included
best renal response, hematological response, minimal residual disease (MRD), progression-free
survival (PFS), overall survival (OS) and safety. Both hematological response and
renal response were assessed by International Myeloma Working Group (IMWG) criteria.
Results: Between Feb 28, 2021 and Jan 21, 2022, sixty-one patients were enrolled across
28 Chinese hospitals, with a median age of 61 years (range: 38-79); 60.7% were male.
Fifty-eight (95.1%) patients had cytogenetic information by FISH at diagnosis, fourteen
(24.1%) of them had high-risk cytogenetic abnormalities (defined as t(4;14), t(14;16),
and/or del(17p)). Median serum creatine level and eGFR were 349 μmol/L (range: 155-1718)
and 12.9 mL/min (range: 2.5-36.3), respectively. Twelve (19.7%) patients received
dialysis. Median number of PVD treatment cycles was 4 (range: 1-9). Six patients had
received ASCT. Among 56 patients with evaluable hematological response, ORR was 91.1%
(69.6% ≥VGPR, 41.1% CR). Forty-eight (78.7%) patients had got the primary endpoint
with renal ORR at 3 months, which was 89.4% (48.9% ≥ renal-PR, 35.4% renal-CR). Median
time to best renal response was 1.5 months (range: 0.3-11.0). Dialysis independence
rate was 58.3% (7/12). Median time from first dose of PVD therapy to dialysis independence
was 1 month (range: 0.5-2.0). Rapid reduction of serum free light chain (FLC) was
seen at the first cycle of PVD therapy. Median reduction proportion of involved FLC
at C1D8 was 77.5% (range: 0-98.5%), while at C1D22 was 91.7% (range: 0-99.9%). The
most common adverse effects (incidence >10%) were infection (29.5%), myelosuppression
(18.0%), peripheral neuritis (16.4%) and skin rash (11.5%). Median follow-up time
was 6 months. Twelve patients (19.7%) discontinued PVD treatment, mainly due to toxicity
and patients’ intention. Median PFS and OS has not been reached.
Summary/Conclusion: PVD regimen is an effective and well-tolerated regimen for NDMM
patients with RI, which offered high renal response (renal ORR at 3 months of 89.4%).
P910: DOES MINIMAL RESIDUAL DISEASE OF STEM CELL COLLECTION HAVE PROGNOSTIC IMPACT
ON PATIENTS WITH MULTIPLE MYELOMA?
X. Jingyu1,*, Y. Wenqiang1, F. Huishou1, L. Jiahui1, L. Lingna1, D. Chenxing1, D.
Shuhui1, S. Weiwei1, X. Yan1, Q. Lugui1, A. Gang1
1State Key Laboratory of Experimental Hematology, National Clinical Research Center
for Blood Diseases, Blood Diseases Hospital & Institute of Hematology, Chinese Academy
of Medical Sciences & Peking Union Medical College, Tianjin, China
Background: Multiple myeloma (MM) is a kind of hematological malignancy involved in
monoclonal plasma cells. In most clinical guidelines autologous stem cell transplantation
(ASCT) is considered as the standard of care for transplant-eligible patients (TEMM)
as long as patients achieving partial response (PR). However, it is unclear that under
the circumstance of not achieving complete response (CR) weather it would lead to
the presence of neoplastic plasma cells in the stem cell collection (SCC) and then
result in negative impact on survival prognosis.
Aims: Here, we evaluated the effect of the minimal residual disease (MRD) of SCC in
the TEMM.
Methods: We analyzed retrospectively clinical data of 90 patients with MM undergoing
ASCT between January 1, 2013 to June 1, 2021 and MRD evaluation of both bone marrow
(BM) and SCC were carried out at the same time. MRD was evaluated by multiparameter
flow cytometry (MFC) with 10-4-10-5 sensitivity. The best response was defined as
the deepest response during the follow-up. Here we defined the time from ASCT to disease
progression or death as modified progression-free survival (mPFS) and the time from
ASCT to death as modified overall survival (mOS).
Results: A total of 90 patients met the inclusion criteria. The median age is 54 (37-69)
and 62.2% were males. There were 25 (27.8%) patients presenting high-risk cytogenetic
abnormalities by FISH, defined as the presence of at least one of t (4;14), t (14;16)
or del (17p). Before ASCT 36.7% of patients achieved MRD negativity in BM and 76.7%
in SCC. After the comparison among MRD-positivity status with different sensitivity
and numbers of detectable MRD neoplastic plasma cells, we found that the percentage
of patients with MRD positivity in SCC was much less than that in BM no matter the
sensitivity (P <0.001). Neither mPFS (P=0.861, median mPFS, 40.83m vs. 34.17m for
negativity vs. positivity) nor mOS (P=0.747, median mOS, 67.02m vs. 58.86m for negativity
vs. positivity) was affected by MRD status in SCC. According to MRD status they were
divided into 4 groups, namely MRD negativity in BM and SCC (Group A, 34.4%), MRD positivity
in BM but negativity in SCC (Group B, 22.2%), MRD positivity in BM and SCC (Group
C, 41.1%) as well as MRD negativity in BM but positivity in SCC (Group D, 2.3%). Having
taken the inaccuracy of biopsy into consideration we excluded Group D. The median
follow-up of the cohort was 26.8 months (15.1-105.1m). Patients among the three groups
experienced similar mPFS (P=0.403, median mPFS, 41.07m vs. 34.17m vs. 40.83m, for
Group A, B and C, respectively) and similar mOS (P=0.933, median mOS, 67.02m vs. 58.86m
vs. 58.61m for Group A, B and C, respectively). Achievement of CR with the presence
of MRD negativity was associated with prolonged mPFS and mOS compared with MRD-positive
CR or ≤VGPR (P<0.001, median mPFS, 55.88m vs. 23.03m vs. 27.10m, respectively; P=0.026,
median mOS, 67.02 vs. 46.16m vs. 41.65m, respectively).
Image:
Summary/Conclusion: Our results demonstrated that neoplastic plasma cells in SCC have
little impact on the survival prognosis in MM patients and it is sound to carry out
ASCT when TEMM patients achieving PR. Also, MRD-negativity status can be considered
more valuable on prognosis than CR.
P911: SYMPTOM BURDEN AND ITS IMPACT ON DAILY LIFE AMONG PATIENTS WITH IDIOPATHIC MULTICENTRIC
CASTLEMAN DISEASE (IMCD) – FINDINGS FROM AN INTERNATIONAL IMCD PATIENT SURVEY
F. Shupo1, N. Mason2, E. Jones2, G. Wayi-Wayi1, M. Repasky3, M. Franklin4, J. Brazier4,
N. Zibelnik1, S. Mukherjee5,*
1EUSA Pharma, Hemel Hempstead; 2BresMed Health Solutions, Sheffield, United Kingdom;
3Castleman Disease Collaborative Network, Paso Robles, United States of America; 4School
of Health and Related Research, Sheffield, United Kingdom; 5Hematology and Medical
Oncology, Cleveland Clinic Main Campus, Cleveland, United States of America
Background: Idiopathic Multicentric Castleman disease (iMCD) is a rare lymphoproliferative
disorder driven by proinflammatory hypercytokinemia. The presentation of iMCD is heterogeneous
and can range from mild constitutional symptoms to chronic burdensome symptoms and
in extreme cases life-threatening multiorgan failure. Consequently, disease-related
symptoms in iMCD patients are likely to adversely impact daily life. To date, characterization
of symptom burden and their impact on daily living in iMCD patients has not been systematically
studied.
Aims: We aimed to investigate, characterize, and map the symptoms and associated burden
on daily life experienced by patients with various subtypes of iMCD.
Methods: We developed an international patient-based online survey informed by clinical
practice and published literature to elicit the burden of disease-related symptoms
and effects of symptoms on daily life from a patient perspective. Eligible patients
were > 18 years old with physician-confirmed diagnosis of iMCD-NOS (not otherwise
specified), TAFRO (thrombocytopenia, anasarca, reticulin fibrosis of the bone marrow,
renal dysfunction, and organomegaly) and POEMS—associated MCD (multicentric Castleman
disease with polyneuropathy, organomegaly, endocrinopathy, monoclonal protein, skin
changes). This survey was shared with iMCD communities in Australia, Canada, the UK,
and the US via the Castleman Disease Collaborative Network (CDCN). Burden of Illness
(BOI) was quantitatively measured using a 5-point frequency Likert scale (from 0 ‘Does
not affect my daily life’ to 4 ‘Very severely affects my daily life’), and mean impact
scores (MIS) were calculated. Ethics approvals/waivers were attained for this one-time,
cross-sectional, bespoke 47-question survey.
Results: A total of 57 patient responses were collected during April–November 2021.
On average, patients experienced 7.0 symptoms (range: 0─22) in the week prior to survey
completion. Tiredness was the most frequently reported symptom (77%), followed by
physical weakness (44%) and night sweats (40%). Individual symptoms were clustered
into clinically relevant categories and frequency of symptom groupings reported by
iMCD subtype (Fig 1). Constitutional (82%) and neuropsychiatric (68%) symptoms were
most frequently experienced across all respondents in the week prior to survey completion.
The average number of symptoms by iMCD subtypes were 7.1 by iMCD-NOS, 5.5 by TAFRO
and 8.5 by POEMS-associated MCD. 91% of all patients with iMCD reported experiencing
at least one symptom in the week prior to survey completion. When rating their most
impacted aspects of daily life due to their symptoms (Table 1), patients with iMCD-NOS
reported pain and discomfort (MIS 2.09) and personal relationships (MIS 2.08), patients
with MCD-POEMS reported sexual functioning (MIS 3.40) and pain and discomfort (MIS
2.67), while patients with TAFRO reported sexual functioning (MIS 2.44) and ability
to travel (MIS 2.22).
Image:
Summary/Conclusion: To our knowledge, this is the first study of its kind to characterize
and map the BOI in iMCD patients assessed by symptom frequency, symptom burden (multiplicity
of symptoms) and its adverse effects on different aspects of daily living. Through
our ongoing work we hope to develop a symptom burden score/scale that captures the
symptom severity and its impact on daily living which can then be incorporated as
a patient reported outcome measure for shared treatment decision-making and response
assessment in addition to the laboratory and radiologic parameters.
P912: COVID19 SEVERITY AND THERAPEUTIC OPTIONS IN PATIENTS WITH MULTIPLE MYELOMA
E. Karimova1,*, E. Zhelnova1, E. Baryakh1, K. Yatskov1, E. Zotina1, E. Grishina1,
E. Paramonova1, D. Gagloeva1, E. Misyurina1
1hematology, Moscow city hospital №52, Moscow, Russia
Background: Patients with multiple myeloma have higher risk of SARS-CoV-2 infection
and higher risk of death than patients without MM
Aims: Analysis of features of the course of COVID-19 in patients with MM.
Methods: 89 pts with MM and COVID-19 were treated in the hospital from March 2020
to October 2021, 53 (60%) female, 36 (40%) male. The median age was 64 years (35-88
years). 32 pts (36%) were presented with active myeloma, 19 (21%) – R/R myeloma, 21
(24%) – CR, 17 (19%) - PR or VGPR. The majority of patients - 56 (63%) had III st.
(Durie-Salmon). Kidney damage was presented in 33 pts (37%). The majority of patients
had severe lung damage: CT 2 st. - 26 pts (30%), III st. - 35 (39%), IV st.-17 (19%).
Patients received standard therapy for COVID19: etiotropic (antiviral, viral neutralizing
MAB, convalescent plasma, immunoglobulin), pathogenetic (GCS, interleukin inhibitors,
anticoagulants), therapy for secondary infectious complications.
Results: IL inhibitor therapy was given 76 patients (85%), 35 (39%) required repeated
administration of IL inhibitors. 58 pts (65%) received GCS therapy in various doses.
Anticoagulant therapy was conducted in all patients, 51 pts (57%) received LMWH, 37
(42%) received continuous infusion of heparin, 1 patient received NOAC. The frequency
of hemorrhagic complications on LMWH and heparin therapy was comparable (5.9% and
5.5%), mortality and the frequency of TEC in patients on heparin therapy were higher
(49% and 13% versus 14% and 2% in patients on LMWH therapy, p<0.001 and p=0.034, respectively).
It can be explained by the initial severity of patient’s condition, which required
heparin therapy (30 pts - 81% had an initially high degree of lung damage - CT 3-4
st.)
Secondary infectious complications were reported in 80% of patients: bacterial pneumonia
in 71 pts (80%), sepsis in 24 (27%). Infectious endocarditis (2 pts), meningitis (2
pts), colitis (9 pts), colon gangrene complicated by peritonitis (1 patient) were
less common. Fungal infections were verified in 19 pts (21%): 6 (7%) – mucosal candidiasis,
13 (14%) – invasive aspergillosis. Infection caused by herpes viruses was detected
in 5 patients (6%). There was no significant connection between severity of COVID19,
frequency of secondary infections and mortality and level of normal immunoglobulins.
The median duration of hospitalization was 19 days versus 9 among patients without
MM. 25 patients (28%) were hospitalized in the ICU. The overall mortality was 27%
(24 pts), among this patients 71% (17 pts) had active or R/R myeloma and stage III
of the disease. The mortality in the ICU was 88% (22 pts). The most frequent causes
of fatality were secondary infectious complications in 92% (22 pts), progression of
MM in 8% (2 pts). 27 patients (30%) required chemotherapy during COVID-19.
Summary/Conclusion: Patient with R/R MM have the highest risk of mortality from COVID19.
For patients with multiple myeloma the same COVID19 therapeutic options are effective
as for non-immunodeficient patients. Patients with MM and COVID19 also have a high
risk of secondary infectious complications.
P914: EFFICACY AND SAFETY OF BELANTAMAB MAFODOTIN MONOTHERAPY IN PATIENTS WITH RELAPSED
OR REFRACTORY LIGHT CHAIN AMYLOIDOSIS: A PHASE 2 STUDY BY THE EUROPEAN MYELOMA NETWORK
E. Kastritis1,*, G. Palladini2, M. A. Dimopoulos1, A. Jaccard3, G. Merlini2, F. Theodorakakou1,
D. Fotiou1, M. C. Minnema4, A. Wechalekar5, S. Gkolfinopoulos6, K. Manousou6, P. Sonneveld7,
S. Schönland8
1Department of Clinical Therapeutics, National and Kapodistrian University of Athens,
School of Medicine, Athens, Greece; 2Amyloidosis Research and Treatment Center, University
of Pavia, Pavia, Italy; 3Referral Center for AL amyloidosis, Limoges, France; 4Department
of Hematology, University Medical Center Utrecht, Utrecht, Netherlands; 5Clinical
Haematology, Cancer Division, University College London Hospital, London, United Kingdom;
6Health Data Specialists, Dublin, Ireland; 7Erasmus MC Cancer Institute, Rotterdam,
Netherlands; 8University of Heidelberg, Heidelberg, Germany
Background: Managing patients (pts) with relapsed/refractory (RR) light chain (AL)
amyloidosis is challenging, as there is no current standard treatment, many available
options are associated with low efficacy and toxicity, and options for daratumumab
(DARA)- and bortezomib-exposed patients are limited. Belantamab mafodotin (belamaf),
a multi-modal antibody-drug conjugate targeting BCMA, has shown efficacy and tolerability
in heavily pretreated pts with RR multiple myeloma, including those refractory to
DARA. Since clonal plasma cells in AL amyloidosis and MM are phenotypically similar,
belamaf could be a novel treatment option in AL amyloidosis.
Aims: To evaluate the efficacy and safety of belamaf monotherapy off-label in pts
with RR AL amyloidosis.
Methods: The ongoing prospective, open-label, multinational, phase 2, EMN27 study
(NCT04617925) aims to enroll 36 adult pretreated pts with AL amyloidosis who require
therapy. Pts at Mayo cardiac stage 3b are excluded. Belamaf monotherapy at 2.5mg/kg
is administered by intravenous infusion every 6 weeks for a maximum of 8 cycles; dosing
can be reduced to 1.92mg/kg for toxicity. Per study design, a safety analysis (after
6 pts received ≥1 treatment cycle) and an efficacy analysis (after 13 pts are enrolled)
were planned. The safety analysis revealed no new safety signals, and pt accrual continued
to 13 pts. The efficacy analysis is currently conducted; however, already 3 pts achieved
complete response, or very good partial response (VGPR), or low difference of involved
to uninvolved serum free light chains (dFLC) response and enrollment is continuing
to include all planned pts. This descriptive analysis included pts initiating study
treatment ≥3 months before the cut-off date (15/01/2022).
Results: Of 11 pts included in the analysis, 4 (36.4%) continued treatment by the
cut-off date, and 7 (63.6%) discontinued (disease progression: 5 [45.5%], death: 2
[18.2%]). The pts median age was 69.0 years (range 46.0–80.0), and most were males
(7, 63.6%). At baseline, 3 (27.3%) and 8 (72.7%) pts had New York Heart Association
class I and II symptoms, respectively; the median N-terminal pro-brain natriuretic
peptide, high-sensitivity troponin T, and dFLC were 1,979 pg/mL (range 190.0–4,135.0),
41.6 pg/mL (range 11.0–80.8), and 34.2mg/dl (range 4.4–279.1), respectively. Except
for the heart, commonly involved organs were the nervous system (4 pts, 36.4%) and
the soft tissue (2 pts, 18.2%). The median number of previous AL amyloidosis treatments
was 3.0 (range 1.0–7.0), including DARA. The median duration of belamaf therapy was
3.1 months (range 1.4–5.7). At a median follow up of 9.4 months (range 3.1–10.0),
the overall response rate was 72.7% (8 pts; VGPR: 27.3% [3 pts] and partial response:
45.5% [5 pts]). Median time to first hematological response was 8.5 days (range 1.0–28.0)
and to VGPR or better 15.0 days (range 8.0–15.0). The 3-month organ (heart, kidney,
or liver) response rate was 36.4% (4 pts). All pts had ≥1 non serious adverse event
(SAE). Four (36.4%) pts had ≥1 SAE, including 2 (18.2%) pts with a belamaf-related
grade 2 and 4 visual impairment (1 [9.1%] pt each). Eight (72.7%) pts had ≥1 adverse
event of special interest. Two (18.2%) pts had a fatal SAE (pneumonia and intestinal
perforation, 1 [9.1%] pt each), both unrelated to belamaf.
Summary/Conclusion: In this prospective study, belamaf monotherapy induced rapid,
clinically meaningful responses with a manageable safety profile in heavily pretreated
pts with RR AL amyloidosis. As the study progresses, additional data will be generated.
P915: EFFICACY AND SAFETY OF DARATUMUMAB MONOTHERAPY IN NEWLY DIAGNOSED PATIENTS WITH
STAGE 3B LIGHT CHAIN AMYLOIDOSIS: A PHASE 2 STUDY BY THE EUROPEAN MYELOMA NETWORK
E. Kastritis1,*, M. C. Minnema2, M. A. Dimopoulos1, G. Merlini3, F. Theodorakakou1,
D. Fotiou1, A. Huart4, K. Belhadj5, S. Gkolfinopoulos6, K. Manousou6, P. Sonneveld7,
G. Palladini3
1Department of Clinical Therapeutics, National and Kapodistrian University of Athens,
School of Medicine, Athens, Greece; 2Department of Hematology, University Medical
Center Utrecht, Utrecht, Netherlands; 3Amyloidosis Research and Treatment Center,
University of Pavia, Pavia, Italy; 4Department of Nephrology and transplantation,
Rangueil University Hospital, Toulouse; 5Lymphoid Malignancies Unit, Henri Mondor
Hospital, Créteil, France; 6Health Data Specialists, Dublin, Ireland; 7Erasmus MC
Cancer Institute, Rotterdam, Netherlands
Background: Cardiac involvement and severity of cardiac dysfunction in light chain
(AL) amyloidosis is a critical prognostic factor. Patients (pts) at Mayo cardiac stage
3b have a poor prognosis with a median overall survival (OS) of just 4 months and
high rates of early death with current therapies; thus, there is a need for novel,
non-toxic, effective treatments for these pts. Daratumumab (DARA), a human anti-CD38
antibody, has shown efficacy and tolerability in pts with AL amyloidosis.
Aims: To evaluate the efficacy and safety of DARA monotherapy used off-label in newly
diagnosed pts with stage 3b AL amyloidosis.
Methods: The ongoing EMN22 phase 2, multinational, open-label study (NCT04131309)
aims to enroll 40 newly diagnosed pts with stage 3b AL amyloidosis. Eligible adult
pts have high-sensitivity troponin T (hsTnT) >54 pg/mL and N-terminal pro-brain natriuretic
peptide (NT-proBNP) ≥8,500 pg/mL. DARA monotherapy, 16 mg/mL by intravenous infusion
(09/2019–01/2020) and 1,800 mg by subcutaneous injection (01/2020 and thereafter),
is administered weekly during cycles (C)1 and 2, every 2 weeks for C3–6, and every
4 weeks thereafter. Pts not achieving a hematological very good partial response (VGPR)
or better by the end of C3 can receive additional weekly bortezomib and low dose dexamethasone
(Vd). Treatment continues up to 2 years from initiation or until disease progression
or initiation of a new therapy. Primary endpoint is OS rate at 6 months. This descriptive
analysis included pts initiating treatment ≥6 months before the cut-off date (14/01/2022);
the median (95% confidence interval [CI]) OS was obtained by Kaplan-Meier analysis.
Results: Of 27 pts included, 8 (30%) continued study treatment by the cut-off date
and 19 (70%) had discontinued. The pts median age was 68 (range 45–84) years, and
most were male (16, 59%). At screening, 10 (37%) and 17 (63%) pts had New York Heart
Association class II and IIIA symptoms, respectively; the median NT-proBNP was 15,512
pg/mL (range 8,816–72,522), hsTnT was 133 pg/mL (range 60–692), and the difference
of involved to uninvolved free light chains was 406 mg/l (range 24–3,377). Beyond
the heart, the median number of other organs involved was 2 (range 0–5), most commonly
kidneys (14 pts, 52%) and peripheral nerves (11 pts, 41%). The median duration of
DARA therapy was 7 months (range <1–24); seven (26%) pts received additional Vd. At
a median observation time of 8 months (range <1–11), the overall response rate (ORR)
was 67% (18 pts; complete response [CR]:19.0% [5 pts], VGPR:37% [10 pts], partial
response:11% [3 pts]). The ORRs at 1, 2, and 3 months were 59% (16 pts), 63% (17 pts),
and 63% (17 pts), respectively. Median time to first response was 7 days (range 6–114),
and to VGPR or better 54 days (range 6–219). Median OS was 9 months (95% CI, 3–not
reached). The 6- and 12-month median (95% CI) OS rates were 63% (42–78) and 49% (28–67),
respectively. Twenty-five (93%) pts had ≥1 non-serious adverse event. Twenty (74%)
pts had ≥1 serious adverse event (SAE), comprising 15 (56%) pts with ≥1 cardiac-related
SAE and 11 (41%) pts with a fatal SAE. Six SAEs were treatment-related: 2 with DARA
(grade 3 pneumonia, grade 5 sepsis); 3 with bortezomib (grade 2 fatigue, grade 2 fall,
and grade 3 troponin I increase); and 1 with dexamethasone (grade 3 cardiac failure).
Summary/Conclusion: Among pts with Mayo stage 3b AL amyloidosis, a subgroup with poor
prognosis, DARA monotherapy induced rapid and deep hematological responses and no
new safety signals; the median OS surpassed that reported previously.
P916: COEXISTENCE OF ≥2 HIGH-RISK MOLECULAR ABNORMALITIES SUPERVENES THE PROGNOSTIC
VALUE OF THE REVISED INTERNATIONAL STAGING SYSTEM FOR MYELOMA: REAL-WORLD DATA ANALYSIS
FROM THE GREEK MYELOMA STUDY GROUP
E. Katodritou1,*, D. Dalampira1, T. Triantafyllou1, M. Gavriatopoulou2, S. Delimpasi3,
A. Pouli4, T. Papadopoulou1, E. Verrou1, A. Sevastoudi1, K. Tsirou1, L. Katsika1,
G. Douganiotis1, N. Karampatzakis1, T.-E. Metallinou1, V. Palaska1, M. Kotsopoulou5,
C. Lalayianni6, M.-C. Kyrtsonis7, E. Spanoudakis8, D. Maltezas5, A. Chatzivasili9,
M.-A. Dimopoulos2, E. Terpos2, E. Kastritis2
1Hematology, Theagenio Cancer Hospital, Thessaloniki; 2Clinical Therapeutics, National
and Kapodistrian University of Athens; 3Hematology and Bone Marrow Unit, Evangelismos
General Hospital; 4Hematology, Agios Savvas Cancer Hospital; 5Hematology, Metaxa Cancer
Hospital, Athens; 6Hematology and Bone Marrow Unit, George Papanicolaou General Hospital,
Thessaloniki; 7First Department of Propaedeutic Internal Medicine, National and Kapodistrian
University of Athens, Athens; 8Hematology, University Hospital of Alexandroupolis,
Alexandroupolis; 9Hematology, Venizelion General Hospital, Heraclion, Greece
Background: Revised International Staging System (R-ISS) has improved the prognostic
value of ISS in multiple myeloma (MM). Recently, the Mayo Additive Staging System
incorporated +1q21 to determine a 5-factor 3-tier system, providing an add-on value
on R-ISS. The prognostic impact of the coexistence of ≥2 high-risk molecular abnormalities,
including +1q21, compared to R-ISS, has not been validated adequately.
Aims: The aim of this study was to evaluate the prognostic impact on survival of the
coexistence of ≥2 high-risk molecular abnormalities, defined as Ultra High Risk (UHR)
MM, in comparison with R-ISS and other established prognostic markers, in the real-world
setting.
Methods: We analyzed the data of 1352 consecutive newly diagnosed MM patients (M/F:
655/697, median age: 66, range: 29-87, IgG: 817, IgA: 363, light chain: 144, IgD:
9, IgM: 3, non-secretory: 16), treated between 2002-2021 and which had been tested
for molecular abnormalities i.e. del17p, t(14;16), t(4;14) and +1q21 using fluorescence
in situ hybridization. Patients with ≥2 high-risk features were classified as UHR.
We compared the two groups for age, performance status, ISS, R-ISS, lactate dehydrogenase
(LDH), albumin, hemoglobin (Hb), β2-microglobulin, estimated glomerular filtration
rate (eGFR), 1st and 2nd line therapies and response rates. A Cox regression model
was used to determine independent prognostic factors for overall survival (OS). Progression-free
survival (PFS) and OS were plotted with Kaplan-Meier; a p<0.05 was considered as statistically
significant.
Results: One hundred sixteen patients (9%) were classified in the UHR group vs. 1236
(91%) in the non-UHR group; 106 patients had 2, and 10 patients had 3 molecular abnormalities.
The most common combination of high-risk features was +1q21 plus t(4;14) (40%). Median
age, sex, performance status, LDH and serum albumin, did not differ, whereas the UHR
group had lower eGFR, higher β2-microglobulin and lower Hb (p<0.05). Early stage (ISS1/R-ISS1)
was more frequent in the non-UHR group (p<0.05); 1st line treatment, including autologous
transplantation (ASCT), and second line treatment were well balanced between groups
(p<0.05); 66% of patients overall, received triplet/quadruplet combinations and 29%
underwent ASCT upfront. Overall response rate after induction therapy was 86% and
did not differ between groups (p<0.05). Complete response was lower in the UHR group
(11% vs. 19%; p=0.03). After a median follow up of 49 months (95% CI: 44-54), 59%
of patients were alive. Median PFS was significantly shorter for the UHR group (15.8
vs. 31.9 months, p<0.001; HR: 0.45, 95% CI: 0.36-0.58). Median OS was 28 months (95%
CI:18-38) for patients in the UHR group vs. 69 months (95% CI: 61-77) for others (p<0.001).
In the univariate analysis, age, anemia, eGFR, upfront ASCT, R-ISS and UHR myeloma
were independent predictors for OS (p<0.05). In the multivariate analysis UHR myeloma
was the strongest independent predictor for OS (p<0.001; HR: 0.42), supervening the
prognostic value of R-ISS (R-ISS1 vs. R-ISS2 HR=0.58, R-ISS2 vs. R-ISS3 HR: 0.75).
Additionally, UHR status singled out a distinct group within R-ISS2 patients with
significantly worse OS (38 months, 95% CI: 28-48 vs. 64mo, 95% CI: 55-72) (p<0.001).
Summary/Conclusion: According to our analysis of a large cohort of newly diagnosed
MM patients UHR myeloma, was the strongest independent predictor for OS, supervening
the prognostic value of R-ISS. Moreover, UHR status could serve as an additional prognostic
marker for R-ISS2 patients, helping thus to optimize therapeutic approach of MM patients.
P917: HOW COMORBID ARE OUR MYELOMA PATIENTS AND HOW MANY MAKE IT TO THE SECOND-LINE
TREATMENT: REAL-WORLD DATA OF 251 MYELOMA PATIENTS TREATED IN THE HEMATOLOGICAL NETWORK
OF THE OEGK
F. Keil1,*, P. Attalla1
13rd Medical Department for Hematology and Oncology, Hanusch hospital, Vienna, Austria
Background: Comorbid conditions have a negative impact on overall survival (OS) in
multiple myeloma (MM) patients. Furthermore, a European multi-center study reported
that only 61% of patients proceed to second-line treatment.
Aims: The objective of this study was to determine the prevalence of common comorbidities
and their impact on OS. Furthermore, we examined the mortality during first-line treatment
and the percentage of patients proceeding to second-line treatment.
Methods: This retrospective study was conducted in the hematology care network “Hämatologieverbund
der Österreichischen Gesundheitskasse (OEGK)”, consisting of the Hanusch hospital
and three outpatient centers, in Vienna, Austria. We included all patients who were
observed and/or treated for multiple myeloma between 2012 and 2018 within the hematology
care network. Follow-up was conducted until March 2021.
Results: We analyzed 251 patients (median age 69 years). Prevalent concomitant conditions
at diagnosis were hypertension (67.4%), further cardiovascular diseases excluding
hypertension (23.1%), obesity (15.2%), diabetes (13.8%) and chronic lung disease (9.2%).
For the evaluation of renal impairment, we used the CCI, which defines kidney disease
as a creatinine >3mg/dl or the need for dialysis, as well as the chronic kidney disease
(CKD) classification system. While 7.8% of patients fulfilled the CCI criteria for
kidney disease, 39.5% had a CKD stage ≥3A.
Comorbidity in general, as well as specific comorbid conditions had a negative effect
on overall survival. Patients with a CCI score of 0 points at diagnosis showed a median
OS of 8.7 years compared to 3.8 years and 2.6 years for patients with a score of 1
point and ≥2 points, respectively (p <0.001). We also observed a reduced OS of 4.9
years in patients with at least one creatinine clearance (CrCl) value <60 ml/min throughout
the observation period compared to 16.4 years in patients with a CrCl consistently
≥60 ml/min (p <0.001).
In total, 232 patients received a first-line treatment. During the observation period
after first-line therapy, 146 patients proceeded to second-line treatment due to progress,
30 showed no sign of progress so far and 12 had an ongoing first-line maintenance
therapy. 21 patients were referred from other centers for a second opinion and/or
ASCT. These patients were still alive at the end of the observation period, but data
about further treatment lines were missing. One patient eligible for second-line treatment
refused further therapeutic measures. Furthermore, 22 patients died during first-line
treatment or the subsequent treatment-free interval. Of these 22 patients, 11 died
during active first-line treatment due to progression of myeloma or treatment-related
toxicity. The other 11 patients died after completion of first-line treatment during
the observation period due to secondary neoplasms (4) and infection (2). For the remaining
five patients the cause of death was unknown. In total, 90.5% of patients who had
received a first-line treatment were or still are eligible for second-line treatment.
Summary/Conclusion: Comorbid conditions were prevalent in our population and exerted
a major impact on mortality. In comparison to the CCI, the chronic kidney disease
classification system showed superior sensitivity for detection of renal impairment,
which is a good predictor of survival. In contrast to previously published data, a
high proportion of patients were eligible for second-line treatment. Disease-specific
mortality prevented only a small number of patients from receiving second-line treatment.
P918: HIGH RESPONSES RATES WITH SINGLE AGENT BELANTAMAB MAFODOTIN IN RELAPSED SYSTEMIC
AL AMYLOIDOSIS
J. Khwaja1,*, J. Bomsztyk2, S. Mahmood2, B. Wisniowski2, R. Shah1, A. Tailor1, K.
Yong1, R. Popat1, N. Rabin1, C. Kyriakou1, J. Sive1, S. Worthington1, A. Hart1, E.
Dowling1, N. Correia1, C. Bygrave3, A. Rydzewski4, K. Jamroziak5, A. Wechalekar2
1Department of Haematology, University College London Hospital; 2National Amyloidosis
Centre, University College London (Royal Free Campus), London; 3Department of Haematology,
University Hospital of Wales, Cardiff, United Kingdom; 4Department of Internal Medicine,
Nephrology and Transplantation Medicine, Central Clinical Hospital of the Ministry
of Internal Affairs; 5Department of Hematology, Transplantation and Internal Medicine,
Medical University of Warsaw, Warsaw, Poland
Background: Systemic AL amyloidosis is an incurable relapsing plasma cell disorder.
Despite therapeutic advances, there are no approved treatments for relapse disease.
Treatment is often challenging due to underlying organ dysfunction. Belantamab mafodotin
is an antibody-drug conjugate targeting B-cell maturation antigen with approval for
relapsed refractory myeloma. In multiply pre-treated myeloma, the DREAMM-2 phase II
trial showed an overall response rate of 32% for those with 2.5 mg/kg dose administered
every three weeks with 2/3rd patients reporting keratopathy. A small case series of
6 patients with relapsed AL amyloidosis (Zhang et al, ASH 2021) was recently reported
and a phase 2 trial is recruiting for patients with refractory amyloidosis (NCT04617925).
Aims: We report our initial results using Belantamab monotherapy for the treatment
of patients with AL amyloidosis with relapsed disease.
Methods: Data for consecutive patients who were administered Belantamab at a specialist
referral centre, National Amyloidosis Centre, University College London, was analysed.
Results: Eleven patients were included 8 male, 3 female. Median age at Belantamab
initiation was 65 (range 42-74) years. Eight patients had λ AL-type and three κ AL-type.
At diagnosis, median involved free light-chain concentration was 534 (range 73-7181)
mg/l. A median of two organs involved at baseline (range 1-3): 4 had cardiac involvement
(half Mayo stage 2; half Mayo stage 3a) and 8 had renal involvement. The median prior
lines of therapy was 3 (range 2-5) with all exposed to prior immunomodulatory drugs,
proteasome inhibitors and 73% to anti-CD38 antibody treatments. Thirty-six percent
had relapsed after melphalan-conditioned autologous stem cell transplantation.
A median of 3 cycles of belantamab were delivered (range 1-8). The most frequent adverse
event was ocular toxicity which was experienced in 8 patients (grade 1-3), necessitating
dose modification of the three-weekly schedule. One patient developed transient grade
1 dyspnoea and liver dysfunction. No patients developed cytopenias, unlike previous
reports (Zhang et al, 2021), nor infections beyond COVID (2 patients mild with no
hospital admissions). The majority of the cohort required dose reduction either at
initiation (patient 4, due to end stage renal failure; patient 11, post-renal transplant)
or during therapy (n=5; three to 1.9mg/kg, two to 1.25mg/kg) due to ocular toxicity.
Only one patient remained on the standard dose of 2.5mg/kg for ≥3 cycles. Ocular toxicity
improved after treatment interruption (drug intervals 4-6 weeks) and no patients required
complete treatment cessation. One patient is too early to assess response. Haematological
responses (PR or better) were seen in 7 patients with 3 complete responses and two
very good partial responses (VGPR) which are ongoing. Both renal patients (patients
4 and 11) commenced a dose of 1.25mg/kg and sustained a VGPR with no additional toxicity.
Patient 3 had a 42% reduction in sFLC after two doses but then a prolonged gap due
to keratopathy and has lost the response. There were no cardiac or renal toxicities
observed.
Image:
Summary/Conclusion: Belantamab mafodotin demonstrates significant activity in patients
with heavily pre-treated AL amyloidosis with 70% achieving a ≥PR. Apart from keratopathy
requiring dose modification, no other substantial toxicity was observed. Two patients
with renal impairment (stage V CKD and ESRD) and one patient post-renal transplant
tolerated treatment with no additional toxicity. Belantamab mafodotin shows promise
in treatment of relapsed AL and needs further prospective trials.
P919: SIGNIFICANCE OF IHC AND BIOCHEMICAL MARKERS OF BONE METABOLISM FOR PREDICTING
OSTEODESTRUCTIVE SYNDROME IN PLASMA CELL PROLIFERATION
Z. Kozich1,*, V. Martinkov1, M. Zhandarov1, J. PUGACHEVA1, S. Mihno1, N. Klimkovich2
1The Republican Research Center for Radiation Medicine and Human Ecology, Gomel; 2Belarusian
Medical Academy for Postgraduate Education, Minsk, Belarus
Background: Multiple myeloma (MM) is a malignant disease of a lymphoid nature accompanied
by the proliferation of tumor plasma cells, in its development passing through the
stage of monoclonal gammopathy of undetermined significance (MGUS) and smoldering
myeloma (SM). One of the main manifestations of MM is the lesion of the skeleton bones,
which may already manifest at the stage of MGUS and SM and further it can lead to
a decrease in the quality of life of patients. Our work is devoted to the study of
the role of markers that contribute to the detection of the progression of the destructive
syndrome at the stage of MGUS and SM.
Aims: To study significance of IHC and biochemical markers of bone metabolism for
predicting osteodestructive syndrome in MGUS and SM.
Methods: All patients underwent aspiration and BM biopsy for cytological and histopathological
assessment of PC infiltration. An immunological study of blood serum and the determination
of biochemical markers of bone metabolism of serum were also performed. All patients
underwent CT and MRI of the whole body. The diagnosis of MGUS was based on international
criteria: the presence of less than 10% of clonal plasma cells in the bone marrow
aspirate, the concentration of M-protein in the blood serum <30 g/l. Among patients
with MM, a group without osteodestructive lesions of the skeleton bones was identified.
Statistical processing of the results was carried out using the Statistica 6.1 software
package. Differences were considered statistically significant at p<0.05.
Results: The study included 132 patients (63 MM patients and 68 MGUS patients), who
did not differ in age at the time of diagnosis, p = 0.089, the median age was 64.0
years (25% and 75% - 56.0 to 69, 0) and 61.0 years (25% and 75% - 53.0 and 66.0),
respectively. In the MG group, female patients predominated (70.1%), they were significantly
more common than in the MM group (53.1%), p=0.045.
Damage to the skeleton bones during primary diagnosis (including SP) was detected
in 37.4% (68) of cases. Bone tissue destruction was more common in males (p=0.029).
Changes in bone tissue for patients with MGUS and SM detected by MRI in most cases
are presented as a diffuse lesion, foci of bone tissue rarefaction without obvious
foci of destruction or SP. The presence of destructive lesions was more frequently
detected in MGUS patients with subsequent progression to MM (p<0.002). Time to progression
was about 14 months on average (range from 3 to 25 months).
When analyzing the obtained results, in MGUS patients, an excess of the β CrossLaps
level in serum occurred in 7.8% of cases. At the stage of MGUS, 25.3% of patients
(according to the level of osteocalcin) and 16.1% (according to the level of VAR)
had disorders in the processes of bone tissue formation. Bone tissue destruction was
detected more often in patients with IgM secretion (p=0.014), the ratio of immunoglobulin
light chains κ/λ <0.1 and >10 and more than 10% of CD 138+ plasma cells in immunohistochemical
studies.
Summary/Conclusion: At the stage of MGUS, disturbances in the processes of bone tissue
remodeling occur, which is accompanied by the appearance of deviations in the level
of biochemical markers (osteocalcin, β CrossLaps). Therefore, these markers have the
potential to be used to identify individuals at increased risk of developing a destructive
syndrome, and in conjunction with other risk factors (IgM secretion of more than 10%
of CD 138+ plasma cells on immunohistochemistry) and to identify patients at increased
risk of progression during MM.
P920: EFFECTIVENESS AND SAFETY OF SELINEXOR-BASED REGIMEN IN THE TREATMENT OF RELAPSED
AND REFRACTORY MULTIPLE MYELOMA: A MULTICENTER REAL-WORLD STUDY FROM CHINA
L. Kuang1, B. Fang2, W. Chen3, A. Liu3, C. Li4, L. Bao5, C. Fu6, J. Chen7, H. Li7,
Y. Pang8, A. Liao9, Y. Liang10, Y. Wei11, J. Li1,*
1First Affiliated Hospital of Sun Yat-sen University, Guangzhou; 2Affiliated Cancer
Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou; 3Beijing Chao-yang
Hospital, Capital Medical University, Beijing; 4Tongji Hospital of Tongji Medical
College, Huazhong University of Science and Technology, Wuhan; 5Beijing Jishuitan
Hospital, Beijing; 6The First Affiliated Hospital of Soochow University, Suzhou; 7Sichuan
Provincial Hospital of University of Electronic Science and Technology of China, Chengdu;
8The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou; 9Shengjing
Hospital of China Medical University, Shenyang; 10Sun Yat-sen University Cancer Center;
11Nanfang Hospital, Southern Medical University, Guangzhou, China
Background: Selinexor is the first approved oral selective nuclear exportin inhibitor.
Clinical trials have shown that selinexor-based regimens improved the survival of
RRMM patients. However, there are limited reports on the effectiveness and safety
of selinexor in the real world.
Aims: To evaluate the effectiveness and safety of selinexor-based regimens in the
treatment of patients with relapsed and refractory multiple myeloma (RRMM) in real
world settings.
Methods: The clinical data of 53 patients with RRMM who were treated with selinexor-based
regimens in 11 centers in China from July 2020 to December 2021 were retrospectively
analyzed, and the response, survival and side effects were evaluated.
Results: The median age of the 53 patients with RRMM at the time of treatment with
the selinexor-based regimens was 60 years (range: 41-79). The median time since initial
diagnosis was 30 months (range: 1-156). The median number of prior lines of therapy
was 4 (range: 1-11), including 22 (41.5%) cases who had received autologous hematopoietic
stem cell transplantation in the past, 8 cases (15.0%) who had been exposed to CART,
34 cases (64.1%) who were double-class refractory (proteasome inhibitors and immunomodulators),
and 20 cases (33.7%) who were triple-class refractory(proteasome inhibitors, immunomodulators
and daratumumab). The number of patients with high-risk cytogenetics [17p-/t (14;16)/t
(4;14)/1q21] at onset was 24 (66.7%), and 20 (55.6%) at the time of the lastest relapse.
The responses were evaluable in 47 patients, and the overall response rate (ORR, ≥PR)
was 44.7%, including 3 cases with CR (6.4%), 4 cases with VGPR (8.5%), and 14 cases
with PR (29.8%). The median follow-up time was 2.75 months (range: 0.14-17.5). In
the responders, the median time to first response was 1.07 months (range: 0.25-2.71),
and the median duration of response (DOR) was 7.75 months (95% CI: 4.705-10.795).
The median progression-free survival (PFS) was 6.107 months (95%CI: 2.856, 9.358),
and the median overall survival (OS) had not been reached. The major grade 3-4 adverse
effets were hematological toxicity, and the incidences of grade 3-4 neutropenia, lymphopenia,
and thrombocytopenia were 41.2%, 37.9%, and 53.0%, respectively. The incidence of
dose reductions of selinexor due to myelosuppression was 7.1%. Among the non-hematological
adverse effects, the most common grade 3-4 adverse effects were nausea and vomiting
(38.7%), fatigue (18.6%), and infection (18.6%).
Image:
Summary/Conclusion: In the real world, the selinesor-based regimens have good effectiveness
and safety profile in the treatment of RRMM.
P921: UPDATED EFFICACY AND SAFETY RESULTS OF TECLISTAMAB, A B-CELL MATURATION ANTIGEN
X CD3 BISPECIFIC ANTIBODY, IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA FROM
MAJESTEC-1
J. Martínez-López1,*, P. Moreau2, S. Z. Usmani3, A. Garfall4, N. W. van de Donk5,
J. F. San-Miguel6, A. Oriol7, A. Chari8, L. Karlin9, M.-V. Mateos10, R. Popat11, A.
K. Nooka12, S. Sidana13, D. Trancucci14, R. Verona15, S. Girgis15, C. Uhlar15, T.
Stephenson15, A. Banerjee15, A. Krishnan16
1Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre Universidad
Complutense, CNIO, CIBERONC, Madrid, Spain; 2Hematology Clinic, University Hospital
Hôtel-Dieu, Nantes, France; 3Levine Cancer Institute/Atrium Health, Charlotte, NC;
4Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA, United States of America; 5Amsterdam University Medical Center,
Vrije Universiteit Amsterdam, Amsterdam, Netherlands; 6Clínica Universidad de Navarra,
CIMA, CIBERONC, IDISNA, Pamplona; 7Institut Català d’Oncologia and Institut Josep
Carreras, Hospital Germans Trias i Pujol, Barcelona, Spain; 8Mount Sinai School of
Medicine, New York, NY, United States of America; 9Service d’Hématologie Clinique,
Centre Hospitalier Lyon Sud, Pierre-Bénite, France; 10University Hospital of Salamanca/IBSAL/CIC/CIBERONC,
Salamanca, Spain; 11University College London Hospitals, NHS Foundation Trust, London,
United Kingdom; 12Winship Cancer Institute, Emory University, Atlanta, GA; 13Stanford
University School of Medicine, Stanford, CA; 14Janssen Research & Development, Raritan,
NJ; 15Janssen Research & Development, Spring House, PA; 16City of Hope Comprehensive
Cancer Center, Duarte, CA, United States of America
Background: Teclistamab (JNJ-64007957), a bispecific antibody targeting both B-cell
maturation antigen (BCMA) and CD3 receptors, mediates T cell activation and subsequent
lysis of BCMA-expressing myeloma cells. In the multi-cohort, open-label, phase 1/2
MajesTEC-1 (NCT03145181) study, the safety and efficacy of teclistamab in patients
with relapsed/refractory multiple myeloma (RRMM) who previously received ≥3 lines
of therapy (LOT) are being investigated. In phase 1, weekly subcutaneous dose of teclistamab
1.5 mg/kg, preceded by step-up doses of 0.06 and 0.3 mg/kg, was identified as the
recommended phase 2 dose (RP2D). Initial results from phase 1/2 showed that teclistamab
at the RP2D was well tolerated and provided encouraging efficacy in patients with
no prior exposure to an anti–BCMA-targeted treatment.
Aims: We report updated efficacy and safety results from MajesTEC-1 in patients treated
at the RP2D, including additional patients and longer follow-up.
Methods: MajesTEC-1 included patients aged ≥18 years with documented MM (as per the
International Myeloma Working Group [IMWG] criteria) who had received ≥3 prior LOT
including a proteasome inhibitor, an immunomodulatory drug, and an anti-CD38 antibody.
Patients previously exposed to BCMA-targeted therapy were not eligible in Phase 1.
Patients received teclistamab at the RP2D. Overall response rate (ORR, assessed per
the IMWG 2016 criteria) was the primary endpoint. CTCAE v4.03 (cytokine release syndrome
[CRS] and ICANS graded per ASTCT guidelines) was used for grading adverse events (AEs).
Results are based on a Sep 7, 2021 data cutoff for safety and a Nov 9, 2021 data cutoff
for efficacy (N=165).
Results: The median age was 64 y (range 33–84), 58% were male, and patients had received
5 (range 2–14) median prior LOT; 100% of patients were triple-class exposed, 78% were
triple-class refractory, 70% were penta-drug exposed, and 30% were penta-drug refractory.
ORR was 64% (95% CI 56–72), with 30% of patients achieving a complete response or
better. Durable responses were observed, which deepened over time. The 12-month duration
of response (DOR) rate was 66% (95% CI 49–79); median DOR was not reached. A reduction
in soluble BCMA was observed in the first cycle of treatment in a majority of patients
who responded to teclistamab. Neutropenia (65%; grade 3/4: 57%), anemia (50%; grade
3/4: 35%), thrombocytopenia (38%; grade 3/4: 21%), and lymphopenia (34%; grade 3/4:
32%) were the most common hematologic AEs. Infections occurred in 104 patients (63%;
grade 3/4: 35%). The most common nonhematologic AE was CRS in 72% patients (grade
3, 0.6%; no grade 4/5). The median (range) time to CRS onset was 2 days (1–6) and
median duration was 2 days (1–9). A total of 9 ICANS events (all grade 1/2; all resolved)
were reported in 5 (3%) patients, of which 7 ICANS events were concurrent with CRS
(all resolved). No dose reductions due to AEs were required, and no treatment-related
deaths were reported.
Summary/Conclusion: The deep and durable responses achieved with teclistamab in patients
with highly refractory MM were reaffirmed with data from ~9 months of follow-up. No
new safety signals were identified. Additional data with longer follow-up, including
subgroup analyses and progression-free survival, will be presented.
P922: HEALTH-RELATED QUALITY OF LIFE WITH TECLISTAMAB, A B-CELL MATURATION ANTIGEN
X CD3 BISPECIFIC ANTIBODY, IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA FROM
MAJESTEC-1
R. Popat1,*, P. Moreau2, S. Z. Usmani3, A. Garfall4, M.-V. Mateos5, J. F. San-Miguel6,
A. Oriol7, A. K. Nooka8, L. Rosinol9, A. Chari10, L. Karlin11, A. Krishnan12, N. Bahlis13,
T. Martin14, B. Besemer15, J. Martínez-López16, M. Delforge17, J. Fastenau18, K. S.
Gries18, N. W. van de Donk19
1University College London Hospitals, NHS Foundation Trust, London, United Kingdom;
2Hematology Clinic, University Hospital Hôtel-Dieu, Nantes, France; 3Levine Cancer
Institute/Atrium Health, Charlotte, NC; 4Abramson Cancer Center, Perelman School of
Medicine, University of Pennsylvania, Philadelphia, PA, United States of America;
5University Hospital of Salamanca/IBSAL/CIC/CIBERONC, Salamanca; 6Clínica Universidad
de Navarra, CIMA, CIBERONC, IDISNA, Pamplona; 7Institut Català d’Oncologia and Institut
Josep Carreras, Hospital Germans Trias i Pujol, Barcelona, Spain; 8Winship Cancer
Institute, Emory University, Atlanta, GA, United States of America; 9Hospital Clínic,
IDIBAPS, University of Barcelona, Barcelona, Spain; 10Mount Sinai School of Medicine,
New York, NY, United States of America; 11Service d’Hématologie Clinique, Centre Hospitalier
Lyon Sud, Pierre-Bénite, France; 12City of Hope Comprehensive Cancer Center, Duarte,
CA; 13Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, CA; 14University
of California San Francisco, San Francisco, CA, United States of America; 15University
of Tuebingen, Tuebingen, Germany; 16Haematological Malignancies Clinical Research
Unit, Hospital 12 de Octubre Universidad Complutense, CNIO, CIBERONC, Madrid, Spain;
17Universitaire Ziekenhuizen Leuven, Leuven, Belgium; 18Janssen Research & Development,
Raritan, NJ, United States of America; 19Amsterdam University Medical Center, Vrije
Universiteit Amsterdam, Amsterdam, Netherlands
Background: Because patients with multiple myeloma (MM) have impaired health-related
quality of life (HRQoL), it is important that patient-reported outcomes (PROs) are
assessed in addition to clinical outcomes. Teclistamab (JNJ-64007957) is a B-cell
maturation antigen (BCMA) x CD3 bispecific antibody designed to mediate T cell-induced
lysis of BCMA-expressing MM cells. In the phase 1/2 MajesTEC-1 trial, initial results
from the pivotal cohort (patients who received ≥3 prior lines of therapy [LOT], including
a proteasome inhibitor, an immunomodulatory drug, and an anti-CD38 antibody) showed
that teclistamab was well tolerated with encouraging efficacy.
Aims: We report PROs of the pivotal cohort from MajesTEC-1.
Methods: MajesTEC-1 included patients aged ≥18 years with documented relapsed/refractory
multiple myeloma (RRMM) as per the International Myeloma Working Group diagnostic
criteria, progressive/measurable disease, and previous exposure to ≥3 prior LOT. Patients
who had received prior anti-BCMA treatment were ineligible. Patients were given weekly
subcutaneous teclistamab at the recommended phase 2 dose (1.5 mg/kg with step-up doses
of 0.06 and 0.3 mg/kg). PRO assessments were performed at screening and at every even
treatment cycle; results for cycles 2-8 are reported here. HRQoL was assessed by EORTC
QLQ-C30 (range: 0–100; with higher scores indicating better global health status [GHS]
but greater symptom severity [symptom scales]) and EuroQol 5-dimensional descriptive
system (using the visual analog scale [VAS] with “0” indicating worst imaginable health
state and “100” indicating best imaginable health state). Mixed-effects model with
repeated measures was used to determine treatment effect. Meaningful improvement was
defined as the proportion of patients with a change of ≥10 points. Kaplan-Meier estimate
was used to determine time to worsening.
Results: The analysis included 110 patients, with median follow-up duration of 7.8
months. The PRO compliance rates were high at baseline (85–90%) and through treatment
cycles 2–8 (80–94%). Overall HRQoL was improved with teclistamab, as shown by improved
GHS scores (cycle 2–8) and reduced pain (-4.2 [cycle 2] to -15.1 [cycle 8]; Table),
with no overall changes in physical functioning and fatigue. Meaningful improvements
from baseline to cycle 8 were observed in GHS (50% of patients), physical function
(35%), pain (65%), and fatigue (73%). Meaningful improvement in overall health (VAS)
was also observed in 50% of patients. Median time to improvement from baseline was
~1.5 months with longer time to improvement seen for nausea/vomiting and fatigue.
Across all symptoms, median time to worsening ranged from 2 months to not estimable.
Image:
Summary/Conclusion: Teclistamab provided rapid, clinically meaningful improvements
in HRQoL in patients with RRMM, which are consistent with clinical outcomes in MajesTEC-1.
P923: TANDEM AUTOLOGOUS STEM CELL TRANSPLANTATIONS STILL BENEFIT FOR MYELOMA PATIENTS:
A POOLING META-ANALYSIS
C.-H. Lee1,*, C.-L. Ho1
1Hematology and Oncology, Tri-Service General Hospital/National Defense Medical Center,
Neihu Dist, Taiwan
Background: Although high-dose therapy and autologous stem cell transplant combined
with novel agents continues to be the hallmark of first-line treatment in newly diagnosed
transplant-eligible multiple myeloma patients, disease progression remains an issue.
To deepen the treatment response and prolong disease free survival, tandem autologous
stem cell transplant is one of treatment choice. Compared to toxicity of sub-sequent
pharmacotherapy or allogeneic transplant, tandem ASCT is well-tolerated. However,
the efficacy of tandem ASCT is not fully defined yet. Thus, we aimed to assess effectiveness
of tandem ASCT.
Aims: To evaluate the true effect of tandem autologous/autologous stem cell transplantation
for myeloma patients in current era.
Methods: Eligible randomized controlled trials published before Dec 2021 were retrieved
from databases. Myeloma patients received tandem autologous stem cell transplantation
was included. Systematic review and pairwise meta-analysis were performed. We calculated
the hazard ratio (HR) with 95% confidence intervals (CIs).
Results: Ten articles, involving 3,123 patients, met the selection criteria. All studies
demonstrated acceptable quality. The mean age was 58.2 year-old with mean 36.2% high
risk cytogenetics feature. Eight trials were international multi-centers phase III
RCTs. Tandem autologous stem cell transplantation compared to single ASCT was associated
with a significantly superior progression free survival (random-effect HR 0.89; 95%
CI 0.82-0.97; I2 = 0.0%), but no significance difference regard to overall survival
(random-effect HR 0.98; 95% CI 0.94–1.03; I2 = 0%). For high risk myeloma subgroup,
4 trials compared tandem ASCT versus lenalidomide, tandem ASCT demonstrated superior
progression free survival (random-effect HR 0.87; 95% CI 0.82–0.91; I2 = 0%)
Image:
Summary/Conclusion: In conclusion, tandem ASCT are still to be considered as an acceptable
treatment options in general myeloma patients. High-risk cytogenetics is frequently
observed in newly diagnosed myeloma with worsens outcome after single autologous,
whereas a tandem autologous transplant strategy may overcome onset poor prognosis.
P924: TREATMENT EFFICACY OF PROGRESSION FREE SURVIVAL FOR REFRACTORY/RELAPSED MULTIPLE
MYELOMA IN GERIATRIC PATIENTS: A NETWORK META-ANALYSIS
C.-H. Lee1,*, C.-L. Ho1
1Hematology and Oncology, Tri-Service General Hospital, Neihu Dist, Taiwan
Background: In recent 10 years, there were many studies explored the treatment efficacy
of refractory/relapse multiple myeloma (R/R MM). Emerging evidences proved combination
of new treatment modalities is significant better than traditional therapy. However,
very few studies focus on geriatric patient who are more fragile and may not be able
to tolerate treatment.
Aims: Our aim was to synthesize all efficacy evidence, enabling an integrated comparison
of all current treatment options in geriatric patients with R/R MM.
Methods: We performed a systematic literature review to identify all publicly available
randomized controlled trials (RCT) evidence. We searched Embase, PubMed, Cochrane
Central Register of Controlled Clinical Trials, and the Web site www.ClinicalTrials.gov.
Geriatric Patients with a diagnosis of R/R MM (including patients in second-line or
more than second-line treatments) received subsequent active treatment were included.
Geriatric patients should be set at least age 65 or above as a cut-off level. Data
was extracted from subgroup analysis of each study. The evidence was synthesized using
a Bayesian network meta-analysis. Hazard ratio (HR) was adopted.
Results: In total, 4,966 citations were retrieved from the databases; 60 full texts
were screened, of which 47 were excluded. In total, 13 RCTs were identified for quantitive
analysis, including 14 treatment options (figure). Total 4337 geriatric patient were
enrolled for network meta-analysis, twelve trials set cut-off level of age about 65,
one set age 75 as a cut-off level. All trials had good quality. The triple combination
therapy of daratumumab, lenalidomide and dexamethasone (DaraLenDex) was identified
as the best treatment option in patients with R/R MM (figure). It was most favorable
in terms of (1) HR for progression free survival (0.15; 95% credible interval (CI):
0.09 to 0.25) with significance and (2) probability of being best (96% of the cumulative
ranking).
Image:
Summary/Conclusion: To the best of our knowledge, this is the first NMA on R/R MM
that includes all regimens currently evaluated in randomized trials in elderly R/R
MM. Our findings suggest that a 3-drug regimen containing the lenalidomide-dexamethasone
backbone, preferentially combined with anti-MM mAbs daratumumab or elotuzumab, has
the highest probability of being ranked as the best treatment in this setting, underlying
the role of immunotherapy in MM. Prospective randomized trials are eagerly awaited
to clarify the optimal sequencing of treatments for MM. We emphasize that it remains
essential to conduct phase III RCTs to obtain more direct head-to-head evidence. Until
such evidence becomes available, our results are highly important for informed decision
making in everyday clinical practice
P925: IMPACT OF PRIOR TREATMENT EXPOSURE ON THE EFFECTIVENESS OF IXAZOMIB-LENALIDOMIDE-DEXAMETHASONE
IN RELAPSED/REFRACTORY MULTIPLE MYELOMA PATIENTS TREATED IN ROUTINE CLINICAL PRACTICE
(THE INSURE STUDY)
H. C. Lee1,*, K. Ramasamy2, M. Macro3, F. E. Davies4, R. Abonour5, F. van Rhee6, V.
T. Hungria7, N. Puig8, K. Ren9, J. Silar10, V. Enwemadu11, D. Cherepanov9, D. M. Stull11,
X. Leleu12
1MD Anderson Cancer Center, Houston, United States of America; 2Department of Haematology,
Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom; 3CHU de
Caen, Caen, France; 4Perlmutter Cancer Center, NYU Langone, New York; 5Indiana University
School of Medicine, Indianapolis; 6University of Arkansas for Medical Sciences, Little
Rock, United States of America; 7Clinica São Germano and Santa Casa Medical School,
São Paulo, Brazil; 8Hospital Universitario de Salamanca Hematología, Instituto de
Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; 9Takeda Development
Center Americas, Inc. (TDCA), Lexington, United States of America; 10Institute of
Biostatistics & Analyses, Ltd, Brno, Czechia; 11Takeda Pharmaceuticals U.S.A., Inc.,
Lexington, United States of America; 12Pôle Régional de Cancérologie, Department of
Hematology, CHU La Milétrie-Poitiers, Poitiers, France
Background: Results from INSURE, a pooled, global analysis of 3 observational studies,
show that the effectiveness of ixazomib-lenalidomide-dexamethasone (IRd) used to treat
relapsed/refractory multiple myeloma (RRMM) in routine clinical practice is comparable
to its efficacy seen in the TOURMALINE-MM1 trial (median progression-free survival
[PFS], 19.9 vs 20.6 months [mos]), with no new safety concerns (Leleu ASH 2021 #2701).
Data on effectiveness outcomes following retreatment with agents used in earlier lines
of therapy (LoTs) are limited, but may be of particular value for MM pts previously
treated with lenalidomide (LEN) or proteosome inhibitors (PIs).
Aims: To characterize the impact of prior exposure & refractoriness to LEN or PIs
on the effectiveness & safety of IRd in RRMM.
Methods: INSURE is a pooled analysis of data from 3 studies: INSIGHT MM, UVEA-IXA,
& REMIX. INSIGHT MM is a prospective, global study of 4307 MM patients (pts) from
15 countries. UVEA-IXA is a multicenter, longitudinal, retrospective cohort study
of 309 RRMM pts receiving ixazomib (IXA)-based therapy via an early-access program
in Europe. REMIX is a retrospective/prospective study of 198 RRMM pts treated with
IRd via a compassionate-use program in France. INSURE included adult RRMM pts who
had received IRd in ≥2nd LoT. Primary outcomes were PFS & time-to-next therapy (TTNT).
Secondary outcomes included: duration of treatment (DOT), overall survival (OS), overall
response rate (ORR), & safety (discontinuations due to adverse events [AEs] were reported
separately for each study). In this prespecified analysis pts were grouped by prior
LEN or PI exposure (naïve, exposed, or refractory).
Results: 562 pts were included: 391/100/71 were LEN-naïve/exposed/refractory & 37/408/117
were PI-naïve/exposed/refractory. In LEN-naïve/exposed/refractory pts, median age
was 69/68/68 years (yrs; 24/14/18% >75 yrs) & 18/13/22% had an Eastern Cooperative
Oncology Group performance status (ECOG PS) ≥2 (missing pts excluded from %); pts
had received a median of 1/2/3 LoT(s) prior to IRd. In PI-naïve/exposed/refractory
pts, median age was 71/68/69 yrs (38/22/15% >75 yrs) & 6/16/27% had an ECOG PS ≥2
(missing pts excluded from %); pts in all 3 subgroups had received a median of 2 LoTs
prior to IRd. The Table shows effectiveness outcomes. Median DOT was 15.3/15.6/4.7
mos & median PFS was 21.6/25.8/5.8 mos in LEN-naïve/exposed/refractory pts. Median
DOT & PFS in PI-naïve/exposed/refractory pts were 20.4/15.2/7.6 mos & not reached/19.7/12.9
mos, respectively. OS data were immature. The proportions of LEN-naïve/exposed/refractory
pts in INSIGHT (n=114/39/28) & UVEA-IXA (n=161/15/19) who discontinued a study drug
due to AEs were: IXA, 32/28/25% & 19/7/11%; LEN, 22/28/18% & 16/7/11%; dexamethasone
(DEX), 18/21/14% & 11/0/11%, respectively. The proportions of PI-naïve/exposed/refractory
pts in INSIGHT (n=9/130/42) & UVEA-IXA (n=18/125/52) who discontinued a study drug
due to AEs were: IXA, 44/28/31% & 22/17/15%; LEN, 33/22/21% & 17/16/12%; DEX, 33/18/17%
& 17/10/8%, respectively. Data for discontinuations due to AEs were not available
for REMIX. Additional safety data will be presented.
Image:
Summary/Conclusion: IRd appeared to be effective in RRMM pts in routine clinical practice
regardless of prior LEN or PI exposure, with better outcomes seen in LEN- &/or PI-non-refractory
vs -refractory pts. PFS outcomes in the naïve/exposed populations are comparable to
those reported in TOURMALINE-MM1. Pts with prior LEN or PI exposure can achieve clinical
benefit when retreated suggesting that prior exposure should not preclude use of these
agents in later LoTs.
P926: DARATUMUMAB, BORTEZOMIB AND DEXAMETHASONE FOR TREATMENT OF PATIENTS WITH RELAPSED
OR REFRACTORY MULTIPLE MYELOMA AND SEVERE RENAL IMPAIRMENT: RESULTS FROM THE PHASE
2 GMMG-DANTE TRIAL
L. Leypoldt1,*, M. Gavriatopoulou2, B. Besemer3, H. Salwender4, M.-S. Raab5, A. Nogai6,
C. Khandanpour7, V. Runde8, M. Zago9, P. Martus10, H. Goldschmidt11, C. Bokemeyer1,
M. Dimopoulos2, K. Weisel1
1Department of Hematology, Oncology and Bone Marrow Transplantation with Section of
Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; 2Department
of Clinical Therapeutics, School of Medicine, National and Kapodistrian University
of Athens, Alexandra General Hospital, Athens, Greece; 3Department of Hematology,
Oncology, Immunology, Rheumatology and Pulmonology, University Hospital of Tuebingen,
Tuebingen; 4Asklepios Tumorzentrum Hamburg, AK Altona and AK St. Georg, Hamburg; 5Department
of Internal Medicine, Heidelberg University Hospital, Heidelberg; 6Department of Hematology,
Oncology and Tumor Immunology, Charite Medical School, Berlin; 7Department of Medicine
A, Hematology, Oncology and Pneumology, University Hospital Münster, Münster; 8Department
of Hematology/Oncology, Wilhelm-Anton Hospital Goch, Goch; 9Center for Clinical Trials,
University Hospital of Tuebingen; 10Department of Clinical Epidemiology and Applied
Biostatistics, Eberhard Karls University of Tuebingen, Tuebingen; 11Internal Medicine
V and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg,
Germany
Background: Renal function impairment is one of the main characteristics of multiple
myeloma (MM) leading to diagnosis and defining treatment requirement in first-line
and subsequent relapse. However, limited data exist on safety and efficacy of anti-MM
regimens in patients (pts) with severe renal impairment and those requiring hemodialysis
as pts are mostly excluded from clinical trials and specific trials in this indication
are rare. Proteasome inhibitors as bortezomib (BTZ) are known to represent one of
the preferable drug classes in patients with renal impairment due to rapid response,
lack of dose adjustment and renoprotective effects. Daratumumab (DARA) is an anti-CD38
monoclonal antibody which enhances response rates and efficacy when added to standard
of care regimens, both in newly diagnosed and relapsed and refractory (RR)MM. The
combination of BTZ, dexamethasone (DEX), and DARA (DVd) has been shown to be efficacious
and safe in RRMM in the phase 3 study CASTOR (Palumbo et al., NEJM 2016). In the CASTOR
trial, pts with a GFR < 20 ml/min were excluded.
Aims: Here, we present results from the primary analysis of the investigator-initiated
multicenter GMMG-DANTE (NCT02977494) trial investigating DVd in RRMM pts with severe
renal impairment including pts on hemodialysis.
Methods: RRMM pts with measurable disease who had received at least 1 prior line of
therapy and a GFR < 30 ml/min or undergoing hemodialysis were eligible. Pts received
DVd in the approved schedule with 8 cycles (21 d/cycle) of BTZ (1.3 mg/m2, SC) on
Days 1, 4, 8, and 11 and DEX (20 mg, PO or IV) on days 1, 2, 4, 5, 8, 9, 11, and 12
+ DARA (16 mg/kg, IV) given weekly for cycles 1-3, Q3W for cycles 4-8, and Q4W thereafter.
36 patients were planned to be included into the trial. The trial was closed prematurely
after 22 patients due to inferior recruitment. The primary endpoint was overall response
rate (ORR). Key secondary endpoints were progression-free survival (PFS), overall
survival (OS) and safety. Response assessment was performed using the IMWG criteria.
Results: 22 pts from 6 German and one Greek site were included from 2017-2020. Analyzed
population included 21 patients. Median age was 70 years. Median GFR was 21.0 ml/min,
8 patients were under hemodialysis. Median number of prior lines was 2 (range 1-10).
All patients started DVd treatment. For one patient it was not possible to obtain
a response. ORR was 67% (14/20) with 6 pts (29%) showing a partial response (PR),
6 pts (29%) a very good partial response (VGPR) and 2 pts (10%) a complete response
(CR). Median treatment duration was 24 weeks (range 2-106). After a median follow-up
of 28 months, median PFS was 10.4 months, median OS was not reached (OS after 24 months
until 39 months plateau 0.504). The most frequent toxicity ≥ grade 3 was hematologic,
with anemia in 23.8%, thrombocytopenia in 23.8% and neutropenia in 9.5%. Main non-hematologic
adverse events (AEs) ≥ grade 3 were infections (23.8%) with mainly pneumonias. Polyneuropathy
all grade was described in 52.4%.
Summary/Conclusion: In this phase II prospective trial we demonstrated that DVd shows
relevant efficacy in RRMM pts with severe renal impairment and can be safely administered.
Efficacy is comparable to the one reported in patients with no severe renal impairment.
Toxicity does not differ from previously reported data on DVd. Acknowledging the generally
reported impaired outcome of MM pts showing severe renal impairment, PFS and OS underline
the importance of effective MM regimens which can be safely administered in a difficult
to treat population.
P927: CLINICAL CHARACTERISTICS, SURVIVAL OUTCOMES AND PROGNOSIS IN 123 IMMUNOGLOBULIN
D MULTIPLE MYELOMA PATIENTS: A RETROSPECTIVE SINGLE-CENTER
J. Liu1,*, H. Fan1, W. Yan1, J. Xu1, L. Li1, L. Qiu1, G. An1
1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences
& Peking Union Medical College, Tianjin, China
Background: Immunoglobulin D (IgD) myeloma is a rare subtype of multiple myeloma (MM),
and has been considered to have a more aggressive course and poor outcome. With the
rapid progress of multiple myeloma treatment, including the application of proteasome
inhibitors and immunomodulatory drugs and the promotion of autologous stem cell transplantation
(ASCT), the prognosis of MM patients has been greatly improved. Considering the development
of MM treatment, the survival outcome and prognostic factors of IgD MM patients are
now controversial.
Aims: To evaluate the clinical characteristics, survival outcomes and prognosis of
IgD myeloma
patients in the era of novel therapy.
Methods: We retrospectively analyzed the clinical characteristics of 123 IgD MM patients
and compared them with other MM subtypes. At the same time, we also discussed the
survival outcomes and prognostic factors of IgD myeloma patients under different treatment
modes.
Results: Male to female ratio was 2.2, median age was 55 years (29-79 years), with
preponderance of lambda light chains (89.4%). IgD myeloma more likely presented advanced
stage when diagnosed: higher frequencies of ISS stage Ⅲ, R-ISS stage Ⅲ, renal dysfunction,
high lactate dehydrogenase (LDH) level, high β2MG level and high bone marrow plasmacytosis.
Additionally, the genetics of IgD myeloma patients were also different from other
subtypes myeloma. Complex karyotypes were more common in IgD myeloma patients (23.9%
vs. 14.8%, p=0.017). Cytogenetic abnormalities demonstrated by FISH could be found
in 86% (80/93) of the IgD myeloma patients. The frequencies of t (11;14) (38.6% vs.
13.6%, p=0.000) and 1q21 amplification (72.0% vs. 53.9%, p=0.001) were much higher
in IgD MM patients than other subtypes myeloma, while deletion of 13q (34.4% vs. 45.7%,
p=0.032) and high risk cytogenetic abnormalities (HRCA) (23.0% vs. 37.9%, p=0.009)
were less. 71 IgD MM patients received induction therapy based on ‘novel therapies’,
26 on ‘traditional therapies’. Compared with ‘traditional therapies’, ‘novel therapies’
significantly improved the prognosis of IgD patients (PFS 16.9 vs. 29.1 months, P=0.008;
OS 37.6 vs. 70.8 months, P=0.002). Further analysis of the ‘novel therapies’ group
found that, in non-transplant patients, the prognosis of the IgD and non-IgD patients
was similar, but in transplant patients, IgD MM patients showed worse outcomes compared
to non-IgD MM patients (figure). Multivariate analysis of 71 IgD patients who received
‘novel therapies’ identified that R-ISS stage Ⅲ, complex karyotypes and deletion of
13q were independent adverse factors for OS, whereas complex karyotypes and thrombocytopenia
were independent adverse factors for PFS.
Image:
Summary/Conclusion: In summary, our study compared the clinical characteristics of
IgD and non-IgD MM patients, IgD myeloma patients are younger, have higher tumor burden
and advanced disease stage. In addition, their genetics are significantly different
from non-IgD MM: IgD MM patients have more complex karyotypes, amplification of 1q21
and t (11;14), while less deletion of 13q and HRCA. Meanwhile, we found that although
new drugs combined with ASCT significantly improved the prognosis of MM patients,
the improvement of IgD MM is less obvious than that of non-IgD MM patients, suggesting
more targeted drugs need to be developed for patients with IgD MM. Finally, we identified
that R-ISS stage Ⅲ, complex karyotypes and deletion of 13q were independent adverse
factors for OS, indicating that R-ISS is still useful way to predict the outcomes
of patients with IgD myeloma.
P928: REAL-LIFE ANALYSIS OF THE MULTIPLE MYELOMA PATIENT’S SURVIVAL IN A LARGE COHORT
OF PATIENTS
N. Lopez-Muñoz1,*, G. Hernandez-Ibarburu2, J. M. Sánchez-Pina1, R. A. Alonso1, C.
Cuellar1, M. Calbacho1, R. Ayala1, R. Iñiguez1, N. García Barrio3, E. Vera1, D. Pérez-Rey2,
L. Meloni4, M. Pedrera-Jiménez3, P. Serrano-Balazote3, J. De La Cruz5, J. Martínez-López1
1Hospital 12 de Octubre; 2Biomedical Informatics Group, Universidad Politécnica de
Madrid; 3Data Science Group, Research Institute imas12, MADRID, Spain; 4Trinetx, LLC,
Cambridge, United States of America; 5Research Institute imas12, MADRID, Spain
Background: Multiple myeloma (MM) constitutes approximately 10% of hematological malignancies,
with a median age at diagnosis of 65 years. Patient survival has improved considerably
over the last 20 years with the introduction of new drugs. In 1999, the first immunomodulatory
drug, thalidomide, was approved, followed by lenalidomide in 2005. In 2003, proteosome
inhibitors such as bortezomib were introduced and, in 2015, anti-CD38 monoclonal antibodies
such as daratumumab.
Aims: By relying on the TriNetx platform, a global health research platform, we have
analyzed the impact on the outcome of the introduction new drugs for MM in the last
20 years. We also analyzed the difference patterns of treatments between networks.
Methods: First, we retrospectively selected patients diagnosed with symptomatic multiple
myeloma between 1999 and 2019 in a tertiary care hospital in Spain, Hospital 12 de
Octubre (H12O) in Madrid. Then, we analyzed data from more than 7500 patients diagnosed
with CML from research networks: EMEA and the US; covering more than 16 million subjects.
We compared the time of survival since the MM diagnosis in three groups depending
of the age at MM diagnosis (less than 65, between 65 and 75, and more than 75 years
old) over three periods of time: 1999-2009, 2010-2014 and 2015-2019 (2020 was excluded
because of pandemic impact). Kaplan-Meier analysis was used for analyzing overall
survival (OS) and differences between groups were tested for statistical significance
using the log-rank test. Also, we analyzed the patterns of treatment along this time.
Results: Approximately, a total 760 patients were included in the study. In the H12O,
median OS was 45.1, 45.2, 65.1 months and not reached for the 1999-2009, 2010-2014
and 2015-2019 periods, respectively (p=0.001). Among the group of patients younger
than 65 years old, the median OS was 64 months (47.5-81.4; 95%) among patients diagnosed
in the period 1999-2009; 80 months (36.1-124; 95%) in 2010-2014; and median not reached
for the 2014-2019 (p<.001).
Similar results were found in a US cohort, including approximately 7000 patients,
between the years 2010-2014 vs 2014-2019 (59.71% vs 64.20%, p<0.0001, HR 1.154 (1.074-
1.241) (figure 1).
The same results were observed for the 65-75 years cohort, median OS was 62.6 months
(50.9-74.3; 95%) for the 1999-2009 period, 70.6 months (33.1-108.1; 95%) between 2010
and 2014, and not reached for the 2014-2019 period (p<.001).
The same occurs among more than 3000 patients from the US base between the years 2010-2014
vs 2014-2019 (53.96% vs 57.73%, p=0.016, HR 1.099 (1.018 - 1.187) (figure 2).
Otherwise, patients older than 75 years have a median OS of 31.1 months and do not
show a statistically significant difference regardless on the year of MM diagnosis
(p=.18). Nor were clinically significant results obtained between the EMEA and US
cohorts.
On the other hand, the first-line treatment used in patients was analysed, and in
both the EMEA and US cohorts, the use of bortezomib (46% vs 47% in the EMEA cohort;
62% vs 70% in US cohort), lenalidomide (43% vs 57% in EMEA cohort, 15% vs 18% in US
cohort) and daratumumab (0% vs 4% in EMEA cohort, 2% vs 4% in US cohort) increases
over the years comparing the 2010-2014 vs 2015-2019 time cohorts. This could explain
the improved survival in more recent times.
Summary/Conclusion: The introduction of new agents for the treatment of MM has transformed
the natural history of the disease, achieving long survival times in younger patients.
Thus, it is essential to continue to advance and develop new therapies.
P929: IXAZOMIB AND DARATUMUMAB WITHOUT DEXAMETHASONE (I-DARA) IN ELDERLY FRAIL RELAPSING
MYELOMA (RRMM) PATIENTS: RESULTS OF THE PHASE 2 STUDY IFM 2018-02 OF THE INTERGROUPE
FRANCOPHONE DU MYELOME (IFM).
M. Macro1,*, C. Touzeau2, C. Mariette3, S. Manier4, S. Brechignac5, L. Vincent6, B.
Hebraud7, O. Decaux8, S. Schulmann9, C. Lenoir10, P. Godmer11, A. Farge12, L. Peyro
Saint Paul13, J.-J. Parienti13, X. Leleu14
1HEMATOLOGY IHBN, University Hospital, CAEN; 2HEMATOLOGY, University Hospital, Nantes;
3HEMATOLOGY, University Hospital, Grenoble; 4Hematology, University Hospital, Lille;
5Hematology, University Hospital, Paris; 6Hematology, University Hospital, Montpellier;
7Hematology, University Hospital, Toulouse; 8Hematology, University Hospital, Rennes;
9Hematology, University Hospital, Nancy; 10Hematology, Private Hospital, Bordeaux;
11Hematology, Community Hospital, Vannes; 12Hematology; 13Clinical Research, University
Hospital, CAEN; 14Hematology, University Hospital, Poitiers, France
Background: Frail patients with multiple myeloma have an inferior outcome, especially
in the relapse setting. This adverse prognosis is mainly related to a high discontinuation
rate due to treatment (Tx) related adverse events.
Aims: The aim of this phase 2 study is to evaluate efficacy and tolerability of Ixazomib-Daratumumab
(I-Dara) without Dexamethasone in elderly frail patients with relapsed myeloma (RRMM)
(NCT03757221).
Methods: Ixa-Dara naïve RRMM patients received oral Ixazomib (4 mg: days 1, 8, 15),
IV Daratumumab (16 mg/kg; days 1, 8, 15, 22, cycles 1-2; days 1, 15, cycles 3-6; days
1, cycles 7+) and IV Methylprednisolone before Daratumumab (100 mg at day 1, 8, cycle
1 and then 60 mg). They were enrolled after 1 or 2 prior therapy if their frailty
score was ≥ 2 by IMWG score. The primary endpoint was ≥ very good partial response
rate (VGPR) at one year. Secondary endpoints included overall response rate (ORR),
progression free survival (PFS), overall survival (OS) & toxicity according to NCI-CTCAE
version 5
Results: Sixty-three patients were screened and 55 enrolled between 03/2018 and 09/2021.
Patient were at first (n = 36) or second relapse (n = 19). Thirty-three patients (60%)
were previously exposed to bortezomib, 37 (67%) were previously exposed to lenalidomide
(Len) and 20 (36 %) were refractory to Len. Median age was 82 (72-93). All patients
had a frailty score ≥2 and 13 (24 %) had a 3 or 4 frailty score. In 41 patients ISS
at diagnosis was stage I (n = 11), II (n = 18) or III (n = 12). Seventeen (36%) patients
harbored high-risk (HR) cytogenetic, including t(4;14) (n = 8) or del17p (n = 10).
The median duration of Tx among 28 pts with ongoing Tx was 10 months [5-32] at data
cutoff (February, 2). The median duration of Tx among 27 pts who stopped Tx was 6
months [0-18]: 18 had progressive disease. Nine patients died during the study: Daratumumab-related
bronchospasm (D1C1); Ixazomib-related overdose (C2); sepsis (n = 4), progressive disease
(n = 3). Regarding toxicity, 27 pts had a ≥grade 3 AE (49%). The most common grade
3-4 toxicities were thrombocytopenia (n = 9), other cytopenias (n = 4), infection
(n = 8), hypertension (n = 3) and gastrointestinal disorders (n = 3). Fourteen out
of 28 were SAE including 5 infections, 1 bronchospasm, 1 acute respiratory failure
and 2 ixazomib overdoses. Overall response rate, including minimal response, was 86
% with a ≥VGPR rate of 32 % in the whole group. In Len refractory patients the ORR
was 82 % and ≥VGPR 41%, in HR cytogenetic patients ORR was 85 % and ≥VGPR 46%. With
a median follow-up of 11.6 months median PFS is 16 months and median OS NR (76% estimated
at one year).
Image:
Summary/Conclusion: In this elderly frail population Ixa-Dara is a feasible combination
with favorable efficacy profile even in Len refractory and HR cytogenetic patients.
Early toxicity remains a concern in this population eventhough more manageable with
Dara SC.
P930: ISATUXIMAB, LENALIDOMIDE, BORTEZOMIB AND DEXAMETHASONE AS INDUCTION THERAPY
FOR NEWLY-DIAGNOSED MULTIPLE MYELOMA PATIENTS WITH HIGH-RISK CYTOGENETICS: A SUBGROUP
ANALYSIS FROM THE GMMG-HD7 TRIAL
E. K. Mai1,*, U. Bertsch1,2, R. Fenk3, D. Tichy4, B. Besemer5, J. Dürig6, R. Schroers7,
I. von Metzler8, M. Hänel9, C. Mann10, A. M. Asemissen11, B. Heilmeier12, E. Nievergall1,
S. Huhn1, K. Kriegsmann1, N. Weinhold1, S. Luntz13, T. A. W. Holderrried14, K. Trautmann-Grill15,
D. Gezer16, M. Klaiber-Hakimi17, M. Müller18, C. Khandanpour19, W. Knauf20, C. Scheid21,
M. Munder22, T. Geer23, H. Riesenberg24, J. Thomalla25, M. Hoffmann26, M. S. Raab1,
H. J. Salwender27, K. C. Weisel11, H. Goldschmidt1,2
1Department of Internal Medicine V, University Hospital Heidelberg; 2National Center
for Tumor Diseases, Heidelberg; 3Department of Hematology, Oncology and Clinical Immunology,
University Hospital Düsseldorf, Düsseldorf; 4Division of Biostatistics, erman Cancer
Research Center (DKFZ) Heidelberg, Heidelberg; 5Department of Internal Medicine II,
University Hospital Tübingen, Tübingen; 6Department for Hematology and Stem Cell Transplantation,
University Hospital Essen, Essen; 7Medical Clinic, University Hospital Bochum, Bochum;
8Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe
University, Frankfurt am Main; 9Department of Internal Medicine III, Clinic Chemnitz,
Chemnitz; 10Department for Hematology, Oncology and Immunology, University Hospital
Gießen and Marburg, Marburg; 11Department of Oncology, Hematology and BMT, University
Medical Center Hamburg-Eppendorf, Hamburg; 12Clinic for Oncology and Hematology, Hospital
Barmherzige Brueder Regensburg, Regensburg; 13Coordination Centre for Clinical Trails
(KKS) Heidelberg, Heidelberg; 14Department of Oncology, Hematology, Immuno-Oncology
and Rheumatology, University Hospital Bonn, Bonn; 15Department of Internal Medicine
I, University Hospital Dresden, Dresden; 16Department of Hematology, Oncology, Hemostaseology,
and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen;
17Clinic for Hematology, Oncology and Palliative Care, Marien Hospital Düsseldorf,
Düsseldorf; 18Clinic for Hematology, Oncology and Immunology, Klinikum Siloah Hannover,
Hannover; 19Medical Clinic A, niversity Hospital Münster, Münster; 20Center for Hematology
and Oncology Bethanien, Frankfurt am Main; 21Department of Internal Medicine I, University
Hospital Cologne, Cologne; 22Department of Internal Medicine III, University Hospital
Mainz, Mainz; 23Department of Internal Medicine III, Diakoneo Clinic Schwäbisch-Hall,
Schwäbisch-Hall; 24Hematology / Oncology Center, Bielefeld; 25Hematology / Oncology
Center, Koblenz; 26Medical Clinic A, Clinic Ludwigshafen, Ludwigshafen; 27Asklepios
Tumorzentrum Hamburg, AK Altona and AK St. Georg, Hamburg, Germany
Background: The multicenter phase III trial GMMG-HD7 (NCT03617731) demonstrated superior
minimal residual disease (MRD) negativity rate after induction therapy in patients
with transplant-eligible newly-diagnosed multiple myeloma (NDMM) by addition of the
anti-CD38 monoclonal antibody isatuximab (Isa) to lenalidomide / bortezomib / dexamethasone
(Isa-RVd), as compared to RVd alone (Goldschmidt H et al., 2021, ASH Annual Meeting).
Aims: Here we present a subgroup analysis on patients with high-risk cytogenetics.
Methods: Patients with transplant-eligible NDMM were equally randomized to receive
three 42-day cycles of RVd (lenalidomide 25 mg/d p.o., d1–14 and d22-35; bortezomib
1.3 mg/m2 s.c. d1, 4, 8, 11, 22, 25, 29, 32; dexamethasone 20 mg/d d1-2, 4-5, 8-9,
11-12, 15, 22-23, 25-26, 29-30, 32-33) in both study arms. Isa was added to Isa-RVd
as follows: 10 mg/kg i.v., cycle 1: d 1, 8, 15, 22, 29; cycles 2-3: d 1, 15, 29. Randomization
for induction was stratified by Revised International Staging System. Primary endpoint
of the trial was MRD negativity rate assessed by next-generation flow (NGF, cut-off
1x10-5) after induction therapy. Fluorescence in-situ hybridization (FISH) analysis
was performed centrally on CD138-purified plasma cells. High-risk and ultra high-risk
cytogenetics were defined as at least one or two of the following aberrations, respectively:
del17p, t(4;14), t(14;16), gain1q21 (≥ 3 copies). Data cut-off for the present analysis
was December 2021.
Results: 660 patients (Isa-RVd: 331 and RVd: 329) were eligible for intention-to-treat
analysis. The study met its primary endpoint, demonstrating superiority of NGF-MRD
negativity rates with Isa-RVd compared to RVd (50.1% vs. 35.6%; odds ratio [OR]=1.82,
95% confidence interval [95% CI]: 1.33-2.48, p<0.001). High-risk cytogenetics were
well balanced between the treatment arms. 264 of 584 (45.2%) and 82 of 580 (14.1%)
evaluable patients had high-risk and ultra high-risk cytogenetics, respectively. Del17p,
t(4;14), t(14;16) and gain 1q21 were present in 59 of 615 (9.6%), 67 of 613 (10.9%),
17 of 609 (2.8%) and 218 of 583 (37.4%) evaluable patients, respectively. Among patients
with high-risk cytogenetics, MRD negativity rates were 50.4% (63/125) vs. 37.4% (52/139;
OR=1.70, 95% CI: 1.04-2.79, p=0.03) with Isa-RVd vs. RVd. MRD negativity rates for
ultra high-risk patients were 56.3% (27/48) vs. 44.1% (15/34; OR=1.63, 95% CI: 0.67-3.99,
p=0.28) with Isa-RVd vs. RVd. Similar results were observed for Isa-RVd vs. RVd among
the common major single high-risk cytogenetic features: del17p: 56.0% (14/25) vs.
35.3% (12/34), OR=2.33, 95% CI: 0.82-6.88; t(4;14): 57.6% (19/33) vs. 47.1% (16/34),
OR=1.53, 95% CI: 0.58-4.06; t(14;16): 66.7% (6/9) vs. 50.0% (4/8), OR=2.00, 95% CI:
0.28-15.67; gain1q21: 48.2% (55/114) vs. 35.6% (37/104), OR=1.69, 95% CI: 0.98-2.92.
Dividing evaluable patients in either standard risk (absence of any high-risk aberration;
320/578, 55.4%) vs. high-risk (exactly one high-risk aberration; 176/578, 30.4%) vs.
ultra high-risk (≥2 high-risk aberrations; 82/578, 14.2%) yielded similar efficacy
results. MRD negativity rates for Isa-RVd vs. RVd were 49.7% (86/173) vs. 36.7% (54/147;
OR=1.70, 95% CI: 1.09-2.68) in standard risk patients, 46.7% (35/75) vs. 34.7% (35/101;
OR=1.65, 95% CI: 0.90-3.05) in high-risk patients and 56.3% (27/48) vs. 44.1% (15/34;
OR=1.63, 95% CI: 0.67-3.99) in ultra high-risk patients.
Summary/Conclusion: Isa-RVd induction therapy is superior to RVd in patients with
transplant-eligible NDMM and high-risk or ultra high-risk cytogenetics, consistent
with the benefit observed in the overall trial population.
P931: PROSPECTIVE OBSERVATIONAL MULTICENTER STUDY OF GAUCHER DISEASE PREVALENCE IN
PATIENTS AFFECTED BY MONOCLONAL GAMMOPATHY OF UNDETERMINED SIGNIFICANCE
G. Giuffrida1,*, U. Markovic1,2,3, C. Conticello1, D. Nicolosi1, V. Calafiore1, A.
Romano1,4, A. Condorelli1, S. Grasso1, A. Duminuco1, M. Calagna1, A. Nardo1, C. Riccobene1,
B. Esposito1, U. Consoli5, V. Di Giacomo6, S. Neri6, M. R. Cingari7, G. Iaria8, V.
Innao5,9, M. Spina10, J. Gentile11, C. Zizzo12, G. Duro12, F. Di Raimondo1,4
1Division of Hematology, AOU “Policlinico G. Rodolico-San Marco”, Catania; 2Division
of Oncohematology and BMT, Istituto Oncologico del Mediterraneo, Viagrande; 3Department
of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of
Messina, Messina; 4Postgraduate School of Hematology, University of Catania; 5Unità
Operativa Complessa (UOC) di Ematologia, Azienda Ospedaliera di Rilievo Nazionale
e di Alta Specializzazione (ARNAS), Catania; 6UOC di Ematologia, Azienda Ospedaliera
Papardo, Messina; 7Unitá Operativa Semplice Dipartimentale Ematologia, Ospedale San
Vincenzo, Taormina; 8Hemato-Oncology and Radiotherapy Department, Azienda Ospedaliera
“Bianchi Melacrino Morelli”, Reggio Calabria; 9Division of Hematology, Department
of Human Pathology in Adulthood and Childhood, University of Messina, Messina; 10UO
Medicina Generale, Ospedale Nuovo di Gragnano, Gragnano; 11Sanofi Genzyme, Milano;
12Institute for Biomedical Research and Innovation (IRIB-CNR), National Research Council
of Italy, Palermo, Italy
Background: Gaucher disease (GD) is a rare, autosomal recessive genetic disorder,
caused by deficiency of glucocerebrosidase, leading to glucosylceramide accumulation
in tissue macrophages of hematological, visceral, and skeletal organ systems. Type
1 GD accounts for more than 90% all patients.
Monoclonal gammopathy of undetermined significance is the most common plasma cell
disorder, occurring in 3% of the population older than 50 years and is typically detected
as an incidental finding. Type 1 GD It is frequently associated with polyclonal and
monoclonal gammopathy (MGUS) and is hypothetically caused by long-term immune activation
due to accumulated lysolipids, particularly glucosylsphingosine, although the cause
remains unknown.
Aims: We evaluated the prevalence of GD in MGUS patients, particularly when other
signs and symptoms are present, in order to determine its potential usefulness as
screening tool and inclusion in diagnostic framework of GD.
Methods: Between January 2018 and February 2022, dried blood spots (DBS) samples were
collected and tested for the acid β-glucosidase (glucocerebrosidase) enzyme activity
from MGUS patients, both adult and pediatric, followed in seven hematology units of
Sicily, Calabria and Naples. The glucocerebrosidase activity was measured with multiplexed
tandem mass spectrometry (MS/MS) using the NeoLSD® assay system, and pathological
range was within 0.2 and 2.5 nmol/h/ml. In case of DBS positive result, a confirmatory
test was carried over in order to confirm the diagnosis of GD. The study was approved
by the local institutional review board and supported by Sanofi Genzyme. All patients
provided informed consent for the prospective collection of their data.
Results: A total of 600 patients with MGUS was enrolled at last study update. Around
half of the study population was male, with median age of 65 years. Immunoglobulin
G was most frequently isolated with serum immunofixation, median level of monoclonal
protein was 0.66 g/dL, while median kappa and lambda values based on monoclonal serum
free light chain type were 24.8 mg/L and 32.7 mg/L, respectively. Out of 161 patients
with available complete blood count median hemoglobin value was 13.5 g/dL with 15
patients having less than 11 g/dL, median platelet count was 224.000/mmc with 10 patients
having less than 100.000/mmc platelets and median neutrophil count of 4.280/mmc and
seven patients with less than 2.000/mmc neutrophils. Median ferritin level was 77 ng/mL,
with more than 400 ng/mL in 10 patients.
The median glucocerebrosidase activity in the entire cohort was 6.9 nmol/h/ml (range
0.3-58.8), with thirteen patients having pathological enzyme activity (median value
1.8 nmol/h/ml, range 0.2-2.5). Sequence analysis of GBA gene evidenced compound heterozygous
mutation of the GBA1 gene in three patients (c.1226A>G - N370S and c.1448T>C -L444P
in two and c.1226A>G -N370S and c.355G>A -G119R in one respectively), and homozygous
mutation (c.1226A>G -N370S) in one patient associated with glucocerebrosidase activity
below normal limits. All three patients with compound heterozygous mutational status
had signs of GD (hepatosplenomegaly and mild thrombocytopenia). As for the rest of
the MGUS patients, in a total of 114 patients sequence analysis evidenced single heterozygous
mutation in 16 patients.
Summary/Conclusion: Although type 1 GD remains a rare lysosomal storage disorder,
patients with the diagnosis of MGUS could be considered for GD screening with DBS,
especially when other symptoms (thrombocytopenia, splenomegaly, hyperferritinemia)
are present.
P932: ON-DEMAND PLERIXAFOR WITH CYCLOPHOSPHAMIDE AND G-CSF FOR HEMATOPOIETIC STEM-CELL
MOBILIZATION IN MULTIPLE MYELOMA PATIENTS: FINAL RESULTS OF THE MOZOBL06877 STUDY
R. Mina1,*, F. Bonello1, F. Fazio1, R. Saccardi1, V. Bongarzoni1, F. Marchesi1, G.
Bertuglia1, P. Curci1, R. M. Lemoli1, S. Ballanti1, T. Dentamaro1, G. Benevolo1, A.
Capra1, R. Floris1, P. Tosi1, A. Olivieri1, D. Rota-Scalabrini1, C. Cangialosi1, M.
Cavo1, P. Corradini1, G. Milone1, M. Boccadoro1, A. Larocca1
1European Myeloma Network, (EMN), Italy
Background: High-dose melphalan and autologous stem-cell transplant (ASCT) are pillars
for the upfront treatment of ASCT-eligible newly diagnosed multiple myeloma (ND)MM
patients (pts) or as salvage strategy at relapse. Failure to collect an adequate number
of hematopoietic stem cells (HSC) to receive ASCT (<2×106 CD34+ cells/kg) occurs in
5%-15% of MM pts undergoing HSC mobilization with granulocyte colony-stimulating factor
(G-CSF) or G-CSF+cyclophosphamide (G-CSF/CY).
Aims: We report the final results of the observational MOZOBL06877 study (NCT03406091;
partially supported by Sanofi investigation funds) to prospectively assess the performance
of HSC mobilization with G-CSF/CY plus on-demand CXCR4 inhibitor plerixafor (PLX)
in NDMM pts treated with novel agents.
Methods: NDMM pts undergoing HSC mobilization with CY (2-4 g/m2) and G-CSF (5-10 mcg/kg/day)
were enrolled and observed up to 30 days after mobilization. According to its label,
“on-demand” PLX was administered in pts with <20 CD34+ cells/ul after ≥4 days of G-CSF
or in case <1×106 CD34+ cells/kg were collected on the first apheresis day. Pts gave
written informed consent. The primary endpoint was the poor mobilizer rate, defined
as the rate of pts collecting <2×106 CD34+ cells/kg or requiring PLX. Secondary endpoints
were the identification of predictive factors for PLX use and safety during mobilization.
Results: 301 NDMM pts were enrolled and analyzed; 72% received induction with bortezomib-thalidomide-dexamethasone,
9% a lenalidomide (Len)-based and 3% a daratumumab (Dara)-based regimen. Overall,
48 pts (16%) were poor mobilizers: 14 (5%) failed to yield ≥2×106/Kg CD34+, while
34 (11%) required rescue with PLX. Among pts yielding <2×106/Kg CD34+ cells, 4 received
PLX, while 10 did not. Among pts who successfully collected ≥2×106/Kg CD34+ (n=287,
95%), 34 (12%) required PLX: 23 (68%) due to a CD34+/uL count ≤20 after ≥4 days of
G-CSF and 11 (32%) to a collection <1×106/Kg after the first apheresis day. In pts
mobilized with PLX, median number of CD34+×106/L cells increased from 17.5 (IQR 10.8-25.6)
before PLX to 58.3 (IQR 34.2-100.2) after PLX. Median number of CD34+/Kg collected
in pts who did not require PLX was 10.2×106 (IQR 8.3-13.2). In pts who required PLX,
a median of 6.5×106 CD34+/Kg (IQR 4.6-9.6) was collected. An adequate HSC yield for
≥2 ASCTs (≥4×106/Kg) was obtained in 96% and 85% of pts not receiving or receiving
PLX, respectively. Median number of apheresis days was 1 (IQR 1-2) in pts not requiring
PLX and 2 (IQR 1-2) in pts requiring PLX. In a multivariate analysis (Table), factors
predicting PLX use were advanced disease stage (R-ISS 3 vs 1-2, OR 5.5, P=0.01), bone
marrow (BM) plasma cell (PC) infiltration at diagnosis (PC>60% vs ≤60%, OR 4.2, P<0.001),
pre-mobilization absolute neutrophil count (ANC, < vs >2500/mmc, OR 2.8, P=0.01),
and the use of Len-based (Yes vs No, OR 3.4, P=0.02) or Dara-based induction regimens
(Yes vs No, OR 5.3; P=0.04). Grade 3 infections occurred in 3 pts (1%), with no grade
4-5 reported.
Image:
Summary/Conclusion: Among NDMM pts treated with novel-agent-based induction and undergoing
HSC mobilization with CY/GCSF, “on-demand” PLX in case of low CD34+ cell count or
insufficient HSC yield was a safe and effective rescue strategy, reducing mobilization
failure from 16% to 5%. Predictive factors for PLX use were advanced disease stage,
high BM plasmacytosis, low ANC values before mobilization and the use of Len/Dara
during induction. Preemptive PLX in pts with ≥1 risk factors should be investigated
to further reduce the risk of mobilization failure.
P933: DARATUMUMAB (D) IN COMBINATION WITH VD OR D-RD IN RELAPSED OR REFRACTORY MULTIPLE
MYELOMA: SUBGROUP ANALYSIS OF CASTOR AND POLLUX STUDIES IN PATIENTS WITH EARLY OR
LATE RELAPSE AFTER INITIAL THERAPY
A. Spencer1,*, P. Moreau2, M.-V. Mateos3, H. Goldschmidt4, K. Suzuki5, M.-D. Levin6,
P. Sonneveld7, S.-S. Yoon8, S. Z. Usmani9, K. Weisel10, D. Reece11, T. Ahmadi12, H.
Pei13, W. Garvin Mayo14, X. Gai15, J. Carey16, R. Carson16, M. A. Dimopoulos17
1Malignant Haematology and Stem Cell Transplantation Service, Alfred Health-Monash
University, Melbourne, Australia; 2Hematology Department, University Hospital Hôtel-Dieu,
Nantes, France; 3University Hospital of Salamanca/IBSAL/Cancer Research Center-IBMCC
(USAL-CSIC), Salamanca, Spain; 4University Hospital Heidelberg, Internal Medicine
V and National Center for Tumor Diseases (NCT), Heidelberg, Germany; 5Department of
Hematology, Japanese Red Cross Medical Center, Tokyo, Japan; 6Albert Schweitzer Hospital,
Dordrecht; 7Erasmus MC Cancer Institute, Rotterdam, Netherlands; 8Department of Internal
Medicine, Seoul National University College of Medicine, Seoul, South Korea; 9Memorial
Sloan Kettering Cancer Center, New York, NY, United States of America; 10Department
of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology,
University Medical Center Hamburg-Eppendorf, Hamburg, Germany; 11Department of Medical
Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada; 12Genmab
US, Inc., Plainsboro, NJ; 13Janssen Research & Development, LLC, Titusville, NJ; 14Janssen
Research & Development, LLC, Raritan, NJ, United States of America; 15Janssen Research
& Development, LLC, Beijing, China; 16Janssen Research & Development, LLC, Spring
House, PA, United States of America; 17National and Kapodistrian University of Athens,
Athens, Greece
Background: High-risk multiple myeloma (MM) is often defined based on cytogenetic
abnormalities (ie, t[4;14], t[14;16], and/or del17p); however, patients who relapse
early (12-18 months) after initial therapy are considered a functional high-risk group
that is also associated with poor prognosis. Daratumumab (DARA), a human IgGk monoclonal
antibody targeting CD38, is approved in combination with standard-of-care regimens
for MM. In the phase 3 CASTOR and POLLUX studies, DARA in combination with bortezomib
plus dexamethasone (D-Vd) and lenalidomide plus dexamethasone (D-Rd) significantly
improved progression-free survival (PFS), regardless of cytogenetic risk, and achieved
higher rates of complete response or better (≥CR) and minimal residual disease (MRD)–negativity
vs Vd or Rd alone in patients with RRMM.
Aims: In this post-hoc analyses of CASTOR and POLLUX we evaluated D-Vd vs Vd and D-Rd
vs Rd in patient subgroups with 1 prior line of therapy based on timing of relapse
(early or late) after initiation of the first line of therapy.
Methods: In CASTOR and POLLUX, patients with RRMM and ≥1 prior line of therapy were
randomized to D-Vd/Vd or D-Rd/Rd, respectively. The primary endpoint was PFS. In this
analysis, the early relapse subgroup included patients with 1 prior line of therapy
who relapsed <18 months after initiating their first line of therapy; patients with
1 prior line of therapy who relapsed ≥18 months after initiating their first line
of therapy were included in the late relapse subgroup.
Results: 49 and 186 patients from CASTOR and 99 and 196 patients from POLLUX were
included in the early relapse and late relapse subgroups, respectively. Median follow-up
was 72.6 months (CASTOR) and 79.7 months (POLLUX). PFS consistently favored the DARA-containing
regimens across subgroups (Table). In CASTOR, ≥CR rates were higher with D-Vd vs Vd
in the early relapse (21% vs 17%; P = 0.7360) and late relapse (51% vs 14%; P <0.0001)
subgroups. In POLLUX, ≥CR rates were higher with D-Rd vs Rd in the early relapse (53%
vs 12%; P <0.0001) and late relapse (62% vs 38%; P = 0.0012) subgroups. MRD-negativity
rates (10–5) were higher with D-Vd/D-Rd vs Vd/Rd regardless of relapse timing (CASTOR:
early, 13% vs 0%; P = 0.1476; late, 23% vs 3%; P <0.0001; POLLUX: early, 30% vs 4%;
P = 0.0006; late, 34% vs 14%; P = 0.0009).
Image:
Summary/Conclusion: These post hoc analyses of CASTOR and POLLUX showed PFS and depth
of response benefits of DARA-containing regimens in patients with 1 prior line of
therapy, regardless of relapse timing (early or late). Our results support the use
of D-Vd and D-Rd in RRMM, including in patients who are considered functional high
risk.
P934: DARATUMUMAB + LENALIDOMIDE, BORTEZOMIB, AND DEXAMETHASONE IN TRANSPLANT-ELIGIBLE
NEWLY DIAGNOSED MULTIPLE MYELOMA: A POST HOC ANALYSIS OF SUSTAINED MINIMAL RESIDUAL
DISEASE NEGATIVITY FROM GRIFFIN
C. Rodriguez1,*, J. L. Kaufman2, J. Laubach3, D. W. Sborov4, B. Reeves5, A. Chari6,
R. Silbermann7, L. J. Costa8, L. D. Anderson Jr9, N. Nathwani10, N. Shah11, N. Bumma12,
A. Jakubowiak13, R. Z. Orlowski14, H. Pei15, A. Cortoos16, S. Patel16, T. S. Lin16,
P. G. Richardson3, P. M. Voorhees17
1Wake Forest University School of Medicine, Winston-Salem; 2Winship Cancer Institute,
Emory University, Atlanta; 3Dana-Farber Cancer Institute, Boston; 4Huntsman Cancer
Institute, University of Utah School of Medicine, Salt Lake City; 5University of North
Carolina – Chapel Hill, Chapel Hill; 6Tisch Cancer Institute, Mount Sinai School of
Medicine, New York; 7Knight Cancer Institute, Oregon Health & Science University,
Portland; 8University of Alabama at Birmingham, Birmingham; 9Simmons Comprehensive
Cancer Center, UT Southwestern Medical Center, Dallas; 10Judy and Bernard Briskin
Center for Multiple Myeloma Research, City of Hope Comprehensive Cancer Center, Duarte;
11Department of Medicine, University of California San Francisco, San Francisco; 12Division
of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus; 13University
of Chicago Medical Center, Chicago; 14Department of Lymphoma and Myeloma, The University
of Texas MD Anderson Cancer Center, Houston; 15Janssen Research & Development, LLC,
Titusville; 16Janssen Scientific Affairs, LLC, Horsham; 17Levine Cancer Institute,
Atrium Health, Charlotte, United States of America
Background: In the primary analysis of the phase 2 randomized GRIFFIN study, Daratumumab
(DARA) + lenalidomide, bortezomib, and dexamethasone (D-RVd) improved the stringent
complete response (sCR) rate by end of consolidation for transplant-eligible newly
diagnosed multiple myeloma (NDMM) (42.4% vs 32.0%; 1-sided P = 0.068). With longer
follow-up (median, 38.6 mo), D-RVd vs RVd improved minimal residual disease (MRD)-negativity
(10–5) rates in clinically relevant subgroups (ISS stage III, 71% vs 36%; high cytogenetic
risk, 44% vs 29% [del17p, t(4;14), or t(14;16)]; revised high cytogenetic risk, 55%
vs 32% [del17p, t(4;14), t(14;16), t(14;20), or gain 1q]).
Aims: In this post-hoc analysis we present results of sustained MRD negativity (median
follow-up, 38.6 mo) in the same subgroups and in patients (pts) with ≥CR.
Methods: Transplant-eligible NDMM pts were randomized 1:1 to 4 D-RVd/RVd induction
cycles, ASCT, 2 D-RVd/RVd consolidation cycles, and 2 years of maintenance therapy
with lenalidomide (R) ± DARA. For induction/consolidation (21-day cycles), pts received
R (25 mg PO Days [D] 1-14), V (1.3 mg/m2 SC D1, 4, 8, 11), and d (40 mg PO weekly)
± DARA (16 mg/kg IV D1, 8, 15 of Cycles 1-4 and D1 of Cycles 5-6). In maintenance
(28-day cycles), pts received R (10 mg PO D1-21; if tolerated, 15 mg in Cycles 10+)
± DARA (16 mg/kg IV Q8W/Q4W or 1800 mg SC per protocol amendments). The primary endpoint
was sCR rate by end of consolidation.
Results: The following features were balanced among randomized pts (D-RVd, n = 104;
RVd, n = 103): high cytogenetic risk (16; 14), revised high cytogenetic risk (42;
37), gain 1q (34; 28), and ISS stage III (14; 14). Sustained MRD-negativity rates
at 10–5 lasting ≥6 and ≥12 months were higher for D-RVd vs RVd among all high-risk
subgroups (Table). D-RVd was superior to RVd for rates of sustained MRD negativity
lasting ≥12 months for pts with ≥CR (53.7% vs 20.3%) and sCR (59.1% vs 17.4%; Table).
Among all pts with sustained MRD negativity, only 1 D-RVd pt subsequently had disease
progression, and 1 RVd pt died. Additional data on MRD at 10–6 and PFS will be presented.
Image:
Summary/Conclusion: MRD data in GRIFFIN show that the addition of DARA to RVd induction/consolidation
and R maintenance may lead to durable MRD-negativity (10–5) rates in pts with transplant-eligible
NDMM with high cytogenetic risk, ISS stage III, and those who achieve ≥CR or sCR,
however larger studies are needed.
P935: REAL WORLD COMPARATIVE ANALYSIS OF THE EFFICACY OF TECLISTAMAB VERSUS CURRENT
TREATMENTS IN PATIENTS WITH TRIPLE-CLASS EXPOSED RELAPSED/REFRACTORY MULTIPLE MYELOMA
FROM THE LOCOMOTION STUDY
H. Einsele1,*, P. Moreau2, M. Delforge3, N. W. van de Donk4, F. Ghilotti5, J. Diels6,
A. Elsada7, V. Strulev6, L. Pei8, R. Kobos8, J. Smit9, M. Slavcev10, K. Weisel11,
M.-V. Mateos12
1Universitätsklinikum Würzburg, Medizinische Klinik und Poliklinik II, Wuerzburg,
Germany; 2Hematology Clinic, University Hospital Hôtel-Dieu, Nantes, France; 3University
of Leuven, Leuven, Belgium; 4Amsterdam University Medical Center, Vrije Universiteit
Amsterdam, Amsterdam, Netherlands; 5Janssen-Cilag SpA, Cologno Monzese, United States
of America; 6Janssen Pharmaceutica NV, Beerse, Belgium; 7Janssen-Cilag, High Wycombe,
Buckinghamshire, United Kingdom; 8Janssen Research & Development, Raritan; 9Janssen
Research & Development, Spring House; 10Janssen Global Services, LLC, Raritan, United
States of America; 11University Medical Center Hamburg-Eppendorf, Hamburg, Germany;
12University Hospital of Salamanca/IBSAL, CIC, Salamanca, Spain
Background: Patients with triple-class exposed relapsed/refractory multiple myeloma
(TCE RRMM) have poor prognosis and limited treatment options after being treated with
≥3 lines of therapy (LOT). Teclistamab (tec), a B-cell maturation antigen × CD3 bispecific
antibody, is currently being evaluated in the MajesTEC-1 (NCT04557098) study. MajesTEC-1
is a single-arm, phase 1/2 study in patients with TCE RRMM, previously exposed to
an immunomodulatory drug, a proteasome inhibitor, and an anti-CD38 antibody and received
≥3 LOT.
Aims: Since MajesTEC-1 lacks a control arm, we performed a real-world comparative
analysis of the efficacy of tec vs currently used treatments in clinical practice
by creating an external real-world control arm from LocoMMotion (NCT04035226), a prospective
study of real-world clinical practice (RWCP) efficacy and safety outcomes in patients
with TCE RRMM who received ≥3 LOT.
Methods: Patients from LocoMMotion (248 patients, clinical cutoff May 21, 2021) who
met MajesTEC-1 eligibility criteria were used to create the external control arm for
MajesTEC-1. Individual patient-level data were included for 150 patients from MajesTEC-1
treated with tec (1.5 mg/kg weekly) at a clinical cutoff of Sep 7, 2021. Inverse probability
of treatment weighting with average treatment effect on the treated was used to adjust
for imbalances in prognostically significant baseline covariates: Eastern Cooperative
Oncology Group performance status, gender, type of MM, prior transplant, refractory
status, International Staging System stage, time to progression on prior LOT, extramedullary
disease, number of prior LOT, time since diagnosis, average duration of prior LOT,
age, hemoglobin, lactate dehydrogenase and creatinine clearance. Efficacy of tec vs
RWCP was measured for overall response rate (ORR), very good partial response (VGPR)
rate, complete response or better (≥CR) rate, duration of response (DOR), progression-free
survival (PFS), and overall survival (OS). Odds ratio, transformed into a response-rate
ratio (RR) (along with 95% confidence interval [CI]), derived from weighted logistic
regression was used for binary endpoints (ORR, VGPR rate, and ≥CR rate). Hazard ratios
(HRs) and 95% CIs for time-to-event endpoints (DOR, PFS, and OS) were computed using
a weighted Cox proportional hazards model.
Results: After reweighting the external RWCP cohort, the 2 cohorts had well balanced
baseline characteristics. Patients treated with tec had improved outcomes vs current
treatments used in RWCP: ORR (RR: 2.31; 95% CI 1.75–2.87; P<0.0001), VGPR rate (RR:
5.54; 95% CI 3.38–7.70; P<0.0001), ≥CR rate (RR 91.50; 95% CI 12.66–661.43; P<0.0001),
DOR (HR 0.17; 95% CI 0.08–0.36; P<0.0001), PFS (HR 0.47; 95% CI 0.34–0.67; P<0.0001),
and OS (HR 0.69; 95% CI 0.46–1.05; P=0.08).
Summary/Conclusion: In this analysis, significantly improved efficacy for almost all
outcomes was observed with tec when compared with RWCP. This highlights the potential
of tec as a highly effective treatment option for patients with TCE RRMM who have
been exposed to ≥3 LOT.
P936: TIME TO RESPONSE, DURATION OF RESPONSE, AND PATIENT-REPORTED OUTCOMES WITH DARATUMUMAB
PLUS RD VS RD ALONE IN TRANSPLANT-INELIGIBLE PATIENTS WITH NDMM: SUBGROUP ANALYSIS
OF THE PHASE 3 MAIA STUDY
T. Facon1,*, S. K. Kumar2, T. Plesner3, P. Moreau4, N. Bahlis5, H. Goldschmidt6, M.
O’Dwyer7, A. Perrot8, C. P. Venner9, K. Weisel10, J. R. Mace11, N. Raje12, M. Tiab13,
M. Macro14, L. Frenzel15, X. Leleu16, H. Pei17, F. Borgsten18, S. Z. Usmani19
1University of Lille, CHU Lille, Service des Maladies du Sang, Lille, France; 2Department
of Hematology, Mayo Clinic Rochester, Rochester, MN, United States of America; 3Vejle
Hospital and University of Southern Denmark, Vejle, Denmark; 4Hematology Department,
University Hospital Hôtel-Dieu, Nantes, France; 5Arnie Charbonneau Cancer Research
Institute, University of Calgary, Calgary, AB, Canada; 6University Hospital Heidelberg,
Internal Medicine V and National Center for Tumor Diseases (NCT), Heidelberg, Germany;
7Department of Medicine/Haematology, NUI, Galway, Ireland; 8CHU de Toulouse, IUCT-O,
Université de Toulouse, UPS, Service d’Hématologie, Toulouse, France; 9Cross Cancer
Institute, University of Alberta, Edmonton, AB, Canada; 10Department of Oncology,
Hematology and Bone Marrow Transplantation with Section of Pneumology, University
Medical Center Hamburg-Eppendorf, Hamburg, Germany; 11Florida Cancer Specialists,
St. Petersburg, FL; 12Center for Multiple Myeloma, Massachusetts General Hospital
Cancer Center, Boston, MA, United States of America; 13CHD Vendée, La Roche sur Yon;
14Centre Hospitalier Universitaire (CHU) de Caen, Caen; 15Department of Clinical Haematology,
Hopital Necker-Enfants Malades, Paris; 16CHU Poitiers, Hôpital la Milétrie, Poitiers,
France; 17Janssen Research & Development, LLC, Titusville, NJ, United States of America;
18Janssen-Cilag, Birkerød, Denmark; 19Memorial Sloan Kettering Cancer Center, New
York, NY, United States of America
Background: In the phase 3 MAIA study, adding daratumumab (DARA) to lenalidomide and
dexamethasone (Rd) improved progression-free survival (primary endpoint), overall
survival, duration of response, and patient-reported outcomes (PROs) in transplant-ineligible
patients with newly diagnosed multiple myeloma (NDMM).
Aims: Here we report a MAIA subgroup analysis of time to response, duration of response,
and PROs.
Methods: Transplant-ineligible patients with NDMM received 28-day cycles of Rd (R
25 mg PO on Days 1-21; d 40 mg PO QW) ± DARA (16 mg/kg IV QW in Cycles 1-2, Q2W in
Cycles 3-6, and Q4W thereafter) until disease progression or unacceptable toxicity.
Secondary endpoints included time to response and duration of response. PROs were
measured using the EORTC QLQ-C30, with treatment effects assessed via mixed-effects
model with repeated measures.
Results: In total, 368 patients were assigned to the DARA plus lenalidomide and dexamethasone
(D-Rd) group and 369 patients to the Rd group; 162 (44%) D-Rd patients and 142 (38%)
Rd patients had renal impairment (defined as baseline CrCl ≤60 mL/min). At a 56.2-mo
median follow-up, median times to very good partial response or better (≥VGPR) and
complete response or better (≥CR) were shorter with D-Rd vs Rd in the overall study
population and in the subgroups of patients with and without renal impairment (Table).
Among patients who achieved ≥CR or partial response or better (≥PR), higher proportions
of D-Rd vs Rd patients had not experienced disease progression at 48 mo (Table). Among
patients with renal impairment, greater improvements from baseline in patient-reported
pain, fatigue, and nausea and vomiting symptom scores were observed with D-Rd vs Rd
across most timepoints; a notably greater meaningful reduction in pain symptom score
was seen with D-Rd vs Rd as early as Cycle 6 Day 1 (least squares mean change from
baseline, −14.9 vs −7.0; P=0.0241). Analyses for additional patient subgroups will
be presented.
Image:
Summary/Conclusion: In transplant-ineligible patients with NDMM, D-Rd showed more
rapid deep responses as well as more durable responses vs Rd, regardless of renal
function. Improvements in patient-reported symptoms were generally greater with D-Rd
vs Rd in patients with renal impairment. Our results support the use of D-Rd in transplant-ineligible
patients with NDMM.
P937: PRECLINICAL DISCOVERY AND EARLY FINDINGS FROM THE PHASE 1, DOSE-ESCALATION STUDY
OF WVT078, A BCMA-CD3 BISPECIFIC ANTIBODY, IN PATIENTS WITH R/R MULTIPLE MYELOMA
M. S. Raab1,*, Y. C. Cohen2,3, F. Schjesvold4,5, K. Aardalen6, A. Oka6, A. Spencer7,
M. Wermke8, P. Hari9, J. L. Kaufman10, A. M. Cafro11, E. M. Ocio12, N. Doki13, K.
Henson6, G. Trabucco6, P.-E. Juif14, R. Chawla14, J. Cotton6, A. Fessehatsion6, L.
Fan6, J. Blankenship6, B. Granda6, H. Lu6, S. De Vita6
1Department of Internal Medicine, Heidelberg University Hospital, Heidelberg, Germany;
2Tel-Aviv Sourasky (Ichilov) Medical Center; 3Sackler Faculty of Medicine, Tel Aviv
University, Tel Aviv, Israel; 4Oslo Myeloma Center, Department of Hematology, Oslo
University Hospital; 5KG Jebsen Center for B cell malignancies, University of Oslo,
Oslo, Norway; 6Novartis Institutes for BioMedical Research, Cambridge, MA, United
States of America; 7The Alfred Hospital, Melbourne, VIC, Australia; 8NCT/UCC Early
Clinical Trial Unit, Universitätsklinikum Carl Gustav Carus an der Technische Universität,
Dresden, Germany; 9Division of Hematology & Oncology, Medical College of Wisconsin,
Milwaukee, WI; 10Winship Cancer Institute, Emory University, Atlanta, GA, United States
of America; 11Department of Hematology, ASST Grande Ospedale Metropolitano, Niguarda,
Milan, Italy; 12Hospital Universitario Marqués de Valdecilla (IDIVAL), Universidad
de Cantabria, Santander, Spain; 13Hematology Division, Tokyo Metropolitan Cancer and
Infectious Diseases Center, Tokyo, Japan; 14Novartis Institutes for BioMedical Research,
Basel, Switzerland
Background: B-cell maturation antigen (BCMA) is a member of the tumor necrosis factor
receptor superfamily with selective expression on benign and malignant plasma cells.
Signaling through BCMA mediates survival and proliferation of multiple myeloma (MM)
cells. BCMA is a validated clinical target with multiple BCMA-directed therapies approved
or in development for patients with relapsed and/or refractory (r/r) MM. Bispecific
antibodies (BsAbs) offer a convenient alternative to chimeric antigen receptor–modified
T-cell (CAR-T) therapies targeting BCMA and are demonstrating high response rates
in patients with MM. Here we describe the discovery of WVT078, a novel BCMA-CD3 BsAb
and report initial clinical data from the ongoing phase 1 study (NCT04123418) evaluating
ascending doses of WVT078 in patients with r/r MM.
Aims: To evaluate the safety and tolerability of the novel BCMA-CD3 BsAb WVT078 in
patients with r/r MM.
Methods: Multiple BCMA-CD3 BsAb candidates were characterized in vitro and in vivo
for T cell activation and anti-MM activity. The activity and tolerability of prioritized
candidates were further assessed in an integrated pharmacology study in cynomolgus
monkeys. WVT078 was selected for the phase 1 clinical study and is being tested in
patients with MM who are r/r to ≥2 standard of care regimens including an immunomodulatory
drug, a proteasome inhibitor, and an anti-CD38 agent (if available). Patients who
have received a prior BCMA CAR-T or BCMA-antibody-drug conjugates therapy are included,
whereas those with prior treatment with BCMA-CD3 bispecifics are excluded.
Results: WVT078, a BCMA-CD3 BsAb with high affinity binding to BCMA (with subnanomolar
KD), was selected to effectively target MM cells with a low BCMA expression, an emerging
mechanism of resistance to BCMA-targeted therapies. WVT078 demonstrated potent anti-MM
activity and efficient T-cell activation in preclinical models, and was well tolerated
in cynomolgus monkeys. As of December 08, 2021, a total of 33 patients with r/r MM
were treated with intravenous WVT078 once weekly at the following dose levels: 3,
6, 12, 24, 48, 64, 96, 192, and 250 µg/kg of body weight. Twenty-five patients (75.8%)
discontinued treatment due to progressive disease (n=22), physician decision (n=2)
or adverse event (AE, n=1). The most frequent (≥20%) treatment-related AEs (TRAEs)
of all grades across all doses included cytokine release syndrome (CRS, 60.6%), pyrexia
(39.4%), increased alanine aminotransferase (ALT, 30.3%), increased aspartate aminotransferase
(AST) and anemia (24.2% each), and the most common grade ≥3 TRAEs (≥10%) included
increased AST, lymphopenia (18.2% each), increased ALT (15.2%), CRS and neutropenia
(12.1% each). Clinical activity was observed starting at the dose of 48 µg/kg. At
the active doses of 48-250 µg/kg (n=26), the overall response rate (partial response
or better) was 34.6% (n=9; 90% CI, 19.4-52.6) and the complete response rate (stringent
complete response+complete response) was 11.5% (n=3; 90% CI, 3.2-27.2). The maximum
tolerated dose has not been reached. Pharmacokinetic data showed a trend towards dose-proportional
relationship and no evidence of accumulation.
Image:
Summary/Conclusion: WVT078, a novel BCMA-CD3 BsAb, demonstrated potent anti-MM activity
and favorable tolerability in preclinical models. In the phase 1 study, WVT078 exhibited
an acceptable safety profile and preliminary evidence of clinical activity. Clinical
evaluation of WVT078 as a single agent and in combination with a gamma secretase inhibitor
is ongoing.
P938: CARFILZOMIB-LENALIDOMIDE-DEXAMETHASONE CONSOLIDATION IN MYELOMA PATIENTS WITH
A POSITIVE FDG PET/CT AFTER UPFRONT AUTOLOGOUS STEM CELL TRANSPLANTATION: A PHASE
II STUDY (CONPET)
J. N. Nørgaard1,2,3,*, N. Abildgaard4,5, A. Lysén1,3, G. Tsykunova6, A. J. Vangsted7,
C. Joao8, N. Remen1, L. Osnes9, J. P. Connelly10, M.-E. R. Revheim2,10, F. Schjesvold1,3
1Oslo Myeloma Center, Department of Hematology, Oslo University Hospital; 2Institute
of Clinical Medicine; 3KG Jebsen Center for B-cell Malignancies, University of Oslo,
Oslo, Norway; 4Department of Hematology, Odense University Hospital; 5Department of
Clinical Research, University of Southern Denmark, Odense, Denmark; 6Division of Hematology,
Haukeland University Hostpital, Bergen, Norway; 7Departmen of Hematology, Rigshospitalet,
Copenhagen, Denmark; 8Department of Hematology, Champalimaud Centre for the Unknown,
Lisboa, Portugal; 9Department of Immunology; 10Division for Radiology and Nuclear
Medicine, Oslo University Hospital, Oslo, Norway
Background: [18F]-Fluorodeoxyglucose positron emission tomography/computed tomography
(FDG PET/CT) positivity after first line treatment with autologous stem cell transplantation
(ASCT), is strongly correlated with reduced progression free survival and overall
survival (Moreau et al., JCO, 2017). However, FDG PET/CT positive patients who obtain
FDG PET/CT negativity after treatment seem to have comparable outcomes to patients
who were FDG PET/CT negative at baseline (Davies et al., Haematologica 2018).
Aims: Aiming for FDG PET/CT negativity may be an important goal in myeloma treatment.
The use of FDG PET/CT positivity as an indication for consolidation therapy after
ASCT has not been studied before. We here present the complete data of the primary
endpoint, PET/CT negativity after KRd consolidation.
Methods: Patients with multiple myeloma who had received standard first line treatment
including ASCT and achieved very good partial response (VGPR) or better, were examined
by FDG PET/CT. Patients who were FDG PET/CT positive defined by the Italian Myeloma
criteria for PET USe (IMPETUS) (Nanni C et al., EJNMMI 2016 and 2018) were included
in the treatment phase of the study and were assessed for minimal residual disease
(MRD) by Euroflow (sensitivity: 10-5) before treatment. FDG PET/CT examinations were
centrally read by two experienced nuclear medicine radiologists. The treatment consisted
of four 28-day cycles of KRd (carfilzomib 36 mg/m2 day 1,2,8,9,15 and 16 (except 20 mg/m2
day 1 and 2 first cycle), lenalidomide 25 mg day 1-21 all cycles and dexamethasone
40 mg day 1,8,15 and 22 all cycles). After 4 cycles, FDG PET/CT and Euroflow for MRD
were repeated for response evaluation. Both patients with FDG PET/CT negativity and
patients with FDG PET/CT positivity at baseline are followed for progression free
survival and overall survival.
Results: 159 patients were screened with FDG PET/CT. Fifty-three patients of 159 (33%)
had a positive FDG PET/CT result. Among FDG PET/CT positive patients, a higher proportion
had ISS score III, high-risk cytogenetics and VCd induction was more common (Table
1). Forty-eight patients completed KRd treatment. Sixteen patients (33%) converted
into FDG PET/CT negativity. More patients with previous VRd induction than VCd induction
converted from FDG PET/CT positive to FDG PET/CT negative (38% vs 20%). No patients
stopped treatment due to adverse events. Twenty-seven of 49 (55%) patients with a
FDG PET/CT positive result were MRD negative before KRd consolidation. A higher proportion
of MRD negative patients vs MRD positive patients before KRd treatment, converted
into FDG PET/CT negativity, with 37% vs 28%, respectively. For the proportion of patients
that after KRd treatment was both FDG PET/CT negative and MRD negative by EuroFlow,
the difference was even higher (37% vs 5% in MRD negative vs MRD positive patients,
respectively). In total, the MRD negativity rate increased from 55% to 76%
Image:
Summary/Conclusion: Before KRd consolidation, 33% of patients in VGPR or better after
first line treatment including ASCT were FDG PET/CT positive. High-risk cytogenetics,
ISS score III and VCd induction were more common in FDG PET/CT positive patients.
After four cycles of KRd, 33% of patients converted from FDG PET/CT positivity to
negativity. A higher proportion converted into FDG PET/CT negativity in patients who
were MRD negative before KRd treatment, and in patients who had received VRd. KRd
consolidation is feasible and converts a clinically significant proportion of FDG
PET/CT positive patients to negativity. Patients are followed for PFS and OS.
P939: SYNERGISTIC EFFECTS OF LOW DOSE BELANTAMAB MAFODOTIN IN COMBINATION WITH A GAMMA-SECRETASE
INHIBITOR (NIROGACESTAT) IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA (RRMM):
DREAMM-5 STUDY
A. Nooka1,*, S. Lonial1, S. Grosicki2, M. Hus3, K. Song4, T. Facon5, N. S. Callander6,
V. Ribrag7, K. Uttervall8, H. Quach9, V. Vorobyev10, C.-K. Min11, S. Cheng12, L. M.
Smith12, J. Yu13, T. Collingwood13, B. Holkova13, B. E. Kremer13, I. V. Gupta13, P.
G. Richardson14, M. C. Minnema15
1Emory University, Winship Cancer Institute, Atlanta, GA, United States of America;
2Department of Hematology and Cancer Prevention, Medical University of Silesia, Katowice;
3Katedra i Klinika Hematoonkologii i Transplantacji Szpiku, Lublin, Poland; 4Vancouver
General Hospital, Vancouver, BC, Canada; 5Department of Haematology, Lille University
Hospital, Lille, France; 6University of Wisconsin, Carbone Cancer Center, Madison,
WI, United States of America; 7Institut Gustave Roussy, Villejuif, France; 8Karolinska
University Hospital, Stockholm, Sweden; 9University of Melbourne, St. Vincent’s Hospital
Melbourne, Melbourne, VIC, Australia; 10S P Botkin City Clinical Hospital, Moscow,
Russia; 11Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, South
Korea; 12SpringWorks Therapeutics, Stamford, CT; 13GlaxoSmithKline, Upper Providence,
PA; 14Dana-Farber Cancer Institute, Boston, MA, United States of America; 15University
Medical Center Utrecht, Utrecht, Netherlands
Background: Preclinical data demonstrate that nirogacestat, a gamma-secretase inhibitor,
may increase cell-surface levels of a B-cell maturation antigen (BCMA) and reduce
soluble BCMA levels, which could enhance anti-BCMA agent activity in multiple myeloma.
In the DREAMM-5 (NCT04126200) Phase I/II platform trial belantamab mafodotin (belamaf;
BLENREP), a BCMA-targeting antibody-drug conjugate, is being evaluated in combination
with nirogacestat.
Aims: The aim of this study is to determine if the combination can result in similar
efficacy and an improved ocular safety profile compared to the currently approved
belamaf schedule (single agent dose 2.5 mg/kg Q3W) in patients with RRMM which showed
a 31% overall response rate (ORR) and 44.5% Gr3/4 keratopathy (BLENREP US prescribing
information).
Methods: This cohort within the DREAMM-5 nirogacestat combination sub-study has a
sequential dose-exploration (DE) phase evaluating 0.95 mg/kg Q3W belamaf with 100 mg
BID nirogacestat continuously, followed by a randomized cohort expansion (CE) comparing
the combination to a belamaf 2.5 mg/kg Q3W arm.
Results: Preliminary results from the 10 patients in the DE cohort with low-dose belamaf
+ nirogacestat, are presented in this abstract. Patients had a median (range) of 4.5
(3–10) prior lines of therapy. At time of data cut-off (Nov 15, 2021), patients received
a median (range) of 7 cycles (1–26). The ORR was 60% (n=6/10) and 20% (n=2) achieved
a very good partial response (Table). The key emergent adverse events (AEs) included
ocular events (n=7 [70%]; ≥Grade [Gr] 3, n=2 [20%] of which Gr 3 keratopathy was reported
in 1 patient [10%]), diarrhea (n=7 [70%]; Gr3, n=1 [10%]) and hypophosphatemia (n=7
[70%]; Gr3, n=1, [10%]). There were 2 Grade 5 AEs, neither of which were related to
study treatment. No patient permanently discontinued study due to treatment related
AEs.
Image:
Summary/Conclusion: Encouraging clinical activity and a manageable safety profile
is observed with low dose belamaf (0.95 mg/kg Q3W) + nirogacestat (100 mg BID continuously)
in patients with RRMM. This ongoing sub-study is actively recruiting patients and
will continue to evaluate belamaf + nirogacestat efficacy and safety. Updated results
will be reported at the congress.
Funding Statement: GSK (208887); drug linker technology licensed from Seagen Inc.;
mAb produced using POTELLIGENT Technology licensed from BioWa.
This abstract was previously submitted to the American Society of Clinical Oncology
(ASCO) Annual Meeting, June 3–7, 2022, and is submitted on behalf of the original
authors with their permission. ©2022 American Society of Clinical Oncology, Inc. Reused
with permission. All rights reserved.
P940: SAFETY AND CLINICAL ACTIVITY OF BELANTAMAB MAFODOTIN WITH PEMBROLIZUMAB IN PATIENTS
WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA (RRMM): DREAMM-4 STUDY
A. Suvannasankha1,*, N. Bahlis2, S. Trudel3, K. Weisel4, C. Koenecke5, A. Oriol6,
P. M. Voorhees7, A. A. Alonso8, N. S. Callander9, M. V. Mateos Manteca10, N. Reddy11,
S. Hakim12, N. Patel13, D. Williams12, R. C. Jewell13, X. Zhou12, I. Gupta14, A. K.
Nooka15
1Indiana University Simon Cancer Center and Roudebush VAMC, Indianapolis, IN, United
States of America; 2Arnie Charbonneau Cancer Research Institute, University of Calgary,
Calgary, AB; 3Princess Margaret Cancer Centre, Toronto, ON, Canada; 4University Medical
Center Hamburg-Eppendorf, Hamburg; 5Hannover Medical School, Clinic for Hematology,
Hemostasis, Oncology and Stem Cell Transplantation, Hannover, Germany; 6Institut Català
d’Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona,
Barcelona, Spain; 7Levine Cancer Institute, Atrium Health, Charlotte, NC, United States
of America; 8Hospital Universitario Quirónsalud Madrid, Madrid, Spain; 9Carbone Cancer
Center, University of Wisconsin, Madison, WI, United States of America; 10Instituto
de Investigación Biomédica de Salamanca and Centro de Investigación del Cáncer, Hospital
Universitario de Salamanca, Salamanca, Spain; 11Merck & Co., Inc., Kenilworth, NJ;
12GlaxoSmithKline, Upper Providence, PA; 13GlaxoSmithKline, Research Triangle Park,
NC; 14GlaxoSmithKline, Philadelphia, PA; 15Winship Cancer Institute, Emory University
Hospital, Atlanta, GA, United States of America
Background: Belantamab mafodotin (belamaf; BLENREP), a B-cell maturation antigen (BCMA)
targeted antibody–drug conjugate approved for adult patients with RRMM, has a multimodal
mechanism that eliminates myeloma cells via direct cytotoxicity and a systemic anti-tumor
immune response, which may be augmented by an immune checkpoint inhibitor.
Aims: DREAMM-4 (NCT03848845) assessed safety and clinical activity of belamaf with
pembrolizumab (pembro) in RRMM.
Methods: This was a Phase I/II, single-arm, open-label study of adults with RRMM after
≥3 lines of therapy (LOT, including anti-CD38 monoclonal antibody, proteasome inhibitor,
and immunomodulator). Part 1 established the dose of belamaf 2.5 mg/kg with pembro
200 mg, both IV Q3W up to 35 cycles, for the Part 2 expansion. Primary efficacy endpoint
was investigator-assessed overall response rate (ORR, ≥partial response [PR] per IMWG
criteria by investigator). Secondary endpoints included duration of response (DoR),
progression-free survival (PFS), adverse events (AEs) per NCI-CTCAE v4.03, and pharmacokinetics
(PK).
Results: This primary analysis of all treated pts (as of Oct 18, 2021) included 34
pts (6 in Part 1 and 28 in Part 2). In both parts, median prior LOT was 5 (range 3–13);
10 pts (29%) had high-risk cytogenetics and 9 (26%) had extramedullary disease. ORR
was 47%, with most responses (10/16 pts) ≥ very good PR (VGPR, Table). Median follow-up
was 14.7 months (mo); median (95% CI) DoR was 8.0 (2.1–not reached) mo; median PFS
was 3.4 (1.4–5.6) mo. Most pts had ≥1 AE (any grade [Gr]: 97%; Gr ≥3: 74%) and treatment-related
AE (TRAE, any Gr: 97%; Gr ≥3: 65%). Most common (≥35%) AEs were keratopathy (any Gr:
76%; Gr ≥3: 38%), vision blurred (any Gr: 38%; Gr ≥3: 0%), and thrombocytopenia (any
Gr: 35%; Gr ≥3: 29%). AEs led to dose delays (65%) and dose reductions (32%), but
not discontinuation. Nine pts had a serious AE (SAE); 4 pts had ≥1 SAE related to
study treatment. Two pts had immune-related AEs of Gr 1 (gout and autoimmune hypothyroidism).
Preliminary PK and soluble BCMA data were consistent with single-agent belamaf therapy.
Image:
Summary/Conclusion: Belamaf + pembro demonstrated a favorable ORR compared with single-agent
belamaf in heavily pre-treated RRMM. No new TRAEs were identified; AE frequency and
severity were similar to single-agent belamaf. Correlative biomarker studies are ongoing.
Funding Statement: GSK (205207) in collaboration with Merck Sharp & Dohme Corp., a
subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA. Drug linker technology licensed
from Seagen Inc. mAb produced using POTELLIGENT technology licensed from BioWa.
This abstract was previously submitted to the American Society of Clinical Oncology
(ASCO) Annual Meeting, June 3–7, 2022, and is submitted on behalf of the original
authors with their permission. ©2022 American Society of Clinical Oncology, Inc. Reused
with permission. All rights reserved.
P941: SAFETY AND CLINICAL ACTIVITY OF BELANTAMAB MAFODOTIN WITH LENALIDOMIDE PLUS
DEXAMETHASONE IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA (RRMM): DREAMM-6
ARM-A INTERIM ANALYSIS
H. Quach1,*, M. Gironella2, C. Lee3, R. Popat4, P. Cannell5, R. S. Kasinathan6, B.
Chopra7, R. Rogers6, G. Ferron-Brady6, S. Shafi-Harji8, N. Patel7, J. Opalinska6,
I. Gupta6, B. Augustson9
1University of Melbourne, St. Vincent’s Hospital Melbourne, Melbourne, Australia;
2Department of Hematology, University Hospital Vall d’Hebron, Barcelona, Spain; 3Royal
Adelaide Hospital, Adelaide, Australia; 4NIHR UCLH Clinical Research Facility, University
College London Hospitals, NHS Foundation Trust, London, United Kingdom; 5Department
of Medicine, Royal Perth Hospital, Perth, Australia; 6GlaxoSmithKline, Upper Providence,
PA, United States of America; 7GlaxoSmithKline, London; 8GlaxoSmithKline, Stevenage,
United Kingdom; 9Department of Haematology, Sir Charles Gairdner Hospital, Perth,
Australia
Background: Belantamab mafodotin (belamaf; BLENREP) is a B-cell maturation antigen-targeting
antibody–drug conjugate approved for patients with RRMM as monotherapy at 2.5 mg/kg
Q3W. Preclinical data demonstrate synergy between belamaf and lenalidomide (Len),
suggesting added benefit when combined with standard of care such as Len + dexamethasone
(Dex).
Aims: DREAMM-6 (NCT03544281) Arm A is evaluating belamaf in combination with LenDex
in patients with RRMM.
Methods: This ongoing, two-part, two-arm, open-label study included patients with
RMMM previously treated with ≥1 line of therapy (LOT). Patients received 4 belamaf
doses/schedules (1.9 mg/kg Q8W or Q4W; 2.5 mg/kg Q4W or Q4W SPLIT dose [50% on Days
(D)1, 8] IV) in combination with Len (20 mg PO D1–21) and Dex (20 mg PO/IV D1, 8,
15, 22). Primary objectives were safety (including treatment-related [TR]AEs related
to the combination belamaf and LenDex), tolerability, and efficacy (including overall
response rate [ORR] defined as ≥partial response).
Results: As of this interim analysis (data cut: July 23, 2021), 45 patients received
≥1 dose (12 at 1.9 mg/kg Q8W; 4 at 1.9 mg/kg Q4W; 16 at 2.5 mg/kg Q4W; 13 at 2.5 mg/kg
Q4W SPLIT). Across cohorts, the median age was 68 years (range: 36–80). Thirteen patients
(29%) had high-risk cytogenetics and 6 (13%) had extramedullary disease. Median prior
LOT was 3 (range: 1–11) and 26 (58%) had prior Len treatment.
The median duration of follow-up and ORR ranged across cohorts (Table). The median
duration of response was only reached in the 1.9 mg/kg Q4W cohort (11.1 mo [95% CI:
3.7–not reached [NR]). At the time of data cut, median progression-free survival was
not reached in the 1.9 mg/kg Q8W or 2.5 mg/kg Q4W cohorts. Gr ≥3 TRAEs occurred in
42–85%. Gr ≥3 keratopathy occurred in 0 patients in 1.9 mg/kg Q8W, 1 patient (25%)
in 1.9 mg/kg Q4W, 8 patients (50%) in 2.5 mg/kg Q4W, and 6 patients (46%) in 2.5 mg/kg
Q4W SPLIT cohorts.
Image:
Summary/Conclusion: Belamaf + LenDex had a tolerable safety profile, with no new safety
signals identified in patients with RRMM. AEs, including keratopathy, were common
but manageable with dose modifications. Encouraging clinical activity is observed
with this combination in patients with RRMM. Follow-up/correlative studies are ongoing.
P942: DREAMM-9: PHASE I STUDY OF BELANTAMAB MAFODOTIN PLUS STANDARD OF CARE IN PATIENTS
WITH TRANSPLANT-INELIGIBLE NEWLY DIAGNOSED MULTIPLE MYELOMA
S. Z. Usmani1,*, M. Mielnik2, Y. Koh3, A. Alonso Alonso4, X. Leleu5, H. Quach6, C.-K.
Min7, W. Janowski8, A.-O. Abdallah9, M. Garg10, I. Sandhu11, E. M. Ocio San Miguel12,
A. Oriol13, P. Rodriquez-Otero14, K. Ramasamy15, K. Weisel16, B. Besemer17, M. Cavo18,
X. L. Zhou19, M. C. Kaisermann20, C. M. Bego Marques21, D. Williams20, F. Carreno20,
B. E. Kremer20, I. V. Gupta20, M. Hus2
1Memorial Sloan Kettering Cancer Center, New York, NY, United States of America; 2Department
of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin,
Poland; 3Seoul National University Hospital, Seoul, South Korea; 4University Hospital
Quirón Madrid, Madrid, Spain; 5CHU de Poitiers, Poitiers, France; 6St.Vincent’s Hospital
Melbourne, Melbourne, Australia; 7Catholic University of Korea Seoul St. Mary’s Hospital,
Seoul, South Korea; 8Calvary Mater Newcastle, Newcastle, Australia; 9University of
Kansas, Kansas City, KS, United States of America; 10Leicester Royal Infirmary, Leicester,
United Kingdom; 11University of Alberta, Edmonton, Canada; 12Hospital Universitario
Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, Santander; 13Institut Català
d’Oncologia and Institut Josep Carreras - Hospital Universitari Germans Trias i Pujol
(HUGTP), Badalona; 14Department of Hematology, Clínica Universidad de Navarra, Pamplona,
Spain; 15Churchill Hospital, Headington, Oxford, United Kingdom; 16University Medical
Center Hamburg-Eppendorf, Hamburg; 17University of Tübingen, Tübingen, Germany; 18IRCCS
Azienda Ospedaliero-Universitaria di Bologna, Istitutodi Ematologia “Seràgnoli”, Universitàdegli
Studi di Bologna, Bolonga, Italy; 19GlaxoSmithkline, Waltham, MA; 20GlaxoSmithKline,
Upper Providence, PA, United States of America; 21GlaxoSmithKline, Barcelona, Spain
Background: The bortezomib, lenalidomide, and dexamethasone (VRd) regimen is a standard
of care for newly diagnosed multiple myeloma (NDMM). Belantamab mafodotin (belamaf)
is a B-cell maturation antigen-binding antibody-drug conjugate that eliminates myeloma
cells by a multimodal mechanism: direct cell kill and anti-myeloma tumor immune response.
Belamaf has demonstrated deep and durable responses as a monotherapy in the DREAMM-2
study of patients (pts) with relapsed/refractory multiple myeloma (RRMM). Preclinical
evidence of belamaf in combination with bortezomib or lenalidomide suggests enhanced
anti-myeloma activity, providing rationale for this treatment combination.
Aims: To evaluate the safety and tolerability of this combination in adult pts with
transplant-ineligible (TI) NDMM and establish the recommended Phase III dose.
Methods: DREAMM-9 (NCT04091126) is an ongoing Phase I, open-label, randomized study
of belamaf + VRd. The belamaf dose cohorts currently being evaluated are Cohort 1
(1.9 mg/kg Q3/4W), Cohort 2 (1.4 mg/kg Q6/8W), Cohort 3 (1.9 mg/kg Q6/8W), Cohort
4 (1.0 mg/kg Q3/4W), and Cohort 5 (1.4 mg/kg Q3/4W). Belamaf is given with VRd Q3W
until Cycle 8, and with Rd Q4W thereafter. After evaluation of safety data for Cohort
1, Cohorts 2–5 were opened in parallel and enrolled pts were randomized 1:1:1:1. Primary
endpoint is safety. Secondary endpoints include efficacy, tolerability, and pharmacokinetics
(PK).
Results: As of data cutoff (07 Dec 2021), 64 pts were analyzed across all cohorts.
Median age (range) was 73.0 (51–88) years, 55% were male, 80% were white, 8% had extramedullary
disease, 59% were International Staging System stage II or III, 20% had amp1q, and
17% had high-risk cytogenetics (≥1 of: t(4;14), t(14;16), del17p). The median duration
of follow-up varied: Cohort 1 (17.4 months [mo]), Cohort 2 (5.9 mo), Cohort 3 (6.1
mo), Cohort 4 (4.7 mo), Cohort 5 (5.8 mo). Median number of belamaf cycles were: Cohort
1 (6), Cohort 2 (3), Cohort 3 (3.5), Cohort 4 (4.5), and Cohort 5 (5). Most common
adverse events (AEs) across cohorts included thrombocytopenia (49%), constipation
(43%), diarrhea (32%), and peripheral sensory neuropathy (30%). AEs related to study
treatment were experienced by 61 (97%) pts. Belamaf-related grade 3/4 AEs occurred
in 24 (38%) pts. Belamaf dose reductions occurred in 11 (18%) pts, with dose delays
in 10 (16%) pts. Three pts experienced a fatal severe AE (unrelated to study treatment);
2 due to COVID-19 infection, 1 due to pancreatic adenocarcinoma. Early deep responses
were observed; 67–92% pts achieved ≥very good partial response (VGPR) (Table), with
median time to VGPR of 2.1–2.9 months across cohorts. Of pts with ≥VGPR, 17 were minimal
residual disease (MRD) negative, 10 in Cohort 1. As of data cutoff, 8–75% of pts achieved
best response of complete response (CR) or stringent CR (sCR). Grade 3 corneal exam
findings were reported in 25–58% of pts; grade 3 visual acuity changes were reported
in 21–75% of pts. No grade 4 corneal exam findings or visual acuity changes were reported
in pts receiving belamaf Q6/8W, compared with 0–17% and 0–8%, respectively, in the
Q3/4W cohorts. Belamaf PK profile was similar to that in pts with RRMM, accounting
for baseline characteristics.
Image:
Summary/Conclusion: Belamaf + VRd demonstrated high response rates in pts with TI
NDMM, with a high rate of MRD negativity indicating deep responses. No new safety
signals were observed relative to DREAMM-2. Study is ongoing to evaluate the safety
and efficacy of variable dose intensities of belamaf in combination with VRd.
P943: SURVIVAL OUTCOMES OF PATIENTS WITH MULTIPLE MYELOMA IN FRANCE: A COHORT STUDY
USING THE FRENCH NATIONAL HEALTHCARE DATABASE (SNDS)
X. Leleu1,*, B. Gorsh2, A. Bessou3, P. Paka2, J. De Nascimento3, X. Colin4, S. Landi5,
P. F. Wang2
1Department of Hematology, Centre Hospitalier Universitaire, Université de Poitiers,
Poitiers, France; 2GlaxoSmithKline, Upper Providence, PA, United States of America;
3IQVIA, Paris; 4GlaxoSmithKline, Rueil-Malmaison, France; 5GlaxoSmithKline, Research
Triangle Park, NC, United States of America
Background: While treatment options for multiple myeloma (MM) have expanded with the
introduction of novel therapies, it remains unclear how this has translated into better
patient outcomes. Although some patients can achieve long-term remission, MM remains
incurable. In oncology, overall survival (OS) is the gold standard for measuring the
clinical benefit of an agent. Real-world studies in several countries have shown increases
in OS in recent years for patients with MM; up-to-date real-world data on MM survival
in France are limited.
Aims: To assess the OS of patients with MM in France from diagnosis and by line of
therapy (LOT) and post triple-class exposure (TCE). Secondary aims were survival outcomes
by different subpopulations based on standard of care (SoC) treatment regimens and
transplant status.
Methods: This longitudinal retrospective cohort study used data from the French National
Healthcare database (Système National des Données de Santé – SNDS) from Jan 2013‒Dec
2019, which included records of outpatient claims, hospital discharges and death registry,
demographics, and reimbursement data for hospital and outpatient services. Eligible
patients (≥18 years) had ≥2 records of an MM diagnosis (ICD-10 codes C90/C90.0), and/or
long-term disease during the study period; ≥1 dispensation/administration of MM drug
treatment and date of death available.
LOT was algorithmically defined based on drug regimen, time since administration,
and gap between regimens. OS was analyzed from five time points: 1) from time of diagnosis;
2) from the start of line of LOT1 to LOT4; 3) from the start of first TCE (defined
as prior exposure to a proteasome inhibitor, an immunomodulatory agent and an anti-CD38
monoclonal antibody); 4) from the subsequent treatment following TCE, and 5) from
the subsequent treatment/no treatment following TCE and LOT5+ (indexed at the date
of the last observed treatment). Patient OS was assessed by Kaplan–Meier method and
death rates were calculated.
Results: A total of 14,309 patients were included in this analysis. Median age was
71 years at diagnosis and 51% of patients were male. Patient survival outcomes deteriorated
as their LOT advanced (Figure). Median OS from initial diagnosis was >5 years with
a slight increase for patients diagnosed more recently (63.8 months for patients diagnosed
in 2017 or later vs 60.8 months in those diagnosed in 2014 or later). From LOT1 to
LOT4 OS decreased from 61 to 14.8 months or about 30% to 50% with each additional
LOT. At the time of first TCE, daratumumab monotherapy/combination therapy were the
most common regimens. Median OS following TCE was 3.8 months. Retreatment with previous
agents were the SoC following TCE. Among TCE patients who had 4 prior LOTs, 43% of
them received a subsequent LOT with a median OS estimate of 8.2 months. For those
who discontinued treatment, median OS was only 1 month, indexing at the date of the
last observed treatment.
Image:
Summary/Conclusion: Patients with MM experience worsening survival outcomes with increasing
time from diagnosis and with subsequent LOT. Prognosis was especially poor among TCE
patients who received subsequent therapy and worse for those who did not receive additional
treatment. Despite therapeutic advances and slight increase in OS for patients diagnosed
more recently, an unmet need for improved access to novel therapies remains, especially
in those TCE patients.
P944: A PHASE I STUDY OF C-CAR088, A NOVEL HUMANIZED ANTI-BCMA CAR T CELL THERAPY
IN RELAPSED/REFRACTORY MULTIPLE MYELOMA
X. Qu1,*, G. An2, W. Sui2, T. Wang2, X. Zhang3, J. Yang3, Y. Zhang4, L. Zhang4, D.
Zhu5, J. Huang5, S. Zhu5, X. Yao5, J. Li5, C. Zheng5, S. Chen5, K. Zhu6, Y. Wei5,
X. Lv5, L. Lan5, Y. Yao5, D. Zhou4, P. Lu3, L. Qiu2, J. Li1
1Department of Hematology, The First Affiliated Hospital of Nanjing Medical University,
Nanjing; 2Department of Hematology, Institute of Hematology and Blood Diseases Hospital,
Tianjin; 3Department of Hematology, Hebei Yanda Lu Daopei Hospital, Langfang; 4Department
of Hematology, Peking Union Medical College Hospital, Beijing; 5Cellular Biomedicine
Group Inc, Shanghai; 6University of Maryland School of Medicine, Maryland, China
Background: Anti-B-cell maturation antigen (BCMA) chimeric antigen receptor-T cell
(CAR T) therapy shows remarkable efficacy in patients with relapsed or refractory
multiple myeloma (RRMM).
Aims: C-CAR088, a novel second-generation humanized anti-BCMA CAR T cell therapy,
was developed.
Methods: This phase I dose-escalation and expansion study assessed the safety and
efficacy of three dosages of C-CAR088 in patients with RRMM. As of July 2, 2021, 31
patients had been infused with C-CAR088.
Results: Any grade cytokine release syndrome (CRS) developed in 29 patients (93.5%),
and grade 3 CRS occurred in three patients (9.7%). One patient from the high-dose
group (4.5-6.0 × 106 CAR T cells/kg) developed grade 1 neurotoxicity. No dose-limiting
toxicities were observed in any dose group, and all adverse events were reversible
after proper management. The overall response, stringent complete response, complete
response (CR), and very good partial response rates were 96.4%, 46.4%, 10.7%, and
32.1%, respectively. The CR rate in the medium-dose (3.0 × 106 CAR T cells/kg) and
high-dose (4.5-6.0 × 106 CAR T cells/kg) groups was 54.5% and 71.4%, respectively.
In the CR group, 15 (93.7%) patients achieved minimal residual disease (MRD) negativity
(test sensitivity >1/10-5). All seven patients with double-hit or triple-hit multiple
myeloma achieved MRD-negative CR.
Summary/Conclusion: Our findings demonstrate the manageable safety profile and potential
efficacy of C-CAR088.
P945: SUBCUTANEOUS ISATUXIMAB ADMINISTRATION BY AN ON-BODY DELIVERY SYSTEM IN COMBINATION
WITH POMALIDOMIDE-DEXAMETHASONE IN RELAPSED/REFRACTORY MULTIPLE MYELOMA PATIENTS:
INTERIM PHASE 1B STUDY RESULTS
H. Quach1,*, G. Parmar2, E. M. Ocio3, H. M. Prince4, A. Oriol5, N. Tsukada6, K. Sunami7,
P. Bories8, C. Karanes9, S. Madan10, D. Semiond11, M. Inchauspe12, S. Macé13, P. B.
Musholt14, F. Suzan13, P. Moreau15
1Clinical Haematology Service, St Vincent’s Hospital, University of Melbourne, Vic;
2Illawarra Cancer Care Centre, Wollongong, NSW, Australia; 3Hospital Universitario
Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, Santander, Spain; 4Molecular
Oncology and Cancer Immunology, Epworth Healthcare and University of Melbourne, Melbourne,
Vic, Australia; 5Institut Català d’Oncologia and Institut Josep Carreras, Hospital
Germans Trias i Pujol, Barcelona, Spain; 6Department of Hematology, Japanese Red Cross
Medical Center, Tokyo; 7National Hospital Organization Okayama Medical Center, Okayama,
Japan; 8Early Phase Unit, Institut Claudius Regaud, Institut Universitaire du Cancer
Toulouse, Toulouse, France; 9Department of Hematology/Hematopoietic Cell Transplantation,
City of Hope National Medical Center, Duarte, CA; 10Banner MD Anderson Cancer Center,
Gilbert, AZ; 11Sanofi, Cambridge, MA, United States of America; 12IT&M Stats for Sanofi,
Neuilly sur-Seine; 13Sanofi Research & Development, Chilly-Mazarin, France; 14Sanofi
Research & Development, Frankfurt, Germany; 15Department of Hematology, University
Hospital of Nantes, Nantes, France
Background: Intravenous (IV) isatuximab (Isa) + pomalidomide-dexamethasone (Pd) is
approved for the treatment of relapsed/refractory multiple myeloma (RRMM) patients
(pts). Subcutaneous (SC) delivery would optimize convenience of administration. Prior
results showed that SC Isa administered by syringe pump has efficacy and safety profiles
comparable to IV Isa; the recommended Phase 2 dose (RP2D) was 1400 mg (IMW21 P-207).
Aims: This multicenter Phase 1b study evaluated safety, pharmacokinetics (PK), and
efficacy of SC vs IV Isa + Pd in RRMM pts after ≥2 prior treatment lines.
Methods: Pts were randomized 2:1 to SC1000 mg or IV 10 mg/kg and to SC1400 mg or IV.
An expansion cohort was later implemented with SC Isa administered at the RP2D via
on-body delivery system (OBDS), a wearable bolus injector applied to the abdomen by
a healthcare professional. Primary endpoints (EPs) were safety, including injection
site reactions (ISRs), and PK. Main secondary EPs were overall response rate (ORR)
and progression-free survival (PFS).
Results: Fifty-six pts were randomized and treated: 12 Isa IV, 12 Isa SC1000, 10 Isa
SC1400, and 22 OBDS pts. At study entry, ISS stage II–III was 67% in IV, 33% in SC1000,
60% in SC1400, and 50% in OBDS pts. On Jan 20, 2022, 33% IV, 25% SC1000, 50% SC1400,
and 86% OBDS pts remained on treatment. Due to sequential accrual, median follow-up
(FU) was longer in IV (20.6 mo) and SC1000 (23.8 mo) than SC1400 (18.1 mo) and OBDS
(6.5 mo) pts. Infusion reactions (IRs) were infrequent (≤10% in each cohort, all Grade
[G] 2), only at first IV or SC infusion/injection, with no IRs in OBDS. Local tolerability
of OBDS was very good, with 5 (22.7%) pts experiencing 7 ISR episodes, all G1, out
of 305 administrations (2.3%): 5 injection site erythemas, 1 injection site hemorrhage,
and 1 injection site induration. Median duration of OBDS injection was 10 min. Incidence
of G4 neutropenia was lower in SC1000 (42%) vs the other cohorts (55–60%). The lower
percentage of ≥G3 treatment-related adverse events in OBDS (77%) vs the other cohorts
(≥80%) may be due to shorter FU. Best overall responses are shown in table; longer
FU is needed for OBDS pts. Median PFS was 22 mo in IV, 12.5 mo in SC1000, and not
reached in SC1400 and OBDS pts. PK and CD38 receptor occupancy results in OBDS pts
are consistent with those observed in SC1400 with pump.
Image:
Summary/Conclusion: SC Isa administered by OBDS shows a safety profile consistent
with IV administration with no IRs and excellent local tolerability. Efficacy in the
SC cohorts was comparable to the Phase 3 ICARIA results. Isa SC administration by
OBDS is well tolerated, requires a short duration of injection, and provides a convenient
hands-free option.
Clinical trial information: NCT04045795. Research Sponsor: Sanofi.
P946: A PHASE I/II SINGLE ARM STUDY OF BELANTAMAB MAFODOTIN, CARFILZOMIB AND DEXAMETHASONE
IN PATIENTS WITH RELAPSED MULTIPLE MYELOMA: AMARC 19-02 BELACARD STUDY.
M. Lasica1,2,*, A. Spencer2,3,4, P. Campbell2,5,6, C. Wallington-Gates2,7,8, N. Wong
Doo2,9,10, W. Janowski2,11, G. McCaughan2,12,13, A. Puliyayil2,14, F. Yuen2,3, K.
Le2,3, J. Reynolds2,3,4, H. Quach1,2,15
1Department of Haematology, St Vincent’s Hospital Melbourne, Fitzroy; 2Australasian
Myeloma Research Consortium; 3Department of Haematology, Alfred Hospital, Melbourne;
4Faculty of Medicine, Monash University, Clayton; 5Department of Haematology, Barwon
Health, Geelong; 6Deakin University, Burwood; 7Flinders University; 8Department of
Haematology, Flinders Medical Centre, Bedford Park; 9Department of Haematology, Concord
Hospital, Concord; 10University of Sydney, Camperdown; 11Department of Haematology,
Calvary Mater Newcastle, Newcastle; 12Department of Haematology, St Vincent’s Hospital
Sydney; 13St Vincent’s Clinical School, University of New South Wales, Sydney; 14Department
of Haematology, Border Cancer Hospital, Albury; 15University of Melbourne, Parkville,
Australia
Background: Belantamab Mafodotin (Belamaf), a first in class anti- B-cell maturation
antigen (BCMA) antibody-drug conjugate is efficacious in patients with triple-class
exposed/refractory multiple myeloma (RRMM). Combining Belamaf (B) with carfilzomib
and dexamethasone (Kd) is potentially synergistic through direct myeloma-cell kill
and immune response against myeloma.
Aims: To characterize the safety, tolerability, and preliminary efficacy of BelaMaf
in combination with carfilzomib and dexamethasone (Kd) in patients with early relapsed
MM.
Methods: BelaCarD is an ongoing, two-part, single-arm, multicentre phase I/II study
evaluating an extended schedule of B every 8 weeks in combination with Kd in patients
with RRMM after 1-3 prior lines of treatment. Prior refractoriness to proteasome inhibitors
was allowed. Here we report a pre-planned analysis of the safety run-in phase of the
first 10 patients who have completed at least 1 treatment-cycle. Belamaf (2.5mg/kg)
was administered on day (D) 1 of every 2nd 28-day cycle, K 70mg/m2 iv D1 (20mg/m2
on C1D1), D8 and D15 of every cycle and dexamethasone 40mg weekly (20mg for patients
>75 years). Treatment was continued until disease progression. Adverse events (AEs)
were graded per CTCAEv4, except for corneal AEs which were graded by the pre-specified
keratopathy and visual acuity (KVA) scale. Response was assessed by the International
Myeloma Working Group (IMWG) criteria.
Results: At cut-off (Feb 3rd 2022), 19 patients had received B-Kd. The median age
of the 10 safety run-in patients was 65 years (range, 48-77); One, five and four patients
had 3, 2 and 1 prior lines of therapy respectively including (exposed/refractory %)
bortezomib (100/30%), carfilzomib (10/0%) lenalidomide (60/50%), pomalidomide (10/10%),
ASCT (70/0%), anti-CD38 monoclonal Ab (mAb) (40/40%). The median number of treatment-cycles
was 7 (5-11). The median number of cycles commenced was 9 (range, 2-13). The most
frequent AE during cycle 1 was thrombocytopenia (all grade 30%, Gr 3/4 20%) and blurred
vision (all grade 20%, Gr 3/4 0%) One patient experienced Gr 4 neutropenia. From cycle
2 onwards, the most frequent AEs included blurred vision (all grade 40%, Gr 3/4 20%),
peripheral neuropathy (all grade 30%, Gr 3/4 10%), upper respiratory tract infection
(all grade 30%, Gr 3/4 20%), dry eyes (all grade 20%, Gr 3/4 0%), neutropenia (all
grade 20%, Gr 3/4 10%) and nausea (all grade 20%, Gr 3/4 0%). Six patients had an
SAE, one was related to Belamaf (Gr 1 infusion reaction). Keratopathy occurred in
8 patients; grade 1, 20%; grade 2, 0%; grade 3, 60%. Decline in best corrected visual
acuity (BCVA) by at least 2 lines occurred in 8 patients (Gr 3 n=6, Gr 2 n=2). One
patient discontinued therapy due to corneal toxicity. Two patients died (progressive
disease n=1, unrelated cause n=1).
Of the 10 patients in the safety run-in, 9 achieved a PR or better (CR=3, VGPR=3,
PR=3) and 2 are known to have subsequently progressed (Figure 1). At estimated median
potential follow-up of 9.95 months, median PFS had not been reached (95%CI: 1.08 –NR).
Image:
Summary/Conclusion: B-Kd with an extended B schedule, has a safety profile that is
in keeping with that expected for each individual drug. Deep responses were seen.
Recruitment is ongoing in an expansion phase based on the preliminary safety and efficacy.
P947: COMPARATIVE EFFECTIVENESS OF ORAL IXAZOMIB-LENALIDOMIDE-DEXAMETHASONE (IRD)
AFTER INITIAL BORTEZOMIB (V)-BASED INDUCTION VS PARENTERAL V-BASED THERAPY IN NEWLY
DIAGNOSED MULTIPLE MYELOMA (NDMM)
R. M. Rifkin1,*, C. L. Costello2, R. E. Birhiray3, S. Kambhampati4, J. Richter5, R.
Abonour6, H. C. Lee7, Y. J. Kim8, K. Ren9, D. M. Stull10, D. Cherepanov9, K. Bogard10,
S. J. Noga9, S. Girnius11
1Rocky Mountain Cancer Centers/US Oncology Research, Denver; 2Moores Cancer Center,
University of California San Diego, La Jolla; 3Hematology Oncology of Indiana/American
Oncology Network, Indianapolis; 4Kansas City Veterans Affairs Medical Center, Kansas
City; 5Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York;
6Indiana University School of Medicine, Indianapolis; 7MD Anderson Cancer Center,
Houston, United States of America; 8Data Analytics, Evidera, St-Laurent, Canada; 9Takeda
Development Center Americas, Inc. (TDCA); 10Takeda Pharmaceuticals U.S.A., Inc., Lexington;
11TriHealth Cancer Institute, Cincinnati, United States of America
Background: Long-term proteasome inhibitor (PI)-based treatment can improve outcomes
for patients (pts) with multiple myeloma (MM). However, prolonged parenteral PI therapy
(e.g. with V) can be challenging to achieve in routine clinical practice, and outcomes
for pts are often poorer in this setting compared with clinical trials. The phase
IV, community-based, single-arm US MM-6 study (NCT03173092) is assessing in-class
transition (iCT) from V-based induction to all-oral IRd in transplant ineligible NDMM
pts treated in routine clinical practice, with the objective of increasing the duration
of PI-based treatment while maintaining quality of life. INSIGHT MM is the largest
global, prospective, observational study of MM pts (>4,200), and provided a subset
of patients as the comparator cohort. This enabled assessment of iCT vs V-based therapy
in NDMM pts in routine clinical practice in the US.
Aims: To examine the comparative effectiveness of IRd following initial V-based induction
(3 cycles; US MM-6 pts; ‘IRd’ cohort) vs continued V-based therapy (INSIGHT MM pts;
‘V-based’ cohort) in NDMM pts.
Methods: A secondary analysis of non-transplant eligible US NDMM pts with ≥stable
disease after 3 cycles of V-based induction and baseline Eastern Cooperative Oncology
Group performance status (ECOG PS) of 0, 1 or 2 from the US MM-6 (Manda CLML 2020)
and INSIGHT MM (Costello Future Onc 2019) studies was performed. Study outcomes included
first-line duration of treatment (DOT), overall response rate (ORR), progression-free
survival (PFS), overall survival (OS), and reasons for treatment discontinuation.
All analyses were weighted using the inverse probability of treatment weighting (IPTW)
approach to reduce the imbalance of potential confounding factors between the two
cohorts (adjusted analyses). Kaplan–Meier methods were used to examine DOT, PFS, OS,
and associated 95% confidence intervals (CIs); the log-rank test was used to compare
distribution of time to events. The Clopper-Pearson method was applied to estimate
95% CIs for ORR. Statistical significance was evaluated at alpha=0.05.
Results: 100 pts from the IRd cohort (MM-6) and 111 pts from the V-based cohort (INSIGHT)
were included. After IPTW, in the IRd vs V-based cohorts: median age was 75.0 vs 74.8
yrs; 56.7 vs 51.3% of pts were male; 37.4 vs 29.1% had an ECOG PS of ≥2; 48.8 vs 41.4%
had International Staging System stage III at initial diagnosis, and 79.5/17.7/2.8
vs 77.3/19.5/3.1% pts had received VRd/ V-cyclophosphamide-d (VCd)/ VRCd as initial
induction therapy.
Adjusted ORRs in the IRd vs V-based cohorts were 74.1 (95% CI 66.0–82.2) vs 57.5%
(95% CI 47.9–67.1; p<0.0001). After a median follow-up of 20.3 and 15.8 months in
the IRd and V-based cohorts, respectively, DOT was 10.8 (95% CI 6.5–24.4) vs 5.3 months
(95% CI 4.3–7.0; p<0.0001) (see Figure). Median PFS was not estimable (NE) in either
cohort; 24-month PFS rates were 85.7 (95% CI 68.1–94.0; IRd cohort) vs 76.5% (95%
CI 62.6–85.8; V-based cohort). Median OS was NE in either cohort; 24-month OS rates
were 94.0 (95% CI 77.7–98.5; IRd cohort) vs 84.9% (95% CI 70.6–92.6; V-based cohort).
In the IRd and V-based cohorts, 16.8 and 16.9% of pts discontinued IRd and V, respectively,
due to an adverse event.
Image:
Summary/Conclusion: US MM-6 NDMM pts who transitioned to IRd after 3 initial cycles
of V-based induction had a significantly higher ORR and longer DOT compared with pts
who received continued V-based therapy in INSIGHT MM. The results suggest that iCT
from continued V-based therapy to all-oral IRd may improve outcomes in pts treated
at community oncology clinics.
P948: INCIDENCE AND CLINICAL OUTCOME OF SARS-COV-2 INFECTION AFTER VACCINATION IN
PATIENTS WITH MONOCLONAL GAMMOPATHY OF UNDETERMINED SIGNIFICANCE (MGUS)
N. Sgherza1,*, P. Curci1, R. Rizzi1,2, D. Roccotelli2, M. Croce2, M. Avantaggiato2,
L. Ruga2, A. Vitucci1, A. Palma1, D. Di Gennaro2, P. Musto1,2
1Hematology and Bone Marrow Transplantation Unit, AOUC Policlinico, Bari, Italy; 2Department
of Emergency and Organ Transplantation, “Aldo Moro” University School of Medicine,
Bari, Italy
Background: Our group recently reported that MGUS patients investigated during the
early waves of pandemic, when vaccines were still not available, neither have an increased
risk of contracting SARS-CoV-2, infection, nor show poorer COVID-19 outcomes with
respect to healthy controls (Sgherza N. et al. Haematologica. 2022).
Aims: Aiming to specifically address the clinical effects of vaccines, we compared
incidence and outcome of SARS-CoV-2 infection of 1,454 previously described, not vaccinated
MGUS patients (91 of whom were SARS-CoV-2 positive), with those observed in a similar
population during the national vaccination campaign.
Methods: We obtained retrospective information from 86 individuals found to be SARS-CoV-2
positive among 1,265 MGUS patients analyzed after at least two doses of anti-SARS-CoV-2
vaccine received between April 2021, and January, 2022. Seventy-one subiects positive
before vaccination or after only one dose were escluded from analysis.
Results: The mean age of this group was 65.7 +/- 13.3 years (range 31-90); 32 patients
were female (37%), and 54 were male (63%). About MGUS-subtypes, the most frequent
one was IgG-kappa (n=35; 40.7%), followed by IgG-lambda (n=28; 32.5%), IgM-kappa (n=11;
12.8%) and others (n=12; 14%). Most of patients (81/46, 94.2%) were at low or low-intermediate
risk, according to Mayo Clinic prognostic model. Thirty-seven (43%) patients developed
SARS-CoV-2 infection after two doses (9, 26 and 2 patients receiving ChAdOx1-S, BNT162b2
mRNA and mRNA-1273 vaccines, respectively), fourty-nine (57%) after three doses (BNT162b2
mRNA or mRNA-1273 as “booster” dose, repectively). The mean number of days between
last dose of vaccine and SARS-CoV-2 infection was 103.04 +/-80.28 (range 2-285). The
two populations of SARS-CoV2 positive MGUS patients (before and after vaccination)
were comparable for age, sex and presence of co-morbidities (data not shown). Overall,
rates of symptoms (59.3% vs 26.7%), hospitalization (20.9% vs 2.3%), hospitalization
in Intensive Care Unit (11% vs 1.2%) and rate of deaths (8.8% vs 1.2%) were significantly
higher in still not vaccinated MGUS patients than in those who had received vaccines
(Table 1).
Image:
Summary/Conclusion: Our data indicate that the incidence of SARS-CoV-2 infection is
not significantly reduced in fully vaccinated MGUS patients (even after three doses),
likely also because of the higher diffusion capacity of the recently recognized Omicron
viral variants. However, as observed in normal population and in other hematological
contexts, the clinical outcome of COVID-19 may be significantly improved after vaccination
in these patients, with symptoms present in less of one third of subjects and no case
of hospitalization or death in our series. These observations support extensive vaccination
programs in patients with MGUS.
P949: A PHASE 2 TRIAL OF ELRANATAMAB, A B-CELL MATURATION ANTIGEN (BCMA)-CD3 BISPECIFIC
ANTIBODY, IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA: INITIAL SAFETY RESULTS
FOR MAGNETISMM-3
A. M. Lesokhin1,*, B. Arnulf2, R. Niesvizky3, M. Mohty4, N. J. Bahlis5, M. H. Tomasson6,
P. Rodrίguez-Otero7, H. Quach8, N. S. Raje9, S. Iida10, M.-S. Raab11, A. Czibere12,
S. Sullivan12, E. Leip12, A. Viqueira13, X. Leleu14
1Division of Hematology and Oncology, Memorial Sloan Kettering Cancer Center/Weill
Cornell Medical College, New York, United States of America; 2Hôpital Saint-Louis,
Paris, France; 3Weill Cornell Medical College - New York Presbyterian Hospital, New
York, United States of America; 4Sorbonne University, Hôpital Saint-Antoine, and INSERM
UMRs938, Paris, France; 5Arnie Charbonneau Cancer Institute, University of Calgary,
Calgary, Canada; 6Holden Comprehensive Cancer Center, University of Iowa, Iowa City,
United States of America; 7Clinica Universidad de Navarra, Madrid, Spain; 8University
of Melbourne, Melbourne, Australia; 9Massachusetts General Hospital Cancer Center,
Harvard Medical School, Boston, United States of America; 10Department of Hematology
& Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan;
11Heidelberg Myeloma Center, Department of Hematology/Oncology, Heidelberg University
Hospital, Heidelberg, Germany; 12Pfizer Inc, Cambridge, United States of America;
13Pfizer SLU, Madrid, Spain; 14Centre Hospitalier Universitaire de Poitiers, Poitiers,
France
Background: Elranatamab (PF-06863135) is a humanized bispecific antibody that targets
both BCMA-expressing multiple myeloma (MM) cells and CD3-expressing T cells. MagnetisMM-3
(NCT04649359) is an open-label, multicenter, non-randomized, phase 2 study to evaluate
the safety and efficacy of elranatamab monotherapy in patients with relapsed/refractory
(R/R) MM.
Aims: Initial safety results from the MagnetisMM-3 trial in patients with R/R MM and
no prior BCMA-targeted treatment are presented.
Methods: MagnetisMM-3 enrolled patients who are refractory to at least 1 proteasome
inhibitor, 1 immunomodulatory drug, and 1 anti-CD38 antibody. Patients were assigned
to 1 of 2 independent, parallel cohorts: those naïve to BCMA-directed therapies (Cohort
A) and those with previous exposure to BCMA-directed antibody-drug conjugates or CAR-T
cells (Cohort B). Patients received subcutaneous elranatamab 76 mg QW on a 28-day
cycle with a 2-step-up priming dose regimen administered during the first week. Dose
modifications were permitted for toxicity. Treatment-emergent adverse events (TEAEs)
were graded by CTCAE (v5.0), and cytokine release syndrome (CRS) and immune effector
cell-associated neurotoxicity syndrome (ICANS) by ASTCT criteria.
Results: As of the data cutoff on Dec 31, 2021, 60 patients in Cohort A had received
≥1 dose of elranatamab; the last patient’s first dose was ~2 months prior to the cutoff.
Median age was 69.0 years (range, 44−89), 48.3% were male, 63.3% were white, 18.3%
were Asian and 11.7% were Black/African American. At baseline, 60.0% of patients had
an ECOG performance status 1−2 and patients had received a median of 5 (range, 2−12)
prior therapies. Median duration of elranatamab treatment was 9.57 weeks (range, 0.1−46.1);
median relative dose intensity was 87.4% (range, 23.1−101.4).
TEAEs were reported in 100% (Grade [G] 3/4, 75.0%) of patients. Most common (≥30%)
hematologic TEAEs were neutropenia (36.7% [G3/4, 35.0%]), anemia (36.7% [G3/4, 30.0%])
and thrombocytopenia (30.0% [G3/4, 21.7%]). Among patients who received the 2-step-up
priming regimen (n=56), CRS and ICANS, respectively, were reported in 58.9% (G3/4,
0%) and 3.6% (G3/4, 0%); of those patients, 57.6% (n=19/33) and 100% (n=2/2) received
tocilizumab and/or steroids. Most common (≥30%) non-hematologic TEAE, other than CRS/ICANS,
was fatigue (31.7% [G3/4, 3.3%]). Infections were reported in 46.7% (G3/4, 18.3%)
of patients; most frequently reported were upper respiratory tract infections (11.7%
[G3/4, 0%]). Discontinuations due to adverse events were reported in 5.0% of patients.
No patients permanently discontinued treatment due to CRS or ICANS. There were 10
deaths; causes were MM progression (n=8), septic shock (n=1) and unknown (n=1). Data
will be updated at the time of presentation to include ~90 patients.
Summary/Conclusion: Preliminary results of MagnetisMM-3 in patients with R/R MM and
no prior BCMA-targeted treatment suggest that 76 mg QW elranatamab with a 2-step-up
priming regimen is well tolerated, with no G ≥3 CRS or ICANS observed.
P950: COMBINED PROTEASOME AND AUTOPHAGY INHIBITION IN RELAPSED/REFRACTORY MULTIPLE
MYELOMA – A PHASE I TRIAL OF HYDROXYCHLOROQUINE, CARFILZOMIB AND DEXAMETHASONE
T. S. Slørdahl1,2,*, F. B. Askeland3,4, M. S. S. Hanssen5, S. M. Sundt-Hansen1, N.
T. Vethe6, H. Hjorth-Hansen1,2, M. H. Fenstad7, A. Waage2, Ø. Hjertner1, F. Schjesvold3,4,
A. Sundan2
1Department of Hematology, St. Olavs Hospital; 2Department of Clinical and Molecular
Medicine, Norwegian University of Science and Technology (NTNU), Trondheim; 3Oslo
Myeloma Center, Department of Hematology, Oslo University Hospital; 4KG Jebsen Center
for B Cell Malignancies, University of Oslo, Oslo; 5Department of Ophthalmology, St.
Olavs Hospital, Trondheim; 6Department of Pharmacology, Oslo University Hospital,
Oslo; 7Department of Immunology and Transfusion Medicine, St. Olavs Hospital, Trondheim,
Norway
Background: Multiple myeloma (MM) is characterized by malignant cells which produce
large amounts of monoclonal immunoglobulin. MM cells are highly sensitive to drugs
that target protein degradation. There are two important intracellular pathways for
protein degradation (Lamark, 2010). The proteasome is responsible for the degradation
of poly-ubiquitinated damaged, modified, misfolded and redundant proteins (Jung, 2009).
This pathway of degradation is the target of proteasome inhibitors. The other main
pathway is that of autophagy and lysosomal degradation, eliminating protein aggregates
and damaged organelles. Hydroxychloroquine (HCQ) is an inhibitor of autophagy. In
vitro studies have shown that HCQ potentiates carfilzomib (K) toxicity towards myeloma
cells and could therefore be an attractive treatment option in myeloma (Baranowska,
2016). Here we present safety and efficacy data from a phase 1 study on the combination
of HCQ, K and dexamethasone (d).
Aims: This study aimed to answer the question whether combined inhibition of the proteasome
and the autophagosome is safe and tolerable. The primary endpoint was to establish
a maximum tolerated dose (MTD) of the combination HCQ-Kd.
Methods: This phase I, single arm, open label dose escalation study (3 + 3 design),
included patients with relapsed and/or refractory MM with at least two prior lines
of therapy including bortezomib and an immunomodulatory agent. All patients started
a 14-day run-in of HCQ at their intended HCQ dose level (DL) before the first cycle
of HCQ-Kd. The HCQ DL were 200 mg (DL1), 400 mg (DL2), 600 mg (DL3), and 800 mg (DL4&5)
daily. Intravenous carfilzomib was given in a dose of 20 mg/m2 in cycle 1 day 1, and
thereafter 56 mg/m2 (DL1-4) and 70 mg/m2 (DL5) on day 8, 15, and in all subsequent
cycles and doses. Oral dexamethasone 40 mg (20 mg pts >75y) was administered on day
1, 8, 15 and 22. Patients were treated with HCQ-Kd for a maximum of six 28-day cycles
and continued Kd if still responding thereafter. The dose limiting toxicity (DLT)
observation period was 28 days. Adverse events (AE) grade (G) 1-5 were registered
from day 1 of run-in cycle and until 90 days post last dose of HCQ.
Results: The clinical trial is completed, and we here report the data for MTD, safety
and efficacy. 19 patients were included. One patient withdrew consent prior to first
treatment cycle and was therefore not included in MTD and efficacy analyses. Median
previous lines of therapy were 4 (2-9), 50% had high-risk cytogenetics. Only one DLT
occurred at DL2 (G3 pneumonia), and MTD was not reached. 104 AEs were recorded, of
which G1 45%, G2 32%, G3 22% and G4 1%. No deaths occurred. Most common AEs were anemia
(74%), neutropenia (42%), thrombocytopenia (19%), skeletal pain (32%), respiratory
tract infections (26%), insomnia (26%) and vomiting/nausea (21%). A total of 13 serious
adverse events (SAEs) occurred during the study of which 2 led to treatment discontinuation.
Responses were seen in 8 of 18 (ORR 44 %) including 3 PR (18%), 3 VGPR (18%), 1 CR
(6%), 1 sCR (6%). Clinical benefit rate was 78 % when including 6 MR (33%) (figure
1). One patient reached MRD negativity. 7 patients did not complete all 6 cycles of
HCQ-Kd, 5 due to PD and 2 due to SAEs.
Image:
Summary/Conclusion: HCQ given up to 800 mg daily in combination with weekly carfilzomib
and dexamethasone is well tolerated in patients with relapsed/refractory multiple
myeloma. Adverse events were mostly grade 1 and 2. The study results indicate a meaningful
clinical efficacy of the combination.
P951: CLINICAL AND LABORATORY CHARACTERISTICS, COURSE AND OUTCOME OF COVID19 INFECTION
IN PATIENTS WITH MULTIPLE MYELOMA
A. Sretenovic1,*, M. Mitrovic1, T. Adzic Vukicevic2, Z. Bukumiric3, O. Markovic4,
M. Zdravkovic4, D. Antic5, J. Bila1
1Myeloma department, Clinic of hematology University Clinical centre of Serbia; 2Pulmology,
COVID Hospital “Batajnica”, University Clinical Center of Serbia; 3Medical Faculty,
University of Belgrade; 4Hematology, Clinical-Hospital Center “Bezanijska Kosa”; 5Lymphoma
department, Clinic of hematology University Clinical centre of Serbia, Belgrade, Serbia
Background: During the Covid19 pandemic, patients with multiple myeloma (MM) are particularly
vulnerable due to the characteristics of their disease.
Aims: The aim of study was to analyze clinical and laboratory characteristics, course
and outcome of Covid19 infection in patients with multiple myeloma.
Methods: The study included 53 patients with MM and Covid 19 infection, diagnosed
during period March 2020 - November 2021 (27 male; 26 female, mean age 62 yrs, range
37- 87 yrs). IgG MM was present in 28pts (53%), IgA in 8 (15%), IgM in 3pts (6%),
and BJ in 14 (26%). According to the clinical stage (CS, Durie-Salmon), distribution
was as follows: III 44pts (83%); II 4pts (8%); I CS 5pts (9%). Renal impairement existed
in 18pts (34%). Regarding ISS score, the group included: ISS1 had 18pts (34%), ISS2
12pts (23%) and 23pts (43%) had ISS3. According to the Revised ISS (R-ISS) score,
R-ISS1 was found in 9pts (17%), R-ISS2 in 20pts (58%) and R-ISS3 was present in 13pts
(25%). All pts were treated according to National Protocol for the Treatment of Covid19
infection, including: antibiotics in 53pts (100%), corticosteroids in 50pts (94%),
low molecular weight heparin in 52pts (98%), and intravenous immunoglobulins in 26pts
(49%). During period January 2021 - November 2021, 9 of 24pts (37.5%) were vaccinated
against Covid19. Variables of importance were analyzed using descriptive and analytical
statistics. All calculations were made in SPSS program version 26.0.
Results: At the moment of detected Covid19 infection, 37pts (70%) had active MM, and
16pts (30%) were in the state of follow-up during remission of disease. Immunoparesis
was present in all of 37pts (74%) with active MM. Elevated interleukin-6 (IL-6) was
found in 19pts (36%), of which 15pts (78.9%) had active MM. Similarly, elevated d-dimer
was found in 46pts (87%), of which 35pts had active disease. During the course of
Covid19 infection, pneumonia was registered in 52pts (98%), bleeding in 2pts (4%)
and thrombosis in 4pts (8%). Bleeding and thrombosis were registered in patients with
active MM. In the Intensive Care Units were treated 7pts (13%), with lethal outcome
of 6pts. In the Semi-Intensive Care were treated 36pts (64%). A total of 36pts (64%)
were cured and 17pts (36%) died. The average time to recovery of our pts was 14 days
(max up to 45 days). After recovery 20pts (55%) had active myeloma, reinfection was
found in 5pts (13.5%) and they were in active phase of disease. All of 5pts (10%),
who were previously vaccinated against Covid19 during MM remission, completely recovered
from Covid19 infection. Additionally, 4pts (8%) were vaccinated after recovery from
Covid19 infection.
Patients with active MM had signficantly worse outcome of Covid19 infection, (Chi-Square,
p=0.003). Immunoparesis was highly frequent in pts with acitive disease (Fisher Exact
Test, p=0.000), as well as elevated d-dimer (Fisher Exact Test, p=0.002). Increased
IL-6 was observed in pts with active MM, but statistical significance was not established
(Chi-Square p=0.088). The poor treatment outcome of pts with MM and Covid 19 was influenced
by: Age (T-Test, p=0.022), Renal impairement (Chi-Square, p=0.032), and high R-ISS
score 3 (Mann Whitney, p=0.042).
Summary/Conclusion: A significantly worse outcome of Covid19 infection may be expected
in patients with active MM and renal impairement, accompanied with findings of elevated
d-dimer and IL-6, as well as immunoparesis and R-ISS score 3. All vaccinated patients
recovered from Covid 19 infection.
P952: THE ROLE OF AGE IN THE LONG-TERM TREATMENT OF MULTIPLE MYELOMA - A LONGITUDINAL
ANALYSIS OF REGULAR CARE DATA
H. T. Steinmetz1,*, M. Heinz1, U. Totzke2
1Onkologie Köln, Outpatient Clinic for Hematology and Oncology, Cologne, Cologne,
Germany; 2TOTZKE & DREHER SCIENTIFIC SA, Basel, Switzerland
Background: Recently, we demonstrated in a longitudinal approach that the percentage
of multiple myeloma (MM) patients reaching consecutive treatment lines in regular
care is much higher than presumed, i.e. 88%, 66%, 44%, and 30% in 2nd, 3rd, 4th, and
5th line, respectively (Oncol Res Treat 44, 662-71).
Aims: As the annual risk of death generally increases with age, we investigated here
the impact of age on the course of MM treatment.
Methods: MM patients treated in our outpatient clinic in Germany over 8 years were
assigned to three groups of equal size according to age at diagnosis. Resulting subgroups
were analyzed regarding their outcome and the probabilities of reaching consecutive
therapy lines using a linear statistical model, as previously described for the analysis
of the whole cohort.
Results: Age groups were <65, 65-74, and ≥75 years. As compared to the two remaining
groups, the proportion of male patients and of those with ISS 1 was considerably higher
in the youngest age group while the median observation time was on average 7 months
shorter in the oldest age group (Tab.).
Age <65 years
Age 65-74 years
Age ≥75 years
Patientsb
50 (34.5%)
46 (31.7%)
49 (33.8%)
womenb
18 (36.0%)
25 (54.3%)
23 (46.9%)
Time observed [month]a
40.5 (0-198)
40 (2-152)
33 (0-197)
Stage at diagnosisb
ISS
R-ISS
ISS
R-ISS
ISS
R-ISS
I
19 (38%)
11 (22%)
6 (13%)
9 (20%)
10 (20%)
3 (6%)
II
11 (22%)
13 (26%)
21 (46%)
17 (37%)
17 (35%)
16 (33%)
III
11 (22%)
6 (12%)
13 (28%)
2 (4%)
16 (33%)
6 (12%)
Unknown
9 (18%)
20 (40%)
6 (13%)
18 (39%)
6 (12%)
24 (49%)
Deathb
12 (24.0%)
8 (17.4%)
27 (55.1%)
Cause of deathb
MM
3 (25.0%)
1 (12.5%)
10 (37.0%)
Other
5 (41.7%)
6 (75.0%)
13 (48.1%)
Unknown
4 (33.3%)
1 (12.5%)
4 (14.8%)
a median (range); b N (%)
The probability of reaching consecutive treatment lines decreased in all age groups,
but most strikingly in that of the oldest patients (Fig.). Age does not appear to
play a major role for reaching subsequent lines at ages up to 75 years though. Up
to this age, patients were thus even more likely to reach subsequent lines than previously
shown for the total cohort, increasing to about a 50% chance for the 5th line of therapy.
The mean age at death was 83 (SD 4.8) years in the oldest age group, but in none of
the age groups, the leading cause of death was MM.
Image:
Summary/Conclusion: Only age beyond 75 years at baseline significantly reduces the
probability of reaching 2nd or higher lines of MM therapy.
P953: PHASE I STUDY OF TQB3602 CAPSULE, AN ORAL PROTEASOME INHIBITOR, IN RELAPSED/REFRACTORY
MULTIPLE MYELOMA
W. Tang1, Y. Li1, X. Zhong1, Q. Liang1, Y. Liu2, Y. Zeng3, B. Fang2, L. Zheng4, T.
Niu1,*
1Department of Hematology, West China Hospital, Sichuan University, Chengdu; 2Department
of Hematology, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University,
Zhengzhou; 3Department of Hematology, First Affiliated Hospital of Kunming Medical
University, Hematology Research Center of Yunnan Province, Kunming; 4GCP Center/National
Institute of Drug Clinical Trial, West China Hospital, Sichuan University, Chengdu,
China
Background: The proteasome inhibitor is an effective treatment strategy for multiple
myeloma (MM). TQB3602 is a novel oral proteasome inhibitor derived from multiple rounds
of optimization based on ixazomib. It has an addition of an acridine ring to the structure
of ixazomib, which maintains high activity and selectivity, resulting in better metabolic
stability. In preclinical models of MM, TQB3602 displayed potent kinase inhibiting
activity for 20S proteasome enzyme with IC50 15.3 nM. It also inhibited cell proliferation
in the MM.1S cell line with IC50 10.1 nM. In the MM.1S MM CB-17 CDX model, TQB3602
also showed antitumor efficacy. TQB3602 represents a promising clinical candidate
for treating MM.
Aims: This study presents the first-in-human trial designed to evaluate the safety,
tolerance, pharmacokinetics, dose-limiting toxicity and maximum tolerated dose of
TQB3602 in relapsed/refractory MM (RRMM).
Methods: This study is a multicenter, open-label, 3 + 3 dose-escalation phase I trial
(NCT04275583). Adult patients with RRMM who had received at least two kinds of therapy,
including bortezomib, lenalidomide or thalidomide, are enrolled. TQB3602 is orally
administered at a dose of 0.5mg, 1mg, 2mg, 3mg, 4mg, 5mg, 5.5mg, 6mg or 7mg on days
1, 8, 15 in 28-day cycle until limiting toxicity or disease progression.
Results: Twenty-five patients with RRMM (16 males, 9 females, median age 65, range
37-73) were enrolled between May 2020 and October 2021, with all patients exposed
to bortezomib and three patients exposed to ixazomib. Patients received a median of
three cycles of TQB3602 (range 1-11). Grade ≥3 drug-related adverse events included
anemia (28%), thrombocytopenia (8%), neutropenia (8%) and diarrhea (4%). No grade
≥3 drug-related peripheral neuropathy occurred. Two dose-limiting toxicities occurred
at 7mg, including diarrhea and grade 2 peripheral neuropathy. The maximum tolerated
dose was determined as 6mg. TQB3602 was rapidly absorbed, resulting in a time to plasma
peak concentration from 0.8 to 1.5 hours. After multiple dosing, the terminal half-life
was 23 to 152 hours. Following administration of 6 mg, the mean half-life was approximately
82 hours. TQB3602 slightly accumulated in the human body. At a dose of 6 mg, the AUClast
and Cmax were approximately 1.9 times higher on day 15 than day 1. The mean plasma
concentration-time profiles of day 1 and day 15 are shown in Figures A and B, and
the geometric mean plasma Cmax and AUClast on day 1 and day 15 are shown in Figures
C and D. Among 25 response-evaluable patients, 65% of patients achieved stable disease
or better.
Image:
Summary/Conclusion: TQB3602 is well tolerated with no severe neuropathy adverse events
and has shown preliminary efficacy in patients with RRMM. Based on the efficacy, safety,
and pharmacokinetic data, 6mg was recommended as the anticipated therapeutic dose
and adopted in an ongoing dose-expansion phase.
P954: ESTABLISHING THE NATIONAL INSTITUTE FOR HEALTH RESEARCH (NIHR) HEALTH INFORMATICS
COLLABORATIVE (HIC) MULTIPLE MYELOMA (MM) REGISTRY
S. Tayabali1,*, S. Moore2, M. Jenner3, F. Djebbari2, L. Romao1,4, L. English1,4, R.
Rodriguez1,4, B. Briot Ribeyre1,4, G. Roadknight2,5, K. Varnai2,5, S. Little2,5, J.
Davies2,5, K. Woods2,5, C. Davis6, F. Borca6, J. Olza Meneses6, H. Phan7, W. K. Wong1,4,
R. Popat1
1University College London Hospitals NHS Foundation Trust, London; 2Oxford University
Hospitals NHS Foundation Trust, Oxford; 3University Hospital Southampton NHS Foundation
Trust, Southampton; 4NIHR University College London Hospitals Biomedical Research
Centre, London; 5NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford;
6NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS
Foundation Trust; 7NIHR Southampton Biomedical Research Centre, University of Southampton,
Southampton, United Kingdom
Background: Real world data provides unique insights into the natural history of MM,
resource utilisation, adherence to standards and outcomes of treatments outside clinical
trials. However, the quality of such initiatives can be poor due to the need for manual
data entry, inability to integrate multiple data sources, lack of harmonisation of
data across centres and limited generalisability due to centre bias. Exemplar registries
collate and transform multidisciplinary routinely collected data from Electronic Health
Records (EHRs) to a common data language allowing integration, analysis and expansion.
Aims:
To develop a secure national MM registry beginning with a three site pilot.
Methods:
Potential registry providers were evaluated against the following criteria: previous
experience, infrastructure and technical capability, data governance, compliance with
legal standards and cost effectiveness. The platform required ethical and data sharing
approvals. A common data model was investigated and a myeloma focused multidisciplinary
dataset was designed and mapped to source data. Data extraction, transformation and
analysis is planned.
Results:
8 providers were assessed for suitability. The NIHR HIC was chosen due to its established
data sharing network across 30 UK sites and overarching governance framework. This
platform permitted the establishment of harmonised data flow, comparable across sites
and retaining clinical interpretability. A minimal myeloma dataset of routinely collected
information was agreed by clinicians and data scientists to contain the following
fields: baseline and disease characteristics including imaging findings eg: extramedullary/paramedullary
disease, skeletal/spinal disease, comorbidities, mode of presentation, cytogenetics,
treatments including chemotherapy, stem cell transplants/CAR-T cells, spinal interventions,
anti-infectives, bisphosphonates and other supportive care. Each variable was mapped
to identify the location of raw data in the EHR, its format and suitability for extraction.
For unstructured data, eg: cytogenetics and imaging reports, natural language processing
is being developed to extract key fields.
To enable harmonisation of data from different systems, data is transformed into a
common data model. The Observational Medical Outcomes Partnership common data model
(OMOP CDM) was chosen as it provided a framework for systematic analysis of data from
disparate data sources, typical of those held within health care information systems.
This model allows for international benchmarking and collaboration for research. OMOP
CDM maps commonly used coding systems and vocabularies, eg ICD-10, SNOMED-CT into
standard concepts permitting raw data entering the registry to be standardised into
a research ready format that is person-centric, disease agnostic and syntactically
and semantically defined.
Anonymised data from multiple sites will be electronically transferred to a ISO27001
certified, GDPR compliant UCL data safe haven for analysis. Oversight is provided
by the Steering Committee of clinicians, data scientists, myeloma patients and Myeloma
UK with patient involvement at every step. Data will be analysed to answer 3 initial
themes (Image 1), and extended further to include control data for Health Technology
Appraisals. To date we have successfully identified an initial cohort of over 4000
myeloma cases since 2015.
Image:
Summary/Conclusion: The NIHR HIC Myeloma registry represents a research ready platform
to integrate and analyse UK wide real word data. Detailed data analysis and progress
in answering the initial 3 themes is ongoing.
Funding: Myeloma UK & NIHR
P955: CARFILZOMIB IN RELAPSED/REFRACTORY MULTIPLE MYELOMA PATIENTS: THE REAL-LIFE
EXPERIENCE OF EMMY
C. Hulin1,*, M. Macro2, A. Perrot3, B. Royer4, D. Caillot5, K. Belhadj6, L. Frenzel7,
R. Benramdane8, H. Demarquette9, B. Bareau10, S. Darre11, C. Calmettes12, R. Le Calloch13,
M. Bouketouche14, K. Laribi15, N. Texier16, M. Willaime16, C. Deal17, P. Moreau18,
O. Decaux19
1CHU Bordeaux, Bordeaux; 2CHU Caen, Caen; 3CHU de Toulouse, IUC T-O, Toulouse; 4bruno.royer@aphp.fr,
paris; 5CHU Dijon, Dijon; 6Hôpital Henri Mondor, Creteil; 7Hôpital Necker, paris;
8CH Cergy Pontoise, Cergy Pontoise; 9CH. Dunkerque, Dunkerque; 10CH Cesson Sévigné,
Cesson Sévigné; 11CH. Arras, Arras; 12CH. Périgueux, Périgueux; 13CH Cornouaille,
Quimper; 14CH. St Quentin, St. Quentin; 15CH. Le Mans, Le Mans; 16kappa santé; 17IFM,
paris; 18CHU Nantes, Nantes; 19CHU Rennes, Rennes, France
Background: EMMY is a large-scale epidemiological study to assess the epidemiology
and real-life management of multiple myeloma (MM). Proteasome inhibitors (PI), immunomodulators
(IMID) and anti-CD38 (aCD38) provide broad solutions to treat patients. Following
pivotal clinical trials, carfilzomib, a second-generation PI, is approved in relapsed/refractory
(RR) MM. Its use in real-life setting can be assessed in EMMY.
Aims: To assess the use of carfilzomib in RRMM patients in real-life conditions.
Methods: EMMY is a descriptive, multicenter, national, non-interventional study conducted
in 72 IFM (Intergroupe Francophone du Myélome, sponsor) sites in France. Any patient
initiating treatment for MM over a 3-month annual observation period, is included,
since 2017 This dynamic cohort has included approximately 900 patients each year (2765
patients included in 2019). Data are updated annually from hospital records. Patients
receiving carfilzomib (K) for RRMM at inclusion or follow up were identified and classified
by the regimens received. The median progression-free survival (mPFS), and median
overall survival (mOS) were estimated per regimen and per treatment line.
Results: 512 patients (18.5%) (median age 68.4 years (y), 20.5% ≥75y) received K for
RRMM, 111 (21.7%) for a second (L2), 97 (18.9%) a third (L3) and 304 (59.4%) a fourth
or more (L4+) line. K was used in a double regimen with dexamethasone (Kd) in 232
patients (45.3%) and in a triple regimen either with an IMID (K-IMID) in 198 (38.7%)
patients or with other agents in 82 patients (16%) (K-aCD38 for 35 (6.8%), K-alkylant
for 27 (5.3%) and K-venetoclax for 19 (3.7%) patients). 25.4% patients (62/244) were
at high cytogenetic risk.
K-IMID was used in patients of 66,8y, with an ECOG ≥ 2 for 20.8%, and comorbidities
(≥ 1) for 24.3% including 3% of cardiovascular diseases. K-IMID was used in L2 (42.9%),
L3 (21.7%) and L4+ (35.4%). 63.1% were priorly exposed to lenalidomide (len) of which
35,9% were refractory. K was combined with len in 144 (28,1%) and with pomalidomide
(pom) in 54 (10,5%) patients. The use of K-pom increased over years: 14,4% in 2018,
28,2% in 2019, 73,9% in 2020. mPFS was overall 10.4 months (m) 95%CI [7.3; 14.7];
it was 23m 95%CI [14.7; -] in L2, 6.2m 95%CI [1.3; 12.2] in L3 and 7.1m 95%CI [4.6;
9.2] in L4+. mOS was not reached in L2, L3 or L4+.
Kd was used in patients of 70.1y, with an ECOG ≥ 2 for 31.3% and comorbidities for
29.3%. 93.5% were priorly exposed to len of which 66.4% were refractory. Kd was used
in L2(6%), L3 (17.2%) and L4+ (76.8%). mPFS and mOS were 4.4m [3.7; 5.8] and 19.5m
[13.7; 22.3] in L4+.
K-aCD38 was used in patients of 63.5y, with an ECOG ≥ 2 for 10%, and comorbidities
(≥ 1) for 37.1%. 91.4% were priorly exposed to len including 45.7% len-refractory.
K-aCD38 (33/35 with daratumumab) was used in L2 (31.4%), L3/L4 (35.7%) and L5+ (22.9%).
mPFS and mOS (all lines included) were 6m [3.8; 15.1] and 21.3m [16.9; -].
K-alkylant (5/27 with bendamustine, 22/27 cyclophosphamide) and K-venetoclax were
used in older patients (71.8y and 70.4y) and in L4 or L5 for >80% of them. More than
90% were pre-exposed to len including 65% of len-refractory patients.
K discontinuation was recorded for 63.4% of patients for progression in 44% and adverse
event in 11.7% of them.
Summary/Conclusion: The EMMY study shows that carfilzomib is used in many combinations
in extended real-life settings compared to pivotal clinical trials including older
patients, largely-exposed to lenalidomide and at advanced disease. The use of lenalidomide
in early stages has led to a shift in the management of RRMM patients with carfilzomib.
P956: EMMY COHORT: MANAGEMENT OF NEWLY DIAGNOSED OR RELAPSED/ REFRACTORY MULTIPLE
MYELOMA IN PATIENTS AGED OF 80 YEARS AND OVER
M. Macro1,*, O. Decaux2, A. Perrot3, B. Royer4, M. Chretien5, K. Belhadj6, M. Mohty7,
L. Frenzel8, X. Leleu9, M. Dib10, O. Allangba11, P. Zunic12, I. Botoc13, J. Malfuson14,
P. Moreau15, R. Garlantazec16, N. Texier17, R. Germain17, C. Deal18, C. Hulin19
1CHU Caen, Caen; 2C.H.U de Rennes, Rennes; 3Institut Universitaire du Cancer Toulouse
Oncopole, Toulouse; 4hôpital st louis, aphp, Paris; 5CHU Dijon, Dijon; 6Hôpital Henri
Mondor, Creteil; 7Hôpital Saint-Antoine; 8Hôpital Necker, paris; 9CHU de Poitiers,
Potiers; 10CHU Angers, Angers; 11C.H. Yves le Foll, ST Brieuc; 12CHU Réunion Sud,
St Pierre; 13CH Saint-Malo, Saint Malo; 14Hôpital d’Instruction des Armées Percy,
Clamart; 15CHU nantes, Nantes; 16CHU Rennes, Rennes; 17kappa santé; 18IFM, Paris;
19CHU Bordeaux, Bordeaux, France
Background: Multiple myeloma (MM) patients aged 80 years and older are a population
more prone to comorbidities, frailty, cognitive impairment or physical decline and
require appropriate management of their myeloma. They are often underrepresented in
pivotal clinical trials, with little data available on their management. The EMMY
study is a large-scale epidemiological study to assess the epidemiology and real-life
management of MM and can focus on the use and real-life efficacy of treatments for
newly diagnosed or relapsed/ refractory elderly MM patients.
Aims: To describe the management of MM in newly diagnosed or relapsed patients aged
80 years and over and to assess the real-life effectiveness of the treatments received.
Methods: EMMY is a descriptive, multicenter, national, non-interventional study conducted
in 72 IFM (Intergroupe Francophone du Myélome, sponsor) centers in France. Any patient
initiating treatment for MM over a 3-month observation period, from October to December,
is included, since 2017. It is a dynamic cohort with the inclusion of approximately
900 patients each year: 2765 patients included at the end of 2019 of which 561 were
aged 80 and over (20.3%). Data are updated annually from hospital records up to 2020
at the time of the analysis.
Four cohorts of patients aged 80 years and older were analysed according to the line
of treatment initiated during their follow-up: L1, L2, L3 or L4+. Median time to next
treatment (mTNT), median progression-free survival (mPFS) and median overall survival
(mOS) were assessed in months(m).
Results: Patients ≥80 years initiated 19.8% of L1s (237/1199) 22.7% (232/1022) of
L2s, 22.2% (167/751) of L3s and 18.7% (288/1540) of L4+. They are 7.5% patients ≥85
years old to initiate L1, 8.7% L2, 7.4% L3 and 5.1% L4+. When the line is started,
an ECOG ≥2 is reported for 39.4% of L1, 27.7% of L2, 35.1% of L3 and 38.5% of L4 patients
and comorbidities recorded in 45.1% of L1, 39.6% of L2, 35.4% of L3 and 38.6% of L4+.
In L1, the ISS is level I, II, III for 20.2%, 27.4% and 52.4% of patients.
In L1, over the 2017-2020 study period, patients received treatment with PI (58.6%),
IMID (32.1%), PI/IMID (4.2%), anti-CD38 (1.3%) or other molecules (3.8%). In L2, they
received PI (22.8%), IMID (48.7%), PI/IMID (11.7%), anti-CD38 (14.2%) or other (2.6%),
in L3 PI (12.6%), IMID (38, 9%), PI/IMID (9.6%), anti-CD38 (29.3%) or other (9.6%)
and in L4+ a PI (23.3%), IMID (20.1%), PI/IMID (5.6%), anti-CD38 (29.2%) or other
(21.9%). More than half received a double combination in L1 (50.2%), L2 (59.9%), L3
(54.4%) and L4+ (53.8%).
Regarding efficacy, mTNT is 20.3m [15.2; 24.6] in L1, 14.7m [12.3; 20.4] in L2, 10.8m
[9; 13.5] in L3 and 8.2m [6.5; 11.2] in L4+. The mPFS is 19.3m [15.1; 23.2] in L1,
12.6m [10.1; 17.5] in L2, 8.9m [10.1; 17.5] in L3 and 5.9m [4.5;7.4] in L4+. The mOS
is not reached in L1 (rate at 30m: 76.7% [70; 83.3]) and L2 (rate at 30m: 58.1% [48.1;
68]; it is 25.7m [17.1; NE] in L3 and 18.3m [13.4; 22.4] in L4+.
Summary/Conclusion: Patients ≥80 years of age account for more than 20% of de novo
and relapsed myeloma patients treated in EMMY. Double combinations are used in half
of the situations. Over the study period, PI-based treatments were preferred in L1,
IMID-based in L2 and antiCD38 for 30% of patients in L3 and L4+. The benefits in terms
of survival are real and this specific population is likely to increase significantly
in the near future, requiring human and logistical resources.
P957: CARFILZOMIB, LENALIDOMIDE, DEXAMETHASONE FOLLOWED BY A SECOND AUTO-HCT IS AN
EFFECTIVE STRATEGY IN FIRST RELAPSE MULTIPLE MYELOMA: A STUDY OF THE CHRONIC MALIGNANCIES
WORKING PARTY OF EBMT
R. Tilmont1,*, I. Yakoub-Agha1,2, D.-J. Eikema3, N. Zinger4, M. Haenel5, N. Schaap6,
C. Herrera Arroyo7, C. Schuermans8, W. Bethge9, M. Engelhardt10, J. Kuball11, M. Michieli12,
N. Schub13, K. M. O. Wilson14, J. H. Bourhis15, M. V. Mateos16, N. Robin17, E. Jost18,
N. Kröger19, J. M. Moraleda20, S. Sica21, P. J. Hayden22, M. Beksac23, S. Schönland24,
S. Manier1,25
1Hematologie Clinique, CHU de Lille; 2CHU de Lille, Univ Lille, INSERM U1286, Infinite,
Lille, France; 3EBMT Statistical Unit; 4EBMT Leiden Study Unit, Leiden, Netherlands;
5Klinikum Chemnitz gGmbH, Chemnitz, Germany; 6Nijmegen Medical Centre, Nijmegen, Netherlands;
7Hosp. Reina Sofia, Cordoba, Spain; 8GZA Hospitals, Antwerp, Belgium; 9Universitaet
Tuebingen, Tuebingen; 10University of Freiburg, Freiburg, Germany; 11University Medical
Centre, Utrecht, Netherlands; 12Centro di Riferimento Oncologico, Aviano, Italy; 13University
Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany; 14Department of Haematology,
Cardiff, United Kingdom; 15Gustave Roussy Cancer Campus, Villejuif, France; 16Hospital
Clínico, Salamanca, Spain; 17University College London Hospital, London, United Kingdom;
18University Hospital Aachen, Aachen; 19University Hospital Eppendorf, Hamburg, Germany;
20Hospital Universitario Virgen de la Arrixaca, Murcia, Spain; 21Universita Cattolica
S. Cuore, Rome, Italy; 22Department of Haematology, Trinity College Dublin, St. James’s
Hospital, Dublin, Ireland; 23Ankara University Faculty of Medicine, Ankara, Turkey;
24Medizinische Klinik u. Poliklinik V, University of Heidelberg, Heidelberg, Germany;
25Univ Lille, Canther, INSERM UMR-S1277 CNRS UMR9020, Lille, France
Background: Multiple myeloma (MM) is an incurable hematologic malignancy despite recent
therapeutic advances. In the context of lenalidomide-sensitive relapse, one of the
preferred options from EHA/ESMO and IMWG recommendations is the combination of carfilzomib-lenalidomide-dexamethasone
(KRd). This regimen has been approved based on the ASPIRE trial with a PFS of 26.3
months vs 17.6 months for Rd alone (HR = 0.69, p=0.0001). In younger patients, a second
autologous hematopoietic cell transplantation (auto-HCT) remains an option in case
of a prolonged remission after frontline auto-HCT. Only few data are available on
the combination of KRd followed by a second auto-HCT in first relapse.
Aims: Primary objective was to estimate progression-free survival (PFS) and overall
survival (OS) in patients who received a second line of treatment with KRd followed
by a second auto-HCT. Secondary objectives were to assess the response rates and identify
statistically significant co-variables on PFS and OS in this population.
Methods: This international retrospective study was performed in 22 centers affiliated
to the European Society for Blood and Marrow Transplant Society (EBMT). Patients with
MM were included if they received a second line of treatment with KRd induction followed
by a second auto-HCT between January 2016 and December 2018.
Results: A total of 51 patients were included, with a median age of 62 years (range
35 – 69). ISS at diagnosis was stage I for 18 patients (35.3%), stage II for 11 (21.6%),stage
III for 14 (27.5%) and missing for 8 patients (15.7%). Twenty-seven patients (52.9%)
were of standard cytogenetic risk and 11 patients (21.6%) of high cytogenetic risk
according to IMWG criteria, data was missing for 13 patients (25.5%). The median time
between the 1st to the 2nd transplant was 40.4 months (range 17.9–87.9). Regarding
the number of cycles of KRd received in induction, 25 patients received 3 or 4 cycles
(49.0%), 17 patients 5 or 6 cycles (33.3%) and 9 patients 7 to 12 cycles (17.7%).
The conditioning chemotherapy was melphalan alone in 46 patients (90.2%), a combination
of melphalan and another drug in 4 patients (7.8%) and cyclophosphamide alone in 1
patient (2.0%). The median follow-up was 36.7 months (range 5.3-58.0). The median
PFS was 32.6 months (95%CI: 30–39.9) and the median OS was not reached, while 36-
and 48-months OS rates were 87.5% (95%CI: 78.5–97.4) and 72.8% (95%CI: 57.0–92.8)
respectively. In univariate analysis, 2 co-variates were found to be associated with
a longer median PFS: an interval between the first and the 2nd auto-HCT of more than
4 years (mPFS of 36.1 vs. 30.6 months, p = 0.02) and the achievement of a very good
partial response (VGPR) or better at the 2nd transplant (mPFS of 33.5 vs. 27.8 months,
p = 0.01). These results were also observed in multivariate analysis, with HR of 0.41
(95%CI: 0.17-0.96, p = 0.04) and HR of 0.45 (95%CI: 0.21-0.98, p = 0.04), respectively.
No statistical association was found between the duration of PFS and cytogenetic risk
profile, with the limitation of a small number of patients with high-risk cytogenetics.
Regarding OS, no significantly relevant co-variables were found in uni- or multivariate
analysis.
Image:
Summary/Conclusion: A second auto-HCT after KRd induction is an effective treatment
for patients with a first lenalidomide-sensitive relapse of MM. Our study suggests
that patients with at least 4 years of remission after a frontline auto-HCT and who
achieved at least a VGPR after KRd induction, benefit the most from this treatment
strategy.
P958: REAL-LIFE CURRENT STANDARD OF CARE IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE
MYELOMA: SUBGROUP ANALYSES FROM THE LOCOMMOTION STUDY
H. Einsele1,*, P. Moreau2, V. De Stefano3, D. Dytfeld4, E. Angelucci5, R. Benjamin6,
H. Goldschmidt7, N. W. van de Donk8, B. Besemer9, C. Scheid10, R. Vij11, E. I. ’.
Groen-Damen12, M. Semerjian13, V. Strulev14, J. M. Schecter15, T. Roccia16, T. Nesheiwat17,
R. Wapenaar12, K. Weisel18, M.-V. Mateos19
1Universitätsklinikum Würzburg, Medizinische Klinik und Poliklinik II, Würzburg, Germany;
2University Hospital Hôtel-Dieu, Nantes, France; 3Section of Hematology, Catholic
University, Fondazione Policlinico A Gemelli, IRCCS, Rome, Italy; 4Poznań University
of Medical Sciences, Poznań, Poland; 5Hematology and Transplant Center, IRCCS Ospedale
Policlinico San Martino, Genova, Italy; 6Department of Haematology, King’s College
Hospital, London and School of Cancer and Pharmaceutical Sciences, King’s College,
London, United Kingdom; 7University Hospital Heidelberg, Heidelberg, Germany; 8Amsterdam
UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; 9University Hospital Tubingen,
Tubingen, Germany; 10University of Cologne, Cologne, Germany; 11Washington University
School of Medicine, St. Louis, MO, United States of America; 12Janssen-Cilag, Breda,
Netherlands; 13Janssen-Cilag, Issy-Les-Moulineaux, France; 14Janssen Pharmaceutica
NV, Beerse, Belgium; 15Janssen R&D, Raritan, NJ, United States of America; 16Janssen
R&D, High Wycombe, United Kingdom; 17Legend Biotech USA, Piscataway, NJ, United States
of America; 18University Medical Center Hamburg-Eppendorf, Hamburg, Germany; 19University
Hospital of Salamanca/IBSAL/CIC/CIBERONC, Salamanca, Spain
Background: Patients (pts) with relapsed/refractory multiple myeloma (RRMM) who have
had previous triple-class exposure to a proteasome inhibitor (PI), immunomodulatory
drug (IMiD), and anti-CD38 monoclonal antibody have limited treatment options and
represent an urgent and unmet clinical need. LocoMMotion (NCT04035226) is the first
prospective multinational study to assess real-life standard of care (SOC) treatment
in pts with triple-class exposed RRMM.
Aims: To assess efficacy in subgroups of pts treated with SOC therapies in the LocoMMotion
study.
Methods: All pts provided informed consent. This noninterventional study was conducted
across 76 sites (63 in Europe, 13 in the United States). Pts were included if they
had received ≥3 prior lines of therapy (LOT) or were refractory to a PI and an IMiD
(double-refractory); received a PI, an IMiD, and anti-CD38 monoclonal antibody and
had documented progressive disease during/after their last LOT. Real-life SOC therapies
were defined as those used in local clinical practice. Responses and disease progression
were based on International Myeloma Working Group criteria and assessed by response
review committee. Patient subgroups were categorized according to the following baseline
characteristics: age, Eastern Cooperative Oncology Group performance status (ECOG
PS), renal function, International Staging System stage, extramedullary plasmacytoma,
lactate dehydrogenase, bone marrow plasma cells percentage, number of prior LOT, triple-class
or penta-drug exposure, and refractoriness.
Results: 248 pts were enrolled as of May 21, 2021 (median follow-up: 11.0 months).
Pts received a median of 4.0 (range: 1-20) cycles of SOC treatment. Efficacy outcomes
were generally worse in pts with refractoriness to 3 classes of antimyeloma therapy,
presence of extramedullary plasmacytomas, high LDH, and ECOG PS ≥1 compared with pts
who did not have these characteristics (Table). Across all subgroups, overall response
rate ranged from 20.0% to 43.1%. Efficacy outcomes were not affected by age and number
of prior LOT.
Image:
Summary/Conclusion: In this first prospective study of real-life SOC treatment in
pts with triple-class exposed RRMM, subgroup analyses indicate that specific pt and
disease characteristics were associated with poor outcomes. Outcomes were poor in
triple-class refractory and non-triple-class refractory pts; however, the latter subgroup
had longer median progression-free survival. These data may help inform bridging strategies
for chimeric antigen receptor T-cell therapy.
P959: CILTACABTAGENE AUTOLEUCEL IN LENALIDOMIDE-REFRACTORY PATIENTS WITH PROGRESSIVE
MULTIPLE MYELOMA AFTER 1-3 PRIOR LINES OF THERAPY: CARTITUDE-2 BIOLOGICAL CORRELATIVE
ANALYSES AND UPDATED CLINICAL DATA
J. Hillengass1,*, A. D. Cohen2, M. Delforge3, H. Einsele4, H. Goldschmidt5, K. Weisel6,
M.-S. Raab7, C. Scheid8, J. M. Schecter9, K. C. De Braganca9, H. Varsos9, T.-M. Yeh9,
P. Mistry10, T. Roccia10, C. Corsale9, M. Akram11, L. Pacaud11, T. Nesheiwat11, M.
Agha12, Y. C. Cohen13
1Roswell Park Comprehensive Cancer Center, Buffalo, NY; 2Abramson Cancer Center, University
of Pennsylvania, Philadelphia, PA, United States of America; 3University Hospitals
(UZ) Leuven, Leuven, Belgium; 4Universitätsklinikum Würzburg, Medizinische Klinik
und Poliklinik II, Würzburg; 5University Hospital Heidelberg, Internal Medicine V
and National Center for Tumor Diseases (NCT), Heidelberg; 6University Medical Center
Hamburg-Eppendorf, Hamburg; 7University Hospital Heidelberg, and Clinical Cooperation
Unit Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg; 8University
of Cologne, Cologne, Germany; 9Janssen R&D, Raritan, NJ, United States of America;
10Janssen R&D, High Wycombe, United Kingdom; 11Legend Biotech USA, Piscataway, NJ;
12UPMC Hillman Cancer Center, Pittsburgh, PA, United States of America; 13Tel-Aviv
Sourasky (Ichilov) Medical Center and Sackler School of Medicine, Tel Aviv University,
Tel Aviv, Israel
Background: Cohort A of the multicohort phase 2 CARTITUDE-2 (NCT04133636) study is
assessing ciltacabtagene autoleucel (cilta-cel), a B-cell maturation antigen (BCMA)-directed
chimeric antigen receptor T-cell (CAR-T) therapy, in patients with multiple myeloma
(MM) who received 1-3 prior lines of therapy (LOT) and were refractory to lenalidomide
(len). This population is difficult to treat and has poor prognosis.
Aims: To present updated results from CARTITUDE-2 Cohort A.
Methods: All patients provided informed consent. Eligible patients had progressive
MM after 1-3 prior LOT that included a proteasome inhibitor (PI) and an immunomodulatory
drug (IMiD). Patients were len-refractory and had no prior exposure to BCMA-targeting
agents. Patients received a single cilta-cel infusion (target dose: 0.75×106 CAR+
viable T cells/kg) after lymphodepletion. Cilta-cel safety and efficacy were assessed.
The primary endpoint was minimal residual disease (MRD) negativity at 10-5 by next
generation sequencing. Patient management strategies were used to reduce the risk
of movement and neurocognitive adverse events (MNTs). Other assessments included pharmacokinetic
(PK) analyses (Cmax and Tmax of CAR+ T-cell transgene levels in blood), levels of
cytokine release syndrome (CRS)-related cytokines (e.g., IL-6) over time, peak levels
of cytokines by response and CRS, association of cytokine levels with immune effector
cell-associated neurotoxicity syndrome (ICANS), and CAR+ T cell CD4/CD8 ratio by response,
CRS, and ICANS.
Results: As of January 2022 (median follow-up: 17.1 months [range: 3.3-23.1]), cilta-cel
was administered to 20 patients (male: 65%; median age: 60 years [range: 38-75]).
Median number of prior LOT was 2 (range: 1-3); median time since MM diagnosis was
3.5 years (range: 0.7-8.0). 95% of patients were refractory to their last LOT; 40%
were triple-class refractory. Overall response rate was 95%, with 90% of patients
achieving ≥complete response and 95% achieving ≥very good partial response. Median
time to first response was 1.0 month (range: 0.7-3.3); median time to best response
was 2.6 months (range: 0.9-13.6). All MRD-evaluable patients (n=16) achieved MRD negativity
at 10-5. Median duration of response was not reached. The 12-month progression-free
survival rate was 75% and the 12-month event-free rate was 79%. CRS occurred in 95%
of patients (grade 3/4: 10%), with a median time to onset of 7 days (range: 5-9) and
median duration of 3 days (range: 2-12). 30% of patients had neurotoxicity (5 grade
1/2 and 1 grade 3/4). ICANS occurred in 3 patients (15%; all grade 1/2); 1 patient
had facial paralysis (grade 2). No MNTs were observed. 1 death due to COVID-19 occurred
and was assessed as treatment-related by the investigator; 2 deaths due to progressive
disease and 1 due to sepsis (not related to treatment) also occurred. Based on preliminary
PK analyses of CAR transgene by qPCR, peak expansion of CAR-T cells occurred at day
10.5 (range: 8.7-42.9); median persistence was 153.5 days (range: 57.1-336.8).
Summary/Conclusion: A single cilta-cel infusion led to deepening and durable responses
at this longer follow-up (median 17.1 months) in patients with MM who had 1-3 prior
LOT and were len-refractory. Follow-up is ongoing. We will present updated and detailed
PK, cytokine, and CAR-T subset analyses as well as clinical correlation to provide
novel insights into biological correlates of efficacy and safety in this difficult-to-treat
patient population, which is being further evaluated in the CARTITUDE-4 study (NCT04181827;
enrollment concluded).
P960: HEALTH-RELATED QUALITY OF LIFE IN THE LOCOMMOTION STUDY OF REAL-LIFE CURRENT
STANDARD OF CARE IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA
M. Delforge1,*, P. Moreau2, H. Einsele3, V. De Stefano4, J. Lindsey-Hill5, L. Vincent6,
S. Mangiacavalli7, A. Perrot8, E. Ocio9, S. ten Seldam10, E. I. ’. Groen-Damen11,
M. Semerjian12, V. Strulev13, J. M. Schecter14, T. Roccia15, K. S. Gries14, T. Nesheiwat16,
R. Wapenaar11, M.-V. Mateos17, K. Weisel18
1University Hospitals (UZ) Leuven, Leuven, Belgium; 2University Hospital Hotel-Dieu,
Nantes, France; 3Universitätsklinikum Würzburg, Medizinische Klinik und Poliklinik
II, Wuerzburg, Germany; 4Section of Hematology, Catholic University, Fondazione Policlinico
A Gemelli, IRCCS, Rome, Italy; 5Nottinghamshire University Hospitals NHS Trust, Nottingham,
United Kingdom; 6Département d’hématologie Clinique, Centre Hospitalier Universitaire
de Montpellier, Montpellier, France; 7Fondazione IRCCS Policlinico San Matteo, University
of Pavia, Pavia, Italy; 8Centre Hospitalier Universitaire de Toulouse, Service d’Hématologie,
Toulouse, France; 9Hospital Universitario Marqués de Valdecilla (IDIVAL), Universidad
de Cantabria, Santander, Spain; 10Myeloma Patients Europe, Brussels, Belgium; 11Janssen-Cilag,
Breda, Netherlands; 12Janssen-Cilag, Issy-les-Moulineaux, France; 13Janssen Pharmaceutica
NV, Beerse, Belgium; 14Janssen R&D, Raritan, NJ, United States of America; 15Janssen
R&D, High Wycombe, United Kingdom; 16Legend Biotech USA Inc, Piscataway, NJ, United
States of America; 17University Hospital of Salamanca/IBSAL/CIC, Salamanca, Spain;
18University Medical Center Hamburg-Eppendorf, Hamburg, Germany
Background: Assessment of patient-reported outcomes (PRO) can inform how real-life
standard of care (SOC) treatments affect health-related quality of life (HRQoL) for
patients with relapsed/refractory multiple myeloma (RRMM). We present measures of
symptoms, functioning, and overall HRQoL from LocoMMotion (NCT04035226), the first
prospective, multinational study of real-life SOC in triple-class exposed patients
with RRMM.
Aims: To assess HRQoL in patients with RRMM receiving real-life current SOC in the
LocoMMotion study.
Methods: All patients provided informed consent. LocoMMotion is a noninterventional
study across 76 sites (63 Europe, 13 United States) in patients who had received ≥3
prior lines of therapy (LOT) or were refractory to proteasome inhibitor (PI) and immunomodulatory
drug (IMiD). Patients received PI, IMiD, and anti-CD38 monoclonal antibody and had
disease progression during/after their last LOT. Real-life SOC treatments were defined
as those used in local clinical practice. The following questionnaires were used:
European Organisation for Research and Treatment of Cancer Core Quality of Life Questionnaire
(EORTC QLQ-C30), 4 single items from EORTC QLQ-myeloma-specific module (EORTC QLQ-MY20),
and EuroQol 5-Dimension 5-Level (EQ 5D-5L). HRQoL data were collected at baseline
(BL), day 1 of each treatment cycle, end of treatment visit, and during follow-up
(every 4 weeks). Established thresholds were used to evaluate improvement compared
with BL health status. Mixed models for repeated measures were used to assess within-group
change.
Results: The questionnaire completion rate was 75.6% during SOC treatment in the LocoMMotion
study (N=248; male: 54.4%; median age: 68 years; median cycles of SOC: 4.0 [range:
1-20]). Most patients did not achieve meaningful improvement (defined by a literature-based
minimally important difference of 10 points in mean score) in PRO scores. This was
most pronounced in pain symptoms, with 62% of patients showing no meaningful improvement
during the first 3 months of treatment and 55% showing no improvement during the full
treatment duration. Least square (LS) mean changes from BL during SOC treatment and
subsequent LOT for the overall population are described (Table). Patients with ≥very
good partial response during SOC treatment showed greater improvement in PRO scores,
including LS mean change for pain score (-14.9 [95% confidence interval: -22.9, -7.0]).
Image:
Summary/Conclusion: This first prospective study of real-life current SOC in triple-class
exposed patients with RRMM reported limited gains in HRQoL, most notably in pain symptoms.
There is an urgent and unmet need for therapies that lead to deep responses and delayed
disease progression, as these are associated with improvements in HRQoL.
P961: CILTACABTAGENE AUTOLEUCEL, A BCMA-DIRECTED CAR-T CELL THERAPY, IN PATIENTS WITH
RELAPSED/REFRACTORY MULTIPLE MYELOMA: 2-YEAR POST LPI RESULTS FROM THE PHASE 1B/2
CARTITUDE-1 STUDY
Y. Lin1,*, T. Martin2, J. G. Berdeja3, A. Jakubowiak4, M. Agha5, A. D. Cohen6, A.
Deol7, M. Htut8, A. Lesokhin9, N. C. Munshi10, E. O’Donnell11, C. C. Jackson12, T.-M.
Yeh12, A. Banerjee13, E. Zudaire13, D. Madduri12, C. Zhou14, L. Pacaud14, S. Z. Usmani9,
S. Jagannath15
1Mayo Clinic, Rochester, MN; 2UCSF Helen Diller Family Comprehensive Cancer Center,
San Francisco, CA; 3Sarah Cannon Research Institute, Nashville, TN; 4University of
Chicago, Chicago, IL; 5UPMC Hillman Cancer Center, Pittsburgh, PA; 6Abramson Cancer
Center, University of Pennsylvania, Philadelphia, PA; 7Karmanos Cancer Institute,
Wayne State University, Detroit, MI; 8City of Hope Comprehensive Cancer Center, Duarte,
CA; 9Memorial Sloan Kettering Cancer Center, New York, NY; 10Dana-Farber Cancer Institute,
Harvard Medical School; 11Massachusetts General Hospital, Harvard Medical School,
Boston, MA; 12Janssen R&D, Raritan, NJ; 13Janssen R&D, Spring House, PA; 14Legend
Biotech USA, Piscataway, NJ; 15Mount Sinai Medical Center, New York, NY, United States
of America
Background: The phase 1b/2 CARTITUDE-1 study (NCT03548207) reported early, deep, and
durable responses with ciltacabtagene autoleucel (cilta-cel), a chimeric antigen receptor
T (CAR-T) cell therapy with 2 B-cell maturation antigen (BCMA)–targeting single-domain
antibodies, in heavily pretreated patients with relapsed/refractory multiple myeloma
(RRMM). At a median follow-up of ~1 year, the overall response rate (ORR) was 97%,
with 67% of patients achieving stringent complete response (sCR). Progression-free
survival (PFS) and overall survival (OS) rates at 1 year were 77% and 89%, respectively.
Aims: To report updated 2-year post last patient in (LPI) results (total median follow-up
of ~30 months). CARTITUDE-1 results at a median follow-up of 21.7 months are reported
here.
Methods: All patients provided informed consent. Patients with RRMM had received ≥3
prior lines of therapy (LOT) or were refractory to a proteasome inhibitor (PI) and
immunomodulatory drug (IMiD). Patients had received a PI, IMiD, and anti-CD38 antibody.
After apheresis, bridging therapy was allowed. A single cilta-cel infusion was administered
at a target dose of 0.75×106 CAR+ viable T cells/kg 5-7 days after lymphodepletion.
The primary objectives were to evaluate the safety and efficacy of cilta-cel. Response
assessments were based on International Myeloma Working Group criteria by independent
review committee. Minimal residual disease (MRD) negativity at 10-5 was assessed by
next-generation sequencing.
Results: 97 patients (male: 59%; median age: 61 years) received cilta-cel as of July
22, 2021. The median number of prior LOT was 6 (range: 3-18). 84% of patients were
penta-drug exposed, 88% were triple-class refractory, 42% were penta-drug refractory,
and 99% were refractory to their last LOT. The ORR was 97.9% (95% CI: 92.7-99.7),
with 94.9% of patients achieving very good partial response and 82.5% achieving sCR.
Median time to first response was 1.0 month, median time to best response was 2.6
months, and median time to ≥CR was 2.9 months. Median duration of response was not
reached. Among 61 MRD-evaluable patients, 92% were MRD negative at 10-5, which was
sustained in 44% of patients (27/61) for ≥6 months and in 18% of patients (11/61)
for ≥12 months. Median PFS and OS were not reached. The 2-year PFS rate was 60.5%
(95% CI: 48.5-70.4) overall. The 2-year PFS rate was 91% for patients with sustained
MRD negativity ≥6 months and 100% for those with sustained MRD negativity ≥12 months.
No new safety signals or new cases of CAR-T cell neurotoxicity, movement/neurocognitive
treatment-emergent adverse events, or treatment-related deaths were reported since
1-year median follow-up. Over a median follow-up of ~2-years, 15 second primary malignancies
occurred in 11 patients.
Summary/Conclusion: A single cilta-cel infusion resulted in deepening and durable
responses in heavily pretreated patients with RRMM at a median follow-up of ~2 years.
The safety profile was manageable. Follow-up in CARTITUDE-1 is ongoing. We will present
landmark 2-year post LPI data, with ~8 months of additional follow-up (~30 months
total median follow-up). Cilta-cel is also being evaluated in earlier LOT and outpatient
settings across the CARTITUDE program, including NCT04133636, NCT04181827, NCT04923893.
P962: CHANGE IN SOLUBLE BCMA LEVEL MAY BE A SURROGATE MARKER OF EARLY RESPONSE TO
THERAPY IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA (RRMM): PRELIMINARY
RESULTS FROM A PHASE I STUDY OF CEVOSTAMAB
R. Nakamura1,*, R. Hendricks1, R. Dokhaei1, M. Susilo1, D. Wilson1, S. Trudel2, A.
Cohen3, S. J. Harrison4, A. Krishnan5, R. Fonseca6, J. I. Adamkewicz1, M. Li1, C.
Wong1, J. Cooper1, T. Sumiyoshi1
1Genentech, Inc., South San Francisco, CA, United States of America; 2Princess Margaret
Cancer Centre and University of Toronto, Toronto, ON, Canada; 3Abramson Cancer Center
and University of Pennsylvania, Philadelphia, PA, United States of America; 4Peter
MacCallum Cancer Centre, Sir Peter MacCallum Department of Oncology, Melbourne University,
and The Royal Melbourne Hospital, Melbourne, VIC, Australia; 5City of Hope, Duarte,
CA; 6Mayo Clinic in Arizona, Phoenix, AZ, United States of America
Background: Cevostamab is an FcRH5xCD3 bispecific antibody that facilitates T-cell
directed killing of myeloma cells, and has shown promising activity and manageable
safety in a Phase I study in patients (pts) with RRMM (NCT03275103; Trudel et al.
ASH 2021). B-cell maturation antigen (BCMA) is a membrane-bound protein that is expressed
preferentially by malignant plasma cells and has become an important therapeutic target
in MM. Shedding of membrane-bound BCMA, mediated by γ-secretase, gives rise to the
soluble form of BCMA (sBCMA) which is often present at elevated levels in pts with
MM. Normalization of sBCMA may be a predictor of response to therapy, which may be
independent of treatment and target.
Aims: To evaluate sBCMA as a biomarker of early response in pts enrolled in the single
step-up dosing cohorts of the cevostamab Phase I study.
Methods: Plasma samples were collected at baseline and at Cycle (C) 1 Day (D) 1 (pre-infusion
and end of infusion [EOI]), C1D2, C1D4, C1D8 (pre-infusion and EOI), C1D9, C1D11,
and C2D1 (pre-infusion and EOI). sBCMA levels were quantified using hybrid immunoaffinity
capture with LC-MS/MS. Pts were stratified by refractory status, prior therapy, prior
transplant, cytogenetic risk and response (responder [≥PR] or non-responder [<PR]),
and samples from each category were grouped for analysis. Associations between baseline
sBCMA levels and baseline patient characteristics or response to treatment were evaluated
using an unpaired 2-sample Wilcoxon test (significance level: p≤0.05). The relationship
between sBCMA dynamics (percent change in sBCMA level from baseline to C2D1 EOI) and
best response was evaluated using logistic linear regression. All pts provided informed
consent.
Results: At cut-off (January 7, 2022), 97/103 pts in the single step-up cohorts were
biomarker evaluable. No clear differences in baseline sBCMA levels were observed in
pts stratified by refractory status, prior transplant, or prior anti-CD38 or anti-BCMA
therapy. Analysis of a subset of pts with cytogenetic data showed a trend towards
lower baseline sBCMA levels in the standard-risk group (n=18) vs the high-risk group
(n=42; p=0.032). In the active dose cohorts (3.6/20mg+; n=77), baseline sBCMA levels
were comparable between responders and non-responders. However, baseline sBCMA levels
were lower in pts achieving a VGPR or better vs those achieving a PR or no response
(median: 47.7ng/mL vs 120.5ng/mL; p=0.037). At C2D1 EOI, most responders had a reduction
in sBCMA relative to baseline, with the pts who achieved sCR having the greatest decrease
(median: –92.10%). In comparison, an increase in sBCMA was observed in non-responders
at C2D1 EOI, with the pts who had PD having the greatest increase (median: 93.14%).
The extent of reduction in sBCMA from baseline to C2D1 EOI was significantly correlated
with best overall response rate (PR or better) (p=1.8x10–5).
Summary/Conclusion: In this study, baseline sBCMA was not associated with refractory
status, prior transplant or treatment in pts with RRMM. Patients with standard-risk
cytogenetics had lower baseline sBCMA levels vs those with high-risk cytogenetics.
The kinetics of sBCMA change from baseline to C2D1 EOI corresponded with response
to cevostamab, with greater reductions increasing the probability of best response.
These early data suggest that sBCMA dynamics may be a potential tool for early disease
monitoring and identification of response in RRMM. Updated data will be presented.
P963: EVOLUTION OF TREATMENT PATTERNS AND OVERALL SURVIVAL IN PATIENTS WITH RELAPSED/REFRACTORY
MULTIPLE MYELOMA RECEIVING A SECOND LINE OF THERAPY BETWEEN 2012 AND 2020: ANALYSIS
OF THE PREAMBLE COHORT
D. Kuter1,*, H. Goldschmidt2, D. Cella3, M. T. Petrucci4, P. Moreau5, B. Durie6, A.
Juarez-Garcia7, L. Trong7, J. Gu7, G. Hernandez Rivera8, S. Dhanasiri8, T. Marshall7,
J. Wang7, L. Lacoin9, K. Ramasamy10, R. Vij11
1Hematology Division, Massachusetts General Hospital, Boston, MA, United States of
America; 2University Hospital Heidelberg, Internal Medicine V and National Center
for Tumor Diseases (NCT), Heidelberg, Germany; 3Department of Medical Social Sciences
and Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago,
IL, United States of America; 4Department of Cellular Biotechnologies and Hematology,
Sapienza University of Rome, Rome, Italy; 5Service d’hématologie clinique, CHU Hôtel
Dieu, Nantes, France; 6Cedars Sinai Cancer Center, Los Angeles, CA; 7Bristol Myers
Squibb, Princeton, NJ, United States of America; 8Bristol-Myers Squibb, Boudry, Switzerland;
9Epi-Fit, Bordeaux, France; 10Department of Haematology, Oxford University Hospitals
NHS Foundation Trust, Oxford, United Kingdom; 11Siteman Cancer Center, Washington
University in St. Louis, St. Louis, MO, United States of America
Background: Treatment (Tx) options such as immunomodulatory agents (IMiDs), proteasome
inhibitors (PIs), and more recently anti-CD38 monoclonal antibodies (mAbs) have improved
clinical outcomes for patients (pts) with multiple myeloma (MM); yet many experience
disease relapse or become refractory to Tx. PREAMBLE is a prospective, observational
cohort study of pts with MM in North America and Europe.
Aims: To assess Tx patterns and overall survival (OS) evolution over time in pts with
relapsed/refractory MM (RRMM) who received a second-line therapy (2L) between 2012
and 2020 in the PREAMBLE study.
Methods: This analysis focused on a subset of pts enrolled in PREAMBLE at the time
of initiating 2L Tx for MM. The index date was defined as the enrollment date. Pts
were followed until death, completion of 3 years of follow-up (FU), date of loss to
FU, or date of last visit prior to database extraction date. Pt characteristics, previous
Tx since diagnosis and 2L regimens received were described at index date. Pts were
grouped by date of enrollment in 3 cohorts (2012-15, 2016-18, and 2019-20). Duration
of 2L Tx and OS were analyzed using the Kaplan Meier method.
Results: 611 pts were included with a median age of 71 years at the start of 2L Tx,
56.8% were male, and 72.7% and 27.3% were from Europe and North America, respectively.
352 pts received 2L in 2012-2015, 199 in 2016-2018 and 60 in 2019-2020. Median FU
was 22.8, 32.3 and 18.4 months in the 3 cohorts. The median time from diagnosis to
the start of 2L Tx was 33.3 months, increasing from 31.8 months in the 2012-15 cohort
to 40.2 months in the 2019-20 cohort. Prior to the study entry, during first line
(1L), 31.6% of pts received a stem cell transplant (SCT) (34.9%, 27.6% and 25.0% respectively
for the 3 cohorts). Overall, the main 1L regimens received were PI-based regimens
(48.0%), IMiD-based regimens (24.9%), and combinations of IMiD and PI (22.7%). Fewer
than 5 pts received an anti-CD38 mAb in 1L. The proportion of pts who received a combination
of IMiD and PI in 1L increased from 19.3% in 2012-15 to 25.1% in 2016-18 and 35.0%
in 2019-20.
Regarding 2L Tx received, single class regimens (IMiD only and PI only) use dropped
from 88.0% in 2012-15 to 62.8% in 2016-18 and 18.4% in 2019-20. It was replaced by
various combinations of Tx as shown in the figure. The combinations of Tx containing
anti-CD38 mAb had increased from 9.0% to 60.1% since the first launch of anti-CD38
in 2016. 9.0% of pts received an SCT during 2L (10.5%, 8.0% and 3.3% for the 3 cohorts).
The unadjusted median duration of 2L for the 3 cohorts was of 6.0, 8.1 and 9.9 months
(log rank test, p=0.01). A significant improvement in unadjusted OS was observed (log
rank test, p<0.001). One year-OS (95% confidence interval [CI]) was 78.4% (73.9-82.9),
84.8% (79.7-89.9) and 93.0% (86.3-99.7); 2 year-OS (95%CI) was 61.3% (55.8-68.5),
74.5% (68.2-80.6) and 77.7% (65.0-90.4) in 2015-16, 2016-18 and 2019-20 cohorts, respectively.
Median OS was not reached.
Image:
Summary/Conclusion: Major changes in 2L Tx patterns were observed in pts with RRMM
between 2012 and 2020 with the decreased use of single class regimens and the increased
use of combination Tx. Pts were likely to be exposed to all 3 main classes of drugs
(IMiD, PIs and anti-CD38) earlier in the Tx pathway. A trend towards improvement in
OS from start of 2L was also observed over time, likely driven by the changes in Tx
patterns. Future analyses of this on-going study will allow to evaluate the long-term
impact of those new Tx and the remaining unmet needs.
P964: IMMUNOPHENOTYPED-SUSPENSION-MULTIPLEX FISH BY IMAGING FLOW CYTOMETRY FOR THE
SIMULTANEOUS DIAGNOSIS OF THREE PIVOTAL IGH TRANSLOCATIONS IN MULTIPLE MYELOMA
T. Tsukamoto1,*, M. Kinoshita2, K. Yamada2, T. Yamaguchi3, Y. Chinen1, S. Mizutani1,
T. Fujino1, Y. Shimura1, T. Kobayashi1, J. Inazawa4, J. Kuroda1
1Division of Hematology and Oncology, Kyoto Prefectural University of Medicine, Kyoto;
2Sysmex Corporation, Hyogo; 3General Laboratory, Bio Medical Laboratories, Inc.; 4Department
of Molecular Cytogenetics, Medical Research Institute, Tokyo Medical and Dental University,
Tokyo, Japan
Background: The cytogenetic abnormalities including t(4;14)(p16;q32), t(14;16)(q32;q23),
and t(11;14)(q13;q32) are associated with the molecular profiles, clinical features,
and treatment outcome in multiple myeloma (MM). Although the double-color interphase
fluorescence in situ hybridization (DC-FISH) is well established to detect those translocations,
the conventional method can analyze only one abnormality on each slide.
Aims: To analyze multiple chromosomal abnormalities simultaneously with high sensitivity
and specificity, we aimed to develop a new diagnostic modality of four-color FISH
for multiple translocations in immunophenotyped cells in suspension using imaging
flow cytometry (immunophenotyped-suspension-multiplex (ISM)-FISH).
Methods: For the ISM-FISH, we first subjected Carnoy-fixed cells in suspension to
immunostaining using a brilliant violet 421-conjugated anti-CD138 antibody. Then,
we hybridized with four different FISH probes, i.e., Texas Red (TxRed)-conjugated
probe for IGH, fluorescein isothiocyanate (FITC)-conjugated probe for FGFR3, Gold-conjugated
probe for cyclin D1 (CCND1), and Cy5-conjugated probe for c-MAF, in suspension. Then,
more than 2.5 × 104 nucleated cells for each sample were analyzed with imaging flow
cytometer MI-1000 (Sysmex) by six channels: one for immunophenotyping, four for FISH
and one for bright field image. In the process of imaging flowcytometric analysis,
we sorted CD138-positive cell fraction, computed the distance between FISH probes
using originally developed spot counting tool, and determined to be positive for translocation
when the distance is under the threshold.
Results: By this ISM-FISH system, we simultaneously examined three chromosomal translocations,
including t(4;14), t(14;16), and t(11;14), in three MM cell lines, KMS-11, KMS-21-BM,
KMS-26, and successfully detect the abnormalities. Using different proportions of
transformation-positive MM cells spiked with transformation-negative leukemic cells,
HL-60, ISM-FISH showed a sensitivity of less than 0.1%. Next, ISM-FISH and conventional
DC-FISH were performed for bone marrow nucleated cells from 70 patients with MM or
monoclonal gammopathy of undetermined significance (MGUS) and the experiments showed
a promising qualitative diagnostic ability in detecting t(11;14), t(4;14), and t(14;16)
of our ISM-FISH, which was more sensitive than standard DC-FISH, examining 200 interphase
cells with its optimal sensitivity of up to 1.0%. Furthermore, the ISM-FISH showed
a positive concordance of 83.3% and negative concordance of 97.4% using standard DC-FISH,
examining 200 interphase cells. Furthermore, in three patients (4%), t(4;14) or t(11;14)
were positive by ISM-FISH but negative by DC-FISH examining 200 cells. Extensive DC-FISH
study examining additional 1,000 cells showed positive in these ISM-FISH-positive/DC-FISH
(200cells)-negative samples, suggesting higher sensitivity of ISM-FISH than that of
the standard DC-FISH.
Image:
Summary/Conclusion: This study developed the new diagnostic system of ISM-FISH, which
enables the simultaneous diagnosis of three clinically pivotal chromosomal translocations,
t(4;14), t(14;16), and t(11;14), in MGUS and MM. This system may facilitate rapid
and reliable cytogenetic diagnosis and promote patient-oriented therapy according
to the type of chromosomal translocation in the clinical practice setting.
P965: FOLLOW-UP ANALYSIS OF THE RANDOMIZED PHASE II TRIAL OF BORTEZOMIB, LENALIDOMIDE,
DEXAMTHASONE WITH/WITHOUT ELOTUZUMAB FOR NEWLY DIAGNOSED, HIGH RISK MULTIPLE MYELOMA
(SWOG-1211)
S. Usmani1,*, A. Hoering2, S. Ailawadhi3, R. Sexton2, B. Lipe4, J. Valent5, M. Rosenzweig6,
J. Zonder7, M. Dhodapkar8, N. Callander9, T. Zimmerman10, P. Voorhees11, B. Durie12,
S. V. Rajkumar13, P. Richardson14, R. Orlowski15
1Memorial Sloan Kettering Cancer Center, New York; 2Cancer Research And Biostatistics,
Seattle; 3Mayo Clinic, Jacksonville; 4University of Rochester Medical Center, Rochester,
NY; 5Cleveland Clinic, Cleveland; 6City of Hope, Los Angeles; 7Karmanos Cancer Institute,
Detroit, MI; 8Emory Winship Cancer Institute, Atlanta GA; 9University of Wisconsin,
Madison; 10Beigene, Chicao; 11Levine Cancer Institute, Charlotte; 12Cedar Sinai Medical
Center, Los Angeles; 13Mayo Clinic, Rochester MN; 14Dana Farber Cancer Institute,
Boston MA; 15MD Anderson Cancer Center, Houston TX, United States of America
Background: The introduction of immunomodulatory agents, proteasome inhibitors, and
autologous stem cell transplantation (ASCT) has improved outcomes for patients with
multiple myeloma (MM), but those with high risk MM (HRMM) have a poor long-term prognosis.
Herein we provide survival outcomes on the first randomized trial in newly diagnosed
HRMM, S1211, to follow-up on the previously reported progression-free survival (PFS)
(NCT01668719, Usmani SZ et al, Lancet Haem 2021).
Aims: S1211 is a randomized phase II trial comparing 8 cycles of lenalidomide, bortezomib
and dexamethasone (RVd) induction followed by dose-attenuated RVd maintenance until
disease progression with or without elotuzumab (RVd-Elo). Stem cell collection was
allowed, but ASCT was deferred until progression.
Methods: HRMM was defined by one of the following: gene expression profiling high-risk
(GEPhi), t(14;16), t(14;20), del(17p), amplification 1q21, primary plasma cell leukemia
(pPCL), or elevated serum LDH (≥ 2X ULN). Median PFS was the primary endpoint, using
a one-sided stratified log-rank test at a one-sided significance level of 0.1. Secondary
endpoints included overall response rate (ORR), adverse events (AE), serious adverse
events (SAE) and OS. Response was assessed using the IMWG 2009 criteria.
Results: S1211 enrolled 103 evaluable patients, RVd n=54, RVd-Elo n=49. 74% had ISS
II/III, 48% amp1q21, 38% del(17p), 11% t(14;16), 9% GEPhi, 7% pPCL, 5% t(14;20) and
4% elevated LDH (17% ≥1 feature). With median follow-up of 72 months (mos.), no difference
in median PFS was observed [RVd-Elo=29 mos., RVd= 34 mos., HR = 1.11 (80% CI=0.82,
1.49, p=0.66]. No difference in OS was observed [RVd-Elo = median not reached (NR),
RVd= 68 mos., HR = 0.85 (80% CI: 0.59, 1.23), p-value = 0.58]. 76% pts had ≥Grade
3 AEs, no differences in the safety profile were observed. Amongst patients with gain/amp
1q21, median PFS was [RVd-Elo=31 mos., RVd= 37 mos., HR = 1.48 (80% CI= 0.95, 2.31),
p=0.25], median OS was [RVd-Elo = 61 mos., RVd= 68 mos., HR = 1.23 (80% CI: 0.72,
2.10), p-value = 0.63]. In patients with del(17p), median PFS was observed [RVd-Elo=41
mos., RVd= 30 mos., HR = 0.98 (80% CI= 0.60, 1.58), p=0.95], median OS was [RVd-Elo
= NR, RVd= 72 mos., HR = 0.77 (80% CI: 0.40, 1.48), p-value = 0.61].
Summary/Conclusion: In the first randomized HRMM study reported to date, the addition
of Elo to RVd induction and maintenance did not improve PFS and OS with a median follow-up
of 6 years. Although the median PFS for Del17p subgroup on RVd-Elo arm is higher than
RVd, it did not achieve statistical significance. The PFS and OS observed for gain/amp
1q21 and del17p in the RVd control arm may serve as important benchmarks for future
enrichment design HRMM clinical trials. The PFS and OS in both arms of the study exceeded
the original statistical assumptions and support the role for PI/IMiD combination
induction/maintenance therapy for this population.
P966: DARATUMUMAB, CARFILZOMIB, POMALIDOMIDE AND ELOTUZUMAB FOR THE TREATMENT OF POEMS
SYNDROME- THE MAYO CLINIC EXPERIENCE
I. Vaxman1,2,*, S. Kumar1, F. buadi1, M. Lacy1, D. Dingli1, A. Fonder1, M. Hobbs1,
S. Hayman1, L. Y. hwa1, T. Kouralis1, R. Warsame1, E. Muchtar1, L. Nelson1, P. Kapoor1,
M. Grogan1, R. Go1, W. Gonsalves1, M. Siddiqi1, K. Robert1, V. Rajkumar1, M. Gertz1,
A. Dispenzieri1
1hematology, Mayo Clinic, Rochester, United States of America; 2hematology, Beilinson,
Hod Hasharon, Israel
Background: POEMS (Polyneuropathy, Organomegaly, Endocrinopathies, Monoclonal protein,
Skin changes) syndrome is a rare paraneoplastic syndrome and therapies are directed
against plasma cells that produce the proteins that cause this syndrome. Novel therapies
are widely used in multiple myeloma aiming for plasma cell eradication. However, data
on their use in POEMS syndrome are lacking.
Aims: To provide the Mayo Clinic experience in treating 16 patients with relapsed
POEMS syndrome with novel agents (daratumumab, carfilzomib, pomalidomide, and elotuzumab).
Methods: We identified all POEMS patients seen at Mayo Clinic Rochester, Minnesota
using a prospectively maintained database of patients seen at our center between June
1979 and May 2021. Of these patients, we identified all the patients that were treated
with a “novel” agent, defined as daratumumab, carfilzomib, pomalidomide or elotuzumab.
The primary endpoints were response to therapy (hematological, PET, VEGF and clinical,
which will be referred to as responseH, P, V, C, respectively), and time to next therapy
(TTNT), defined as time from institution of regimen of interest to next therapy. The
secondary outcome was safety.
Results: The median age at POEMS diagnosis was 57 years (range 39-79) and 15 patients
(93%) were men. Among all patients at diagnosis, 10 (63%) had skin changes, 15 (94%)
had signs of extravascular volume overload, and 15 (94%) patients had endocrine disorders.
Ten and six patients had IgA and IgG isotypes, respectively, and all the patients
had lambda light chain isotype. Radiological evidence for sclerotic lesions were found
in 13 (81%) patients.
The median time from diagnosis to novel agent’s first dose administration was 50 months
(IQR 23-122) and the median lines of therapy prior to novel agent was 2 (range 1-4).
Twelve patients (75%) underwent prior autologous stem cell transplantation (ASCT),
and 5 patients had prior lenalidomide. The median age at novel agent administration
was 63 years (IQR 51-70) and 4 patients (24%) were 70 years or older.
The patients were treated with a a doublet including dexamethasone (N=5) (31%) or
in various combinations with other agents: DRd (N=6), DC(V)d (N=3), KRd (N=3), KPd
(N=1), DP(V)d (N=5), and EloRd (N=1).
The outcomes with novel agent therapies were favorable (Table 1). Among patients treated
with daratumumab based therapies (N=17), 9 patients achieved CR/VGPRH, 7 patients
achieved CRV, and 5 patients achieved CRP. Among patients treated with carfilzomib-based
therapies (N=6), 3 patients achieved CR/VGPRH and one achieved PRH. Only one patient
treated with carfilzomib-based therapies achieved a clinical response.
Neither patient who received pomalidomide and dexamethasone or elotuzumab with lenalidomide
and dexamethasone responded. At a median follow-up of 38 months since starting of
the novel agent (IQR 24-57), 15 of the patients (94%) are still alive, and the median
TTNT was not reached.
None of the patients discontinued therapy due to adverse events and no deaths occurred
on therapy. Novel therapies were safe with 7 events of hospitalization due to pneumonia
(4 in daratumumab-based therapies and 3 on carfilzomib-based therapies), and 4 patients
were hospitalized due to volume overload (all received dexamethasone with therapy).
Three patients experienced infusion-related reactions (IRR) to the first dose of IV
daratumumab.
Image:
Summary/Conclusion: Response rate was high and the responses were deep. Novel agent
therapies were safe, and no death case occurred on therapy. Future studies are needed
to clarify the optimal sequence of novel agents and the best combination.
P967: TUMOR BURDEN AS A CRITICAL PROGNOSTIC FACTOR OF PRIMARY EXTRAMEDULLARY DISEASE
M. Vlachová1,*, M. Štork2, S. Ševčíková1, T. Jelínek3, J. Minařík4, J. Radocha5, P.
Krhovská4, L. Pospíšilová6, I. Špička7, J. Straub7, P. Pavlíček8, A. Jungová9, V.
Sandecká2, V. Maisnar5, R. Hájek3, L. Pour2
1Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk
University; 2Clinic of Internal Medicine, Hematology and Oncology, University Hospital
Brno, Brno; 3Department of Hematooncology, University Hospital Ostrava and Faculty
of Medicine University of Ostrava, Ostrava; 4Department of Hemato-Oncology, University
Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc,
Olomouc; 54th Department of Internal Medicine – Hematology, Faculty Hospital and Charles
University in Hradec Kralove, Hradec Kralove; 6Institute of Biostatistics and Analyses,
Ltd., Brno; 71st Medical Department - Clinical Department of Haematology, the First
Faculty of Medicine and General Teaching Hospital Charles University; 8Department
of Internal Medicine and Hematology, University Hospital Kralovske Vinohrady, Prague;
9Hematology and Oncology Department, Charles University Hospital, Pilsen, Czechia
Background: Multiple myeloma (MM) is the second most common hematological malignancy
characterized by malignant plasma cell (PC) infiltration of the bone marrow (BM).
In the case of extramedullary MM (EMD), plasma cells survive and proliferate outside
of the bone marrow microenvironment. Primary EMD is found in newly diagnosed MM (NDMM)
patients.
Aims: The aim of this study was to describe clinical features and treatment outcomes
of primary EMD patients and to define possible risk factors.
Methods: In total, 724 primary EMD patients were diagnosed in the Czech Republic between
2004 and 2021 by using modern imaging methods. As a reference group, 2440 MM patients
without any evidence of EMD were used. All patients enrolled into the analysis were
treated by novel agents. Patients’ data were analyzed from the Registry of Monoclonal
Gammopathies of the Czech Myeloma Group (RMG).
Results: Primary EMD patients had less frequently advanced ISS (ISS 3) (OR 0.51 [95%
CI: 0.42–0.63], p<0.001), high paraprotein levels (>20g/l) (OR 0.64 [95% CI: 0.54–0.76],
p<0.001) and high PC infiltration of bone marrow (>10%) (OR 0.35 [95% CI: 0.30–0.42],
p<0.001) when compared to reference MM patients. Median PFS in newly diagnosed primary
EMD patients was comparable to reference MM patients (23.2 months [95% CI: 21.2–26.2]
vs. 23.3 months [95% CI: 22.5–24.8], p=0.800). Median OS was comparable to reference
patients (52.6 months [95% CI: 46.3–62.0] vs. 55.0 months [95% CI: 51.6–58.9], p=0.504).
By multivariate analysis, adjusted for ISS, we found high levels of BM PCs (>10%)
together with 3 and more EMD lesions as an independent risk factor for inferior survival
in primary EMD patients (PFS: HR 1.92 [95 % CI: 1.28–2.87]; OS: HR 2.06 [95 % CI:
1.35–3.14], both p=0.001).
Summary/Conclusion: We found primary EMD patients distinct from reference MM patients.
Survival intervals of both groups of patients were comparable. We found that the intra-
and extramedullary tumor burden is an independent key factor influencing primary EMD
prognosis.
This work was supported by grant AZV NU21-03-00076.
P968: ADDITION OF IXAZOMIB TO POMALIDOMIDE AND DEXAMETHASONE IMPROVES PROGRESSION-FREE
SURVIVAL FOR MULTIPLE MYELOMA PATIENTS PROGRESSING ON LENALIDOMIDE AS PART OF 1ST
LINE THERAPY: ALLIANCE A061202
P. Voorhees1,*, V. Suman2, Y. Efebera3, N. Raje4, S. Tuchman5, C. Rodriguez6, K. Santo2,
M. Bova-Solem7, D. Carlisle7, U. Saad8, P. McCarthy9, P. Richardson10
1Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute,
Atrium Health, Charlotte; 2Alliance Statistics and Data Management Center, Mayo Clinic,
Rochester; 3Department of Hematology and Oncology, Ohio Health, Columbus; 4Division
of Hematology and Oncology, Massachusetts General Hospital, HArvard Medical School,
Boston; 5Division of Hematology, Lineberger Comprehensive Cancer Center, the University
of North Carolina, Chapel Hill; 6Tisch Cancer Institute, Mount Sinai School of Medicine,
New York City; 7Protocol Operations Program, Alliance for Clinical Trials in Oncology,
Chicago; 8Division of Hematologic Oncology, Memorial Sloan Kettering Cancer Center,
New York City; 9Transplant and Cellular Therapy Program, Roswell Park Comprehensive
Cancer Center, Buffalo; 10Department of Hematology and Oncology, Dana Farber Cancer
Institute / Partners Cancer Care, Harvard Medical School, Boston, United States of
America
Background: Lenalidomide (LEN) maintenance and continuous LEN-based induction therapy
until disease progression have become standard of care for frontline therapy of multiple
myeloma (MM). As such, an increasing number of patients (pts) in need of 2nd line
therapy have LEN-refractory disease. Optimal treatment in this setting has not been
rigorously assessed in randomized studies. The phase I portion of Alliance A061202
demonstrated the safety of the ixazomib-pomalidomide-dexamethasone (IXA-POM-DEX) combination
for the treatment of pts with LEN and proteasome inhibitor (PI)-refractory MM.
Aims: In the randomized phase II portion, we evaluated the addition of IXA to POM-DEX
for PI naïve / sensitive pts progressing on LEN as part of 1st line therapy. The primary
endpoint was progression-free survival (PFS). Key secondary endpoints included overall
response rate (ORR), depth of response, survival and safety.
Methods: Pts were randomized 1:1 to IXA-POM-DEX or POM-DEX and stratified by prior
bortezomib exposure, International Staging System stage (1 and 2 vs 3) and the presence
of high-risk cytogenetics. POM was administered at 4 mg on days 1 – 21; IXA 4 mg on
days 1, 8 and 15; and DEX 20 mg (>75 years (yrs)) or 40 mg (≤75 yrs) on days 1, 8,
15 and 22 of a 28-day cycle. Treatment was continued until disease progression, the
emergence of unacceptable side effects or withdrawal of treatment consent.
Results: 38 and 39 eligible pts were assigned to IXA-POM-DEX and POM-DEX, respectively.
The median age was 66 yrs (range 41 – 83) and 64 yrs (range 52 – 85). A planned first
interim analysis was conducted after 43 out of 57 required events had occurred. PFS
favored the IXA-POM-DEX arm (one-sided log rank test value = 4.6345, p=0.03134 [<
p-value boundary of 0.058]), yielding a hazard ratio of 0.528 (upper 90% bound = 0.777).
A stratified log-rank test found that PFS was superior for the triplet after adjusting
for stratification factors (one-sided stratified log rank test value = 5.8371; p=0.0157),
adjusted hazard ratio 0.451 (upper 90% bound = 0.694). The ORR favored IXA-POM-DEX
(63.2% vs 43.6%, p=0.0853), and the ≥very good partial response was 26.3% vs 5.1%,
respectively (p=0.01). The clinical benefit rate (ORR + minimal response rate) was
73.7% and 56.4%. The most common grade 3/4 adverse events included lymphopenia, neutropenia,
anemia, and fatigue in 40%, 37%, 16% and 16% of IXA-POM-DEX-treated pts and 26%, 21%,
13%, and 15% of POM-DEX-treated pts. Therapy was discontinued for disease progression
in 47.4% of pts on IXA-POM-DEX and 76.9% of pts on POM-DEX and for adverse events
in 7.9% and 7.7% of pts, respectively.
Summary/Conclusion: The addition of IXA to the POM-DEX backbone improved the depth
of response and PFS for pts relapsing on LEN as part of 1st line therapy. Hematologic
toxicity was increased with the addition of IXA, but side effects were manageable.
The ease of administration of this all-oral combination allowed for safer, uninterrupted
treatment during the COVID pandemic. Our results should be confirmed in phase III
trials but lend support for this regimen as part of 2nd line therapy for this patient
population.
P969: COMPARISON OF VENOUS THROMBOEMBOLISM INCIDENCE IN MULTIPLE MYELOMA PATIENTS
RECEIVING LENALIDOMIDE-BASED REGIMENS WITH RIVAROXABAN OR ASPIRIN THROMBOPROPHYLAXIS
Y. Wang1, X. Tang2, W. Zheng1, Z. Wang1, J. Xu1, X. Tan2, Y. Tian2, R. Xu1, S. Cui1,*
1Affiliated Hospital of Shandong University of Traditional Chinese Medicine; 2Shandong
University of Traditional Chinese Medicine, Jinan, Shandong, China
Background: Thromboprophylaxis is routinely used with lenalidomide-based regimens
in multiple myeloma because of a substantial risk of venous thromboembolism (VTE).
Aspirin is one of the most common thromboprophylaxis agent but also increases bleeding
complications.Rivaroxaban represents a selective direct inhibitor of activated coagulation
factor X (FXa) having peroral bioavailability and prompt onset of action which can
mitigate the risk of higher incidence of VTE without an observable increase in bleeding
rates.However, little is known about the incidence of VTE in lenalidomide-based regimens
with rivaroxaban thromboprophylaxis.
Aims: The purpose of our study was to compare of venous thromboembolism incidence
in multiple myeloma patients receiving lenalidomide-based regimen with rivaroxaban
or aspirin thromboprophylaxis.
Methods: We conducted a single-centre retrospective study designed to assess the rates
of VTE, including pulmonary embolism (PE) and deep vein thrombosis (DVT), in MM patients
receiving lenalidomide-based regimen with aspirin or rivaroxaban thromboprophylaxis.A
total of 130 patients who were not anticoagulated for other clinical indications and
received at least 1 cycle of lenalidomide-based regimen between January 2015 and December
2021 were included. Clinical bleeding events after treatment were also important indicators
in our study.
Results: The incidence of venous thromboembolism in the rivaroxaban group was significantly
lower than that in the aspirin prevention group (P<0.05). The incidence of bleeding
in the rivaroxaban group was also lower than that in the aspirin prevention group,but
there was no statistical significance(P﹥0.05).
Table II.
Analysis of VTE and Bleeding.
Regimen + prophylaxis
R-based regimens+ASA(n=84)
R-based regimens+XA(n=46)
P-value
VTE
13(15.5%)
1(2.2%)
0.041
Bleeding
6(7.1%)
0
0.081
ASA, aspirin; XA, rivaroxaban; VTE, venous thromboembolism.
Image:
Summary/Conclusion: In this pilot study, we found that rivaroxaban was safe and well-tolerated
as the primary preventive measure for venous thromboembolism in patients with multiple
myeloma receiving lenalidomide-based regimen. However, these findings warrant further
investigation in a larger randomized study to be validated.
P970: SAFETY AND EFFICACY OF BCMA-CAR-T IMMUNE CELLS DERIVED FROM CORD BLOOD IN THE
TREATMENT OF RELAPSED AND REFRACTORY MULTIPLE MYELOMA
Y. Wang1, X. Hu1, Q. Gao1, H. Wang1, Y. Gao1,*, W. Zhang1, X. Ru1, L. Hou1, W. Zhou1,
H. Zhang1, Y. Zhang1, F. Wang2, X. Li3, F. Guan3
1Department of Hematology, Shaanxi Provincial People’s Hospital, Xi’An; 2School of
Medicine, Shanghai Jiao Tong University, Shanghai; 3School of Medicine, Northwest
University, Xi’An, China
Background: Multiple myeloma (MM) is the second most prevalent hematologic malignancy.
The prognosis of MM patients varies, with low-risk patients surviving more than 10
years with standard care and high-risk patients surviving less than 1 year. CAR-T
therapy shows promising clinical results on relapsed or refractory MM (R/RMM) patients.
However, generating clinically significant doses of CAR-T cells from heavily pre-treated
patients for autologous CAR-T therapy is not always possible. Although allogeneic
CAR-T cells therapy could potentially overcome such limitations, it poses a considerable
risk of graft-versus-host disease (GVHD). Cord blood (CB) is a convenient source of
T cells with clear advantages. T cells in CB have low immunogenicity and a relatively
low risk of GVHD. Here we present a novel approach to CAR-T cells production from
CB, which we hope could overcome the constraints mentioned above.
Aims: The study aims to evaluate the safety and efficacy of the CB-generated BCMA-CAR-T
infusion in R/RMM patients.
Methods: This study is based on a clinical investigation during Jan 2021 and Dec 2021
in Shaanxi Provincial People’s Hospital. In this study, CD3+ T cells from the CB samples
were selected, activated, and modified by lentivirus to produce anti-BCMA CAR-T cells.
The cells were administrated intravenously to patients with R/RMM after expanding
for 5-10 days in vitro. 2-3 days after being administered the lymphodepleting chemotherapy
regimen of cyclophosphamide and fludarabine, patients received CAR-T cells infusion.
We followed the occurrence of adverse events, the level of inflammatory cytokine concentration,
CAR-T cells expansion in the peripheral blood, serum monoclonal protein levels, and
the proportion of plasma cells in bone marrow smears after CAR-T cells infusion.
Results: This study included 11 patients with R/RMM (7 males and 4 females). The median
age was 58 years (ranging from 44 to 65). 18.2% (2/11) of the patients had received
other CAR-T therapies and relapsed before the enrolment. 72.73% (8/11) of the patients
had the extramedullary disease (EMD). 63.64% (7/11) of the patients had received autologous
hematopoietic stem cell transplantation (Auto-HSCT). All patients received CAR-T infusion
at the average dose of 7.11 (4.4-15) ×106/kg. After infusion, 18.2% (2/11) of the
patients had a fever lasting for 48 hours, and 18.2% (2/11) of them had an increase
in heart rate. Nausea and diarrhea occurred in 18.2% (2/11) of the patients. No patients
had transient consciousness disorder. No patients received dexamethasone to relieve
symptoms. CAR-T cells had expanded in all patients, with no increase in the levels
of IL-6, IL-8 and IFN-r in peripheral blood in 2 weeks after infusion. Grade 1 cytokine
release syndrome (CRS) was found in 2 patients. No patients had GVHD after infusion.
The ORR was 54.5% (6/11), with 18.2% (2/11) of the patients achieving CR, and 36.4%
(4/11) achieving PR. Except 1 female died from severe pulmonary infection, other 10
patients were followed up for more than 1 month. Unfortunately, 1 patient who reached
CR relapsed during 3 months of follow-up. Those 4 patients who didn’t achieve PR received
autologous CAR-T therapy afterwards and had responses better than PR (1 CR, 1 VGPR,
2 PR).
Summary/Conclusion: BCMA-CAR-T cells manufactured from CB show a promising safety
profile and certain clinical efficacy for R/RMM patients who are not eligible for
autologous CAR-T cells therapy. We also found allogeneic CAR-T cells manufactured
from CB could prepare patients for later subsequent autologous CAR-T therapy.
P971: ADJUSTED COMPARISON OF PATIENT REPORTED OUTCOMES FROM CARTITUDE-1 AND LOCOMMOTION
COMPARING CILTACABTAGENE AUTOLEUCEL VERSUS REAL WORLD CLINICAL PRACTICE IN TRIPLE-CLASS
EXPOSED MULTIPLE MYELOMA
K. Weisel1,*, M.-V. Mateos2, L. Vincent3, T. Martin4, J. G. Berdeja5, A. Jakubowiak6,
S. Jagannath7, Y. Lin8, P. Thilakarathne9, F. Ghilotti10, J. Diels9, B. Haefliger11,
C. Hague12, A. Gonzalez11, J. M. Schecter13, K. S. Gries14, V. Strulev15, T. Nesheiwat16,
L. Pacaud16, H. Einsele17, P. Moreau18
1Oncology, Hematology and BMT, University Medical-Center Hamburg-Eppendorf, Hamburg,
Germany; 2CIC, University Hospital of Salamanca/IBSAL, Salamanca, Spain; 3Clinical
Haematology Department, Montpellier University Hospital, Montpellier, France; 4UCSF
Helen Diller Family Comprehensive Cancer Center, San Francisco; 5Sarah Cannon Research
Institute, Nashville; 6University of Chicago, Chicago; 7Mount Sinai Medical Center,
New York; 8Mayo Clinic, Rochester, United States of America; 9Health Economics, Market
Access & Reimbursement, Janssen Pharmaceutica NV, Beerse, Belgium; 10Health Economics,
Market Access & Reimbursement, Janssen-Cilag SpA, Cologno Monzese, Italy; 11Health
Economics, Market Access & Reimbursement, Cilag GmbH International, Zug, Switzerland;
12Health Economics, Market Access & Reimbursement, Janssen, High Wycomb, United Kingdom;
13Research and Development, Janssen R&D, Raritan; 14Research and Development, Janssen
R&D, Los Angeles, United States of America; 15EMEA Medical Affairs, Janssen Pharmaceutica
NV, Beerse, Belgium; 16Legend Biotech USA, Piscataway, United States of America; 17Medizinische
Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany; 18Clinical
Hematology, University Hospital Hotel-Dieu, Nantes, France
Background: Adjusted comparisons of the single-arm CARTITUDE-1 clinical trial of ciltacabtagene
autoleucel (cilta-cel) and the prospective LocoMMotion study of therapies used in
real-world clinical practice (RWCP) have shown the clinical benefits of cilta-cel
on response and survival outcomes in patients with triple-class exposed multiple myeloma
(TCE-MM). Yet, patients with TCE-MM have a reduced health-related quality of life
(HRQoL) compared with age- and sex-matched populations.
Aims: To present adjusted comparisons of patient-reported outcomes (PROs) from TCE-MM
patients who received cilta-cel in CARTITUDE-1 vs. RWCP in LocoMMotion.
Methods: EQ-5D-5L, EORTC QLQ-C30 and EORTC QLQ-MY20 questionnaires were administered
to all patients in the CARTITUDE-1 Phase 2 and LocoMMotion studies at baseline, day
7, day 28 and every 4 weeks up to 52 weeks. Mixed model repeated measures (MMRM) analyses
were performed to analyze changes from baseline (CFB) for each patient cohort and
the difference in CFB between cilta-cel and RWCP over time. MMRM models included the
baseline PRO score and prognostic characteristics as covariates to balance patient
cohorts and to adjust for confounding bias. A sensitivity analysis was implemented
assigning worst PRO values to patients who dropped out of the analyses due to death.
Results: A total of 61 patients in CARTITUDE-1 and 202 patients in LocoMMotion had
PRO assessments at baseline and during follow-up. At day 7, PRO scores worsened versus
baseline in both cohorts, and worsening was more pronounced for cilta-cel in physical,
role and social functioning, fatigue and lack of appetite and constipation, coinciding
with short term adverse events associated with the cilta-cel infusion and lymphodepleting
chemotherapy. From the next assessment at week 4 onwards, PRO values significantly
improved over time for cilta-cel patients versus baseline, while improvement from
baseline was lower for RWCP for most domains and symptoms. The average improvement
versus baseline from week 4 onwards, represented by the absolute difference in CFB
between both patient cohorts, was significantly in favor of cilta-cel for Visual Analogue
Scale (8.0), Global health status (8.5), pain (-11.4), dyspnea (-8.9), constipation
(-8.3), future perspective (16.5) (all p<0.01), emotional functioning (7.4) and feeling
restless or agitated (-7.2) (p<0.05) (Table). All other PROs were numerically in favor
of cilta-cel across the follow-up period, except for nausea and vomiting and lack
of appetite, for which small differences in improvement are numerically in favor of
RWCP. The sensitivity analyses assigning worst PRO values to patients who dropped
out of the main analyses due to death illustrate that these results are inherently
biased against the more effective treatment on survival and underestimate the PRO
benefit for cilta-cel.
Image:
Summary/Conclusion: Patients with TCE-MM treated with cilta-cel demonstrated significant
improvements vs. RWCP increasing over time in multiple PRO endpoints. These findings
indicate that cilta-cel can significantly improve patients’ HRQoL in addition to significant
efficacy benefits on response, progression free and overall survival and can help
address unmet patient needs.
P972: INDIRECT COMPARISON OF TECLISTIMAB IN MAJESTEC-1 VERSUS PHYSICIAN’S CHOICE OF
THERAPY IN LONG-TERM FOLLOW-UP OF TRIPLE-CLASS EXPOSED RELAPSED/REFRACTORY MULTIPLE
MYELOMA IN DARATUMUMAB TRIALS
K. Weisel1,*, A. Chari2, S. Z. Usmani3, H. Goldschmidt4, M.-V. Mateos5, K. Qi6, A.
Londhe6, S. Nair7, X. Lin8, L. Pei9, E. Ammann10, R. Kobos9, J. Smit11, T. Parekh12,
M. Slavcev10, P. Moreau13
1University Medical Center Hamburg-Eppendorf, Hamburg, Germany; 2Mount Sinai School
of Medicine; 3Memorial Sloan Kettering Cancer Center, New York, United States of America;
4University Hospital Heidelberg and National Center of Tumor Diseases, Heidelberg,
Germany; 5University Hospital of Salamanca/IBSAL/CIC/CIBERONC, Salamanca, Spain; 6Janssen
Research & Development, Titusville, United States of America; 7Janssen Pharmaceutica
NV, Beerse, Belgium; 8Janssen Global Services, Horsham; 9Janssen Research & Development;
10Janssen Global Services, Raritan; 11Janssen Research & Development, Spring House;
12Janssen Research & Development, Bridgewater, United States of America; 13Hematology
Clinic, University Hospital Hotel-Dieu, Nantes, France
Background: Teclistamab (tec), a B-cell maturation antigen × CD3 bispecific antibody,
is currently being investigated in MajesTEC-1, a phase 1/2, single-arm trial (NCT04557098)
in patients with triple-class exposed relapsed/refractory multiple myeloma (TCE RRMM)
who had received ≥3 prior lines of therapy (LOT) and previously exposed to an immunomodulatory
agent, a proteasome inhibitor, and an anti-CD38 monoclonal antibody. Currently, there
are no head-to-head trials comparing the efficacy of tec versus physician’s choice
of therapy.
Aims: To evaluate the comparative efficacy of tec versus physician’s choice of therapy.
Methods: Patients in the long-term follow-up of 4 clinical trials of daratumumab (CASTOR,
POLLUX, EQUULEUS, and APOLLO) who met the eligibility criteria for MajesTEC-1 were
used to create an external control arm for MajesTEC-1. After discontinuation of trial
treatments, patients (N=427) subsequently received physician’s choice of therapy,
with disease progression and best treatment response assessed by the investigator.
The MajesTEC-1 cohort included individual patient-level data from patients who received
tec (1.5 mg/kg weekly) at a clinical cutoff of Sep 7, 2021. Inverse probability of
treatment weighting with average treatment effect on the treated population was used
to adjust for imbalances in baseline covariates of prognostic significance such as
International Staging System stage, number of prior LOT, extramedullary plasmacytoma,
time since diagnosis, age, hemoglobin, cytogenetic risk, refractory status and progression
on last LOT. Efficacy assessments included overall response rate (ORR), rate of complete
response or better (≥CR), rate of very good partial response or better (≥VGPR), progression-free
survival (PFS), time to next treatment (TTNT), and overall survival (OS) were assessed.
For binary endpoints (ORR, ≥CR rate, ≥VGPR rate), odds ratio (OR) along with 95% confidence
interval (CI) derived from a weighted logistic regression analysis was used to estimate
the relative effect of tec vs physician’s choice of therapy. Hazard ratios (HRs) and
95% CIs were computed using a weighted Cox proportional hazards model for time-to-event
endpoints (PFS, OS, and TTNT). Multiple sensitivity analyses were also conducted.
Results: Baseline characteristics were comparable between the 2 cohorts after applying
inverse probability of treatment weighting. Patients treated with tec had improved
outcomes vs physician’s choice of therapy: ORR (OR 4.58; 95% CI 2.83–7.53; P <0.0001);
≥CR rate (OR 12.62; 95% CI 5.20–38.55; P<0.0001); ≥VGPR rate (OR 11.64; 95% CI 6.49–21.98;
P<0.0001); PFS (HR 0.62; 95%CI 0.45–0.84; P=0.0024); TTNT (HR 0.38; 95% CI 0.27–0.52;
P<0.0001); and OS (HR 0.47; 95% CI 0.32–0.69; P=0.0001). All sensitivity analyses
showed similar results.
Summary/Conclusion: In the present analysis, improved efficacy in all clinical outcomes
was observed with tec versus physician’s choice of therapy, highlighting the therapeutic
potential of tec to address unmet needs in patients with TCE RRMM who received ≥3
prior LOT.
P973: REAL-WORLD STUDY ON ADOPTION OF STANDARD OF CARE (SOC) FOR FRONTLINE TRANSPLANT-ELIGIBLE
MULTIPLE MYELOMA (FLTEMM) PATIENTS BETWEEN 2017 AND 2020/2021 ACROSS FRANCE, GERMANY,
SPAIN, AND ITALY
K. Weisel1,*, A. Wadlund2, G. Gungor3, E. Dergarabetian4, C. Pacheco5, N. Masurkar5,
P. Rodriguez-Otero6
1Oncology, Hematology and BMT, University Medical Center Hamburg-Eppendorf, Hamburg,
Germany; 2Janssen, Janssen-Cilag AB, Kolonnvägen 45, Solna, Sweden; 3Janssen, Kavacik,
Keçeli Plaza, Ertürk Sk. No:13, 34810 Beykoz, Istanbul, Turkey; 4Janssen-Cilag Limited,
50-100 Holmers Farm Way, High Wycombe, Buckinghamshire, United Kingdom; 5Cerner Enviza,
France SAS, 198 avenue de France, Paris, France; 6Department of Hematology, Clinica
Universidad de Navarra, Pamplona, Spain
Background: Newer therapies for frontline transplant-eligible multiple myeloma (FLTEMM)
are underway, but an accurate and updated overview of standard MM treatment management
is lacking. Repeated analyses and real-world data are warranted to describe current
MM standard of care (SoC), treatment lines, and patient clinical outcomes in daily
practice.
Aims: This non-interventional, cross-sectional, retrospective observational database
study described the current SoC for FLTEMM patients in France, Germany, Spain, and
Italy, and recorded the evolution in regimen adoption in distinct elements of frontline
treatment (induction, consolidation, and maintenance) during 2017-2020/2021.
Methods: Clinical information on ongoing (population I) or previous (population II)
FLTEMM patients was extracted from the Cancerology database. The primary objective
was to describe the treatment regimen considered current SoC for FLTEMM management
in all four countries using study population I, and to study the evolution in the
use of different regimens in induction phase using study population II. The secondary
objective was to describe clinical outcomes, including best tumor response and minimal
residual disease (MRD) status, associated with each induction regimen in study population
II.
Results: In study populations I and II, the median (min, max) age of the patients
at induction initiation was 59.0 (35.0, 86.0) and 60.0 (23.0, 80.0) years and 62.9%
(227/361) and 64.8% (287/443) patients were male, respectively. In study population
I, the most common induction regimens were bortezomib/lenalidomide/dexamethasone (VRd)
in France (75.3%, 33/44) and Spain (44.1%, 53/120), bortezomib/thalidomide/dexamethasone
(VTd) in Italy (65.2%, 76/116), and others in Germany (58.9%, 48/81) (including daratumumab-based
[19.8%, 16/81] or not). Maintenance was ongoing or planned for 78.3% (34/44), 62.3%
(51/81), 65.2% (78/120), and 61.4% (71/116) patients in France, Germany, Spain, and
Italy, respectively. Among study population I patients on an ongoing maintenance regimen,
lenalidomide was received by 99.6% (20/20) patients in France, 86.8% (34/40) in Germany,
80.2% (45/56) in Spain, and 90.7% (30/33) in Italy. In study population II, VRd use
as 1L induction increased from 27.0% (10/36) in 2017 to 65.7% (12/18) in 2019 in France
and was relatively low in Germany, Spain, and Italy. The most common 1L induction
regimen in Germany was VCd, but its use decreased from 85.2% (31/37) in 2017 to 64.1%
(24/38) in 2019. VTd was the most common 1L induction in Spain and Italy, but its
use declined from 58.3% (16/27) and 72.4% (34/47) in 2017 to 17.3% (4/22) and 52.8%
(19/36) in 2019, respectively. In total, 37.5% (166/443) study population II patients
achieved at least a complete response, which was the highest for those on VRd (59.2%
versus 34.3% for VTd, 33.4% for VCd, and 33.7% for other non-daratumumab-based regimens,
respectively). In total, 42.5% (188/443) study population II patients were tested
for MRD status and 18.2% (81/443) were found negative. MRD testing rate varied from
26.2% in France to 14.8% in Germany, 84.4% in Spain, and 47.7% in Italy.
Summary/Conclusion: The use of bortezomib triplets in induction varied markedly over
time and between selected countries. Despite not being approved by the European Medicines
Agency specifically for FLTEMM patients, VRd use tended to increase with time in France
and to a lesser extent in Spain and Italy in this population.
P974: THE CLINICAL CHARACTERISTICS AND PROGNOSIS OF PATIENTS WITH PRIMARY PLASMA CELL
LEUKEMIA (PPCL) UNDER THE NEW IWMG DEFINITION CRITERIA
W. Yan1,*, J. Xu1, H. Fan1, J. Liu1, L. Li1, L. Qiu1, G. An1
1State Key Laboratory of Experimental Hematology, National Clinical Research Center
for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy
of Medical Sciences & Peking Union Medical College, Tianjin, China
Background: Primary plasma cell leukemia has been described as a distinct entity of
plasma cell disorders with special features and poor prognosis. In 2021, IMWG proposed
that revised the diagnostic criteria of PCL as CPCs ≥ 5% by peripheral blood (PB)
smear. As the new diagnostic standard has been established recently, it is of great
significance that illustrate the clinical characteristics and outcomes of the new
pPCL cohort.
Aims: Given the limited data for the rare entity, we performed a large retrospective
analysis about the clinical characteristics, survival outcomes, and risk factors for
the new-defined pPCL patients treated in our hospital.
Methods: We conducted a retrospective analysis of 158 pPCL patients diagnosed from
2000 to 2019 in our hospital. This pPCL cohort was redefined from NDMM patients who
appear CPCs ≥ 5% by morphologic evaluation of their PB smear. And we compared them
to a control group with 485 NDMM patients. Statistical analyses were performed using
the R version 4.1.2.
Results: The characteristics and comparisons between pPCL patients and NDMM group
are depicted in Table 1.In general, bone marrow suppression and adverse prognostic
biomarkers (ie, anemia, thrombocytopenia, elevated LDH, hypodiploidy, and high-risk
cytogenetics) were more common in pPCL patients compared with NDMM patients (P<0.05).
For 130 pPCL patients whose treatment and prognosis data were available, with a median
follow-up time of 54.7 months, the median PFS and OS were 16.9 months and 30.0 months
respectively, both significantly longer than the control NDMM patients (P<0.001).
Interestingly, we find the cohort who attended after 2007 got obvious longer PFS than
those who received treatment in 2000-2006(20.6 vs 10.0 months, P=0.017), which showed
a similar result in OS (31.6 vs 16.0 months, P=0.034).
A Cox-regression multivariate analysis was performed among the baseline variables.
The presence of hypodiploidy and elevated serum LDH were found to be prognostic for
worse PFS, whereas age>60 and elevated LDH were the independent predictors for worse
OS. As for the cytogenetic aberrations, did not play an important role in the outcome
of pPCL patients other than the presence of del(17p) which could impact OS in the
univariate analysis.
There are 98 patients who had response data known, with 80(81.6%) patients achieving
objective response in the first-line treatment, 56.4%≥VGPR, and 38.9% CR. When stratifying
the cohort by the best response, the median PFS for patients achieved: NR, PR, and
≥VGPR were 2.4, 11.2, and 31.0 months, respectively (P<0.001). The OS for patients
who had ever achieved NR, PR, and≥VGPR were 2.4, 24.9, and 62.1 months, respectively
(P<0.001). Then, we divided the cohort who had achieved≥PR into early response group
(≤2 courses), intermediate response group (3-4 courses), and late response group (>4
courses). Whereas, there is no statistical difference in the prognosis of 3 groups
with diverse remission speed rates (PFS: P=0.35; OS: P=0.77). We also found that achieving
deep remission was the best independent favorable predictor for long survival in the
multivariate analysis.
Image:
Summary/Conclusion: In conclusion, primary plasma cell leukemia (pPCL) defined by
the new revised diagnostic criteria (CPCs≥5%), remains an aggressive disease characterized
with poor prognosis despite the advancement of treatment regimens. And achieving deep
remission (≥VGPR) in the first-line therapy predicts the best prognosis regardless
of response rate.
P975: INTERIM ANALYSIS OF PHASE II STUDY OF DARATUMUMAB IN COMBINATION WITH BORTEZOMIB
AND DEXAMETHASONE IN PATIENTS WITH MULTIPLE MYELOMA WHO RECEIVED 1 PRIOR LINE OF THERAPY
(KMM1906)
K. H. Yoo1,*, G. W. Gang2, J. H. Yi2, M. K. Kim3, H. J. Kim4, S.-H. Kim5, J. S. Park6,
J.-J. Lee7, C.-K. Min8, J. H. Lee1, D. Cho9, K. Kim10
1Division of Hematology, Department of Internal Medicine, Gachon University Gil Medical
Center, Gachon University College of Medicine, Incheon; 2Division of Oncology and
Hematology, Department of Internal Medicine, Korea University Medical Center, Seoul;
3Department of Internal Medicine, Yeungnam University College of Medicine, Daegu;
4Division of Hematology/Oncology, Department of Medicine, Hallym University Sacred
Heart Hospital, Hallym University College of Medicine, Anyang; 5Department of Internal
Medicine, Dong-A University College of Medicine, Busan; 6Department of Hematology-Oncology,
Ajou University School of Medicine, Suwon; 7Department of Hematology-Oncology, Chonnam
University Hwasun Hospital, Hwasun; 8Department of Internal Medicine, Seoul St. Mary’s
Hospital, The Catholic University of Korea; 9Department of Laboratory Medicine and
Genetics; 10Division of Hematology and Oncology, Department of Medicine, Samsung Medical
Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
Background: Daratumumab in combination with bortezomib and dexamethasone (DVd) demonstrated
efficacy in heavily pretreated patients with relapsed or refractory multiple myeloma
(MM). In a previous pivotal phase 3 study, DVd showed better efficacy in patients
receiving only first-line treatment, while the limited administration of bortezomib
and dexamethasone for 8 cycles was criticized.
Aims: We conducted a phase 2 study with DVd regimen in patients with MM who received
1 prior line of therapy.
Methods: We evaluated daratumumab (16 mg/kg IV), bortezomib (1.3 mg/m2 SQ), and dexamethasone
(20 mg IV or PO) with maintaining all three drugs after 9 cycles until disease progression
or unacceptable toxicity. The primary endpoint was objective response rate ≥ very
good partial response (VGPR). Secondary objectives included progression-free survival
(PFS), overall survival (OS), safety and tolerability, and minimal residual disease
(MRD)-negativity. MRD was assessed in patients who achieved in stringent complete
response (sCR) or CR, and who maintained VGPR over 6 months, with EuroFlow-based next-generation
flow (NGF).
Results: From June 2020 to June 2021, 26 MM patients who received 1 prior line of
therapy and disease progressed were enrolled at 10 centers in Korea. The median age
of all patients was 72 years (range, 47-85), and 8 patients were male. Five patients
(19%) had high-risk cytogenetic abnormalities (t(4;14), t(14;16), or del17p), and
6 patients (23%) had extramedullary disease. All patients were treated with 1 prior
line, including VTD (N = 11, 42%), VMP (N = 8, 31%), Rd (N = 6, 23%), and CMP (N =
1, 4%). Nineteen patients (73%) and 18 patients (69%) were exposed to bortezomib and
immunomodulatory drugs, respectively, from previous treatment (Table 1). During the
median follow-up period of 11.7 months, 15 patients (58%) maintained treatment, and
11 patients (42%) discontinued the study due to disease progression (N = 6), death
from other causes (N = 2), or withdrawal of consent (N =3). 16 patients (62%) achieved
≥ VGPR (2 sCR, 7 CR, and 7 VGPR). The MRD-negativity (10-5) was 5/12 (42%) (Table
2). The treatment response and PFS of each patient were given in the swimmer plot
(Figure 1). The most common adverse event (AEs) ≥ grade 3 was thrombocytopenia (N
= 6, 23%). Serious AEs were reported in 8 patients (31%). Dose delay or dose reduction
have occurred in 17 patients (65%).
Table 2.
Response and MRD-negativity rates
Response category
No. with response (%)
CR or better
9 (35%)
sCR
2 (8%)
CR
7 (27%)
VGPR or better
16 (62%)
VGPR
7 (27%)
PR
6 (23%)
MR
2 (8%)
SD
2 (8%)
PD
0
MRD-negativity (10-5)
N = 12
Negative
5 (42%)
Image:
Summary/Conclusion: Among patients with MM who received 1 prior line of therapy, DVd
regimen with maintenance strategy showed an acceptable clinical response and MRD-negativity
with manageable toxicity profile.
P976: BENDAMUSTINE-POMALIDOMIDE-DEXAMETHASONE (BPD) FOR RELAPSED AND/OR REFRACTOR
MULTIPLE MYELOMA WITH EXTRAMEDULLARY DISEASE
Z. Yuping1,*, H. Y. Wu1, X. Chu2, X. Deng3, X. Feng4, C. Yuan5, X. Ran6, G. Liu7,
C. Fan8, H. Hao5, X. Zhou1
1Qingdao Municipal Hospital, Qingdao City, Shandong Province; 2The Affiliated Yantai
Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province; 3Weihai
municipal hospital, Weihai City, Shandong Province; 4The Affiliated Hospital of Qingdao
University (West Coast Hospital area), Dongying City, Shandong Province; 5QILU HOSPITAL
OF SHANDONG UNIVERSITY(Qingdao), Qingdao City, Shandong Province; 6Weifang people’s
Hospital, Weifang City, Shandong Province; 7Shengli Oilfield Central Hospital, Dongying
City, Shandong Province; 8Department of Hematology, Qingdao Hospital of Traditional
Chinese Medicine (Qingdao Hiser Hospital), Qingdao City, Shandong Province, China
Background: Extramedullary disease (EMD) is a rare manifestation of multiple myeloma
(MM). The incidence is about 6-20% in the relapsed MM patients. Although novel agents
prolong the survival of patients with MM, patients with EMD are prone to relapse or
progress again easily,EMD may be associated with decreased overall survival in MM.
No standard therapy has been established for this high-unmet need population. Bendamustine
is an old bi-functional alkylating agent which has proved to be effective in Multiple
Myeloma (MM). Pomalidomide is a potent immunomodulatory drug agent with anti-angiogenic,
anti-proliferative, and immunomodulatory activity against MM. Preclinical studies
showed that bendamustine and pomalidomide had higher penetration rate of cerebrospinal
fluid. In China, the price of two drugs is relatively economical. In a phase I/II
study,the combination of BPd demonstrates deep Response(ORR:61%) and has a favorable
tolerance profile in patients with RRMM. To summarize,BPD is an safe,effective, and
economical treatment, and there are no studies of BPD in the regimen of RRMM with
EMD,so it is worth exploring in domestic clinical trials.
Aims: To evaluate the efficacy and safety of bendamustine, Pomalidomide, and dexamethasone
(BPd) for relapsed / refractor multiple myeloma with EMD.
Methods: This is an open-label, multicenter, prospective study(ChiCTR2100048829).Eligible
patients were aged≥18 years, and had a diagnosis of RRMM with EMD, and an Eastern
Cooperative Oncology Group performance status of 0-2,Patients received bendamustine
75 mg/m2 on days 1 and 2(If there is no severe side effects in the first cycle, it
can be increased to 80mg/m2), 6 cycles. Pomalidomide 4 mg on days 1-21,until progression
or intolerable toxicity. Dexamethasone 20mg on days1-4, 15-18, until progression or
unacceptable toxicity. Each cycle is 28days. The primary endpoint is overall response
rates (ORR).
Results: A total of 21 patients were enrolled in the study,12(57.14%) patients with
first relapse multiple myeloma. The median age is 57.4years(47-72). According to IMWG
criteria, all first relapse patients responded(ORR100%)and CR:3pts(25%),VGPR:7pts(58.33%),PR:2pts(16.67%).Patients
with ≥2nd relapese, the ORR was44.44%,≥VGPR:0pts(0%), PR:4pts(44.44%),SD:3pts (33.33%),PD:2pts(22.22%).This
results established that BPD has a deep Response in first relapse MM with EMD.The
1-year PFS was79.16%of all patients.The most common grade 3 or 4 hematologic toxicities
were neutropenia7(33.33%),thrombocytopenia4(19.05%),anemia 2(9.52%). Overall,the severe
toxicities are manageable.
Table 1.
Patient characteristics
Age, years, median (range)
57.4(47-72)
ECOG performance status(n,%)
ISS disease stage(n,%)
Number of prior therapies,median (range)(n,%)
0
4
19.05%
I
2
9.52%
1
12
57.14%
1
10
47.62%
II
3
14.29%
≥1
9
42.86%
2
7
33.33%
III
16
76.19%
Table 2.
International Myeloma Working Group best response
IMWG
first relapse(n,%)
≥2nd relapese(n,%)
ALL(n,%)
ORR
12
100.00%
4
44.44%
16
76.19%
SCR
0
0.00%
0
0.00%
0
0.00%
CR
3
25.00%
0
0.00%
3
14.29%
VGPR
7
58.33%
0
0.00%
7
33.33%
PR
2
16.67%
4
44.44%
6
28.57%
SD
0
0.00%
3
33.33%
3
14.29%
PD
0
0.00%
2
22.22%
2
9.52%
Image:
Summary/Conclusion: Safety data from the study demonstrates that BPD regimen has a
favorable tolerance profile in patients for RRMM with EMD. Early efficacy is encouraging,
Longer-term results still require more patients have been enrolled and further follow-up.
The combination of BPD will be an effective treatment strategy in RRMM with EMD.
P977: BISPECIFIC CS1-BCMA CAR-T CELLS ARE CLINICALLY ACTIVE IN RELAPSED OR REFRACTORY
MULTIPLE MYELOMA
C. Li1,2, X. Wang1,2, Z. Wu1,2, W. Luo1,2, Y. Zhang1,2, M. Du1,2, C. Lu1,2, H. Kou1,2,
Y. Kang1,2, J. Xu1,2, P. Huang1,2, W. Xiong3, J. Zheng1, J. Deng1, Y. Hu1,2, H. Mei1,2,*
1Institute of Hematology, Wuhan Union Hospital, Tongji Medical College, Huazhong University
of Science and Technology; 2Hubei Clinical Medical Center of Cell Therapy for Neoplastic
Disease; 3Wuhan Sian Medical Technology Co., Ltd, Wuhan, China
Background: BCMA-targeted CAR-T cell therapy has showed high response rates in multiple
myeloma (MM); however, remission is transient in most of patients (pts). Variable
and even absent BCMA expression on MM cells have been documented after single BCMA-targeted
CAR-T cell treatment. CS1 (CD319, SLAMF7) plays a vital role in myeloma pathogenesis,
including promoting MM cells growth, survival and adhesion. CS1 is highly expressed
on tumor cells in almost all MM pts, and is also retained at significant levels at
relapse. Therefore, we propose to augment BCMA targeting with CS1. Bispecific CS1-BCMA
CAR-T cells are effective in targeting MM cells in preclinical studies (Biomedicines
2021, 9, 1422).
Aims: Here we report the outcomes of 13 pts with refractory or relapsed (RR) MM in
our phase I clinical trial (NCT 04662099).
Methods: CS1-BCMA bispecific CAR contained a murine anti-CS1 scFv (clone 7A8D5) and
a murine anti-BCMA scFv (clone 4C8) in tandem, and a 4-1BB costimulatory domain (Figure
1A). The enrolled pts must have received at least 2 prior lines of therapy, and previous
BCMA- or CS1-targeted immunotherapies were allowed. Pts were subjected to lymphodepleting
regimens with cyclophosphamide (250 mg/m2, d-5 to d-3) and fludarabine (30 mg/m2,
d-5 to d-3) daily prior to the CAR-T infusion (d0). Planned dose levels were 0.75,
1.5, and 3.0x106 CAR+T cells/kg, and repeated infusions were allowed. Primary objectives
were incidence of adverse events. Cytokine release syndrome (CRS) and neurotoxicity
were graded using the ASTCT criteria, and other AEs using CTCAE v5.0. Secondary objectives
were overall response rate(ORR), overall survival(OS), duration of response (DOR),
and progress-free survival (PFS). Response was assessed per the IMWG criteria (2016).
Other objectives included in vivo kinetics of CAR-T cells.
Results: As of February 10, 2022, 13 pts received the infusion of CS1-BCMA CAR-T cells
and were included in the final analysis. Six pts (46%) carried cytogenetic abnormalities,
and 3 pts had only extramedullary diseases (EMD) without detectable MM cells in bone
marrow (BM). BCMA and CS1 were highly expressed on MM cells in BM (median BCMA+ 77.2%
and CS1 + 96.5%) and EMD (Figure 1B). Eight pts (62%) were refractory, and median
prior lines of treatment was 5.5 (range 2-10), and 7 pts (54%) received autologous
stem cell transplantation.
Four pts (31%) experienced CRS, and grade 3 CRS occurred once and lasted for 5 days.
Neurotoxicity was not observed. The ORR is 76.9% (10/13), including 4 stringent complete,
2 very good partial, and 4 partial response (Figure 1C). Although CAR-T cells infiltrated
into the tumor tissue, response was not observed in the 3 pts with only EMD (Figure
1D). Soluble BCMA decreased remarkably in peripheral blood (PB) and BM (Figure 1E).
The median follow-up and DOR were 290 days. The median OS and PFS weren’t reached,
and OS and PFS at 9-month were 77.1%.
On day 14 after infusion, CAR-T cells peaked at 464842 copies/ug DNA in PB (n=13)
and 282920 copies/ug DNA in BM (n=10) by droplet digital PCR; CAR-T cell expanded
at 400 CAR+T cells/ul PB (n=13) and 271 CAR+T cells/ul BM (n=10) by flow cytometry.
CAR was detected in vivo at a median of 5 months. Moreover, we systematically evaluated
the immune characteristics of infused products (Figure 1F). Limited by small sample
size, correlation analysis wasn’t done
Image:
Summary/Conclusion: Our study demonstrates bispecific CS1-BCMA CAR-T cells are clinical
active with a good safety profile in heavily pretreated patients with MM.
P978: IXAZOMIB VERSUS LENALIDOMIDE OR IXAZOMIB AND LENALIDOMIDE COMBINATION AS MAINTENANCE
REGIMEN FOR PATIENTS WITH MULTIPLE MYELOMA: INTERIM ANALYSIS OF A MULTI-CENTER PROSPECTIVE
STUDY IN CHINA
Z. Zhuang1, Y. Tian2, H. Yu3, L. Shi4, W.-W. Tian5, Q. Liu6, D. Zou7, F. Dong8, Y.
Ma9, R. Feng10, S. Liu1, H. Liu10, H. Jing8, W. Sun7, L.-M. Ma5, L. Bao4, R. Fu3,
Y. Wu2, W. Chen2, J. Zhuang1,*
1Department of Hematology, Peking Union Medical College Hospital; 2Department of Hematology,
Beijing Chao-Yang Hospital of Capital Medical University, Beijing; 3Department of
Hematology, Tianjin Medical University General Hospital, Tianjin; 4Department of Hematology,
Beijing JishuitanHospital, Beijing; 5Department of Hematology, Shanxi Bethune Hospital
of Shanxi Medical University, Taiyuan; 6Department of Hematology, The First Affiliated
Hospital of Anhui Medical University, Hefei; 7Department of Hematology, Xuanwu Hospital
of Capital Medical University; 8Department of Hematology, Peking University Third
Hospital, Beijing; 9Department of Hematology, The second Hospital of Shanxi Medical
University, Taiyuan; 10Department of Hematology, Beijing Hospital, National Center
of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science,
Beijing, China
Background: Maintenance therapy (MT) deepens response and prolongs progression free
survival (PFS) in patients with newly diagnosed multiple myeloma (NDMM) after frontline
regimens. Ixazomib, a 2nd generation oral proteasome inhibitor (PI), has been approved
for MT because of its convenience and tolerability.
Aims: We conducted this prospective multi-center study to compare the efficacy and
safety of Ixazomib (I-MT) or Ixazomib plus Lenalidomide (IL-MT) to Lenalidomide (L-MT)
as maintenance regimen in NDMM patients.
Methods: This study was approved by the Institutional Review Board of Peking Union
Medical College Hospital and registered (NCT04217967). NDMM patients were enrolled
from 10 centers of North China MM Registry since September 2019. After 4 cycles of
front-line induction therapy, patients reached partial response (PR) would receive
autologous stem cell transplantation (ASCT) if eligible, or keep up to 5 cycles of
front regimens if ineligible, then start maintenance therapy. Patients did not reach
PR would switch to 2nd-line induction for 2-5 cycles and start MT once PR was achieved.
For MT, 4mg of Ixazomib was given on day 1,8,15, and 25mg of Lenalidomide every other
day on days 1–21 of 28day cycles. Patients in dual drug group were administrated with
both Ixazomib and Lenalidomide, dose as listed above. The primary endpoint was PFS
from MT.
Results: A total of 176 patients were enrolled, including 58 in I-MT, 72 in L-MT and
46 in IL-MT. The demographic and clinical characteristics, including gender ratio,
age, paraprotein isotype, ISS, R-ISS, were comparable among different MT regimen groups
either receiving ASCT or not at baseline (Table 1). The proportions of patients with
high-risk cytogenetic abnormalities (HRCAs), defined as amplification 1q21, deletion
17p, t(4,14) and t(14,16), were lower in L-MT without ASCT and I-MT with ASCT.
The median follow-up duration since MT was 10.6, 12.6 and 11.1 months in I-MT, L-MT
and IL-MT groups in non-ASCT patients, while 9.3, 14.0 and 12.2 months in ASCT patients,
respectively. In non-ASCT groups, the disease progression rates were 13.5%, 10.2%
and 10.8%, whereas 0, 21.7% and 0 in ASCT groups. The median PFS and OS were not reached
(NR) in all groups. There were 73.1%, 73.4% and 78.3% of the patients reached VGPR
or better before MT in non-ASCT patients, while the rates improved to 80.8%, 81.6%
and 86.5% during follow-up. The depth of response did not change in ASCT patients.
In the whole cohort, the prevalence of peripheral neuropathy (PN) was 18.9% on I-MT,
8.3% on L-MT and 21.7% on IL-MT. Grade 2 PN occurred in 3, 0 and 2 patients, respectively.
The incidence of gastrointestinal (GI) events was 12.1%, 1.4% and 15.2%, respectively.
Grade 3 GI events occurred in 3, 0 and 1 patients. The incidence of grade 3-4 hematologic
toxicities was 1.7%, 4.2%, and 2.2%. Infections developed in 10.3%, 2.8% and 4.3%
patients. No drug withdrawal was related to adverse events.
Image:
Summary/Conclusion: Due to inadequate access to melphalan and low rate of ASCT, the
PFS of NDMM in Chinese patients was relatively inferior. We design this multi-centered
prospective study to evaluate if dual drug MT will further strengthen response and
make up the gap. Though the primary endpoint--PFS has not been reached in all treatment
groups, dual MT improves response most and is quite tolerable.
P979: EXOSOMES IN POLYCYTHEMIA VERA: “MINI PLATELETS“ WITH THROMBOGENIC POTENTIAL
A. Abulafia1,2,*, G. Spectre1,2, E. Ziv3, K. R. Geiger1, Z. Sarsor3, N. Ron2,3, E.
Beery3, S. Revel-Vilk4,5, M. Naamad6, P. Raanani1,2,3, O. Uziel2,3, U. Rozovski1,2
1Institute of Hematology, Rabin Medical Center, Petah Tikva; 2Sackler School of Medicine,
Tel Aviv University, Tel Aviv; 3Felsenstein Medical Research Center, Rabin Medical
Center, Petah Tikva; 4Gaucher Unit, Pediatric Hematology/Oncology Unit, Shaare Zedek
Medical Center; 5Faculty of Medicine, Hebrew University of Jerusalem; 6Flow Cytometry
Unit, Shaare Zedek Medical Center, Jerusalem, Israel
Background: Polycythemia vera (PV) is characterized by clonal proliferation of myeloid
progenitor cells that is caused by activating mutation in the Janus Kinase (JAK)2
gene. Patients with PV tend to develop thrombotic complications across the vasculature
tree. Yet, how PV-clonal cells induce pro-thrombotic environment is still under investigation.
Secreted by all types of cells, exosomes are nano-scaled particles which carry a molecular
cargo that reflects their cell of origin. Exosomes travel in blood, may reach distant
sites and affect various physiological and pathological processes. These features
led us to think that PV-derived exosomes (hereafter PV-exosomes) carry the mutated
JAK2 oncogene and contribute to the thrombotic manifestations seen in PV.
Aims: Determine whether PV-exosomes carry mutated JAK2 transcripts.
Determine the prothrombotic potential of PV-exosomes and their effect on endothelium.
Methods: Exosomes were isolated from eythroleukemia (HEL 92.1.7) cell line (positive
for mutated JAK2) and from patients with PV by serial ultracentrifugation. To confirm
the presence of exosomes, electron microscopy images were obtained and to quantify
their concentration nanoscale tracking analysis was applied (Malvern).
The exosomes were stained with the FM-4-11 lipophilic dye and their uptake by HUVECs
and HaCaT cells was followed by flow cytometry. To determine the presence of JAK2
transcript, a 465 bp long DNA segment within exon14 of JAK2 was amplified by qRT-PCR
and by Sanger sequencing JAK2 mutational status was determined. Thrombin generation
assay (TGA) (Ceveron®, Alpha, Technoclone) was used to assess the thrombotic potential
of PV-exosomes. Platelet activation status was assessed by the expression of PAC-1,
CD-62 and CD-63 in platelets and Trans Epithelial Electric Resistance (TEER) (Electric
cell substrate impedance sensing device, Applied Biophysics) was used to assay endothelial
dysfunction in HUVECs.
Results: We isolated exosomes from the sera of 24 healthy donors and 31 patients with
PV. We found the presence of the mutated JAK2 mRNA in all samples confirming that
the mutated JAK2 transcript is packaged into sera-exosomes. Yet, while healthy donors’
derived exosomes carry only the wild type JAK2 mRNA, we detected both the wild type
and the mutated JAK2 isoforms in patients with PV.
We showed that PV-exosomes induced thrombin generation. Then we compared PV-exosomes
and healthy-individuals’ derived exosomes and found that thrombin generation potential
of PV-exosomes is higher in all measured parameters (p< 0.05 in all parameters). Furthermore,
using various platelet markers (CD62, CD63, PAC1) we confirmed that PV-exosomes induced
platelet activation. Together, these findings suggest that PV-exosomes may promote
thrombosis directly.
Under physiological conditions the integrity of the vascular wall prevents uncontrolled
thrombi. Therefore, we wondered whether PV-exosomes induce endothelial dysfunction,
and in this way contribute indirectly to the pro-thrombotic phenotype. PV-exosomes
were taken up by HUVEC endothelial cells in a dose- and time- dependent manner and
TEER assay revealed that exosomal uptake by HUVECs induced endothelial dysfunction
by breaking the tight junction between adjacent endothelial cells.
Summary/Conclusion: Exosomes may act as “mini platelets” that promote thrombosis in
patients with PV. PV-exosomes may promote thrombosis either directly by activating
platelets and inducing thrombin generation or indirectly by inducing endothelial dysfunction.
P980: JAK2V617F-DEPENDENT DOWN REGULATION OF SHP1 EXPRESSION PARTICIPATES IN THE SELECTION
OF MPN CELLS IN THE PRESENCE OF TGFΒ
C. Aoun1,*, N. Salomao1, N. Maslah1, S. Awan Toor1, G. Letort1, P. Gou1, J.-J. Kiladjian2,
S. Giraudier1, B. Cassinat3
1INSERM U1131-hopital saint louis, Paris, France; 2Centre d’investigations cliniques;
3Laboratoire de biologie cellulaire, INSERM U1131-Hopital Saint Louis, Paris, France
Background: Myeloproliferative neoplasms (MPNs) are characterized by an increased
production of blood cells, due to acquisition in HSC of mutations in the JAK2, CALR
or MPL genes. The JAK2V617F mutation, leading to a constitutively active form of JAK2,
is the most frequent mutation. During disease evolution the clonal selection of mutated
cells is dependent on cell-intrinsic and extrinsic factors present in the niche microenvironment
such as cytokines. TGFβ, whose local secretion is increased in MPNs, and more particularly
in myelofibrosis is known to negatively regulate normal hematopoietic stem cells (HSC)
proliferation. However, its mode of action is not clearly established. We hypothesized
that JAK2V617F mutant HSC are resistant to TGFβ antiproliferative effect explaining
the pathological stem cell invasion in myelofibrosis. Using multiomics approaches
to study the intracellular signaling pathways in MPNs we recently identified the down
regulation of SHP-1 expression in JAK2V617F cells compared to wild type cells. Of
note, the phosphatase SHP-1 has recently been shown to be required for the TGFβ-induced
quiescence of HSCs.
Aims: We hypothesized a relationship between SHP-1 down regulation and the resistance
of JAK2V617F mutated cells to the antiproliferative effect of TGFβ. The work presented
herein aimed at verifying this hypothesis.
Methods: We used the HEL erythroleukemia cell line which carries several copies of
the JAK2V617F mutated gene. We also used an isogenic model obtained by transducing
either the JAK2 wild type (JAK2wt) of JAK2V617F allele in the UT-7 megakaryoblastic
cell line. Proliferation was measured using the CCK8. Protein expression was measured
using western blot while gene expression was measured using RT-qPCR.
Results: The expression of SHP-1 was reduced by 30% at the RNA level and 50% at the
protein level in UT-7 cells transduced with JAK2V617F compared to JAK2wt -transduced
cells. In bone marrow cells taken from JAK2V617F knock-in mice the protein level of
SHP1 was reduced by 70% compared to wild-type mice. EPO treatment of UT-7 cells (which
express the EPO-R) showed a down regulation of SHP-1 expression which was abrogated
in the presence of the JAK2 inhibitor ruxolitinib. These results suggest that SHP-1
expression is modulated by JAK2 signaling leading to its down regulation in JAK2V617F
cells. Although TGFβ receptor and SMAD2/3 expression was similar in both cell type,
JAK2V617F UT-7 cells were less responsive to TGFβ signaling compared to JAK2wt cells
(reduced phosphorylation of SMAD2/3). The proliferation of JAK2wt UT-7 cells was significantly
reduced in the presence of TGFβ 10ng/mL while JAK2V617F UT-7 cells were not affected.
To confirm the involvement of SHP-1 we transduced HEL cells with the cDNA coding for
SHP-1 in order to restore higher levels of the protein in this JAK2V617F mutated cell
line and observed a significant reduction in the proliferation when treated with TGFβ
compared to cells transduced with the empty vector. Finally, we performed an in vitro
competitive assay where JAK2V617F UT-7 cells transduced by an m-cherry fluorescent
vector were mixed 50/50 with JAK2wt UT-7 and cultured with TGFβ. After 14 days of
culture with TGFβ 10ng/mL we observed a significant positive selection of the JAK2
mutant cells over JAK2wt cells.
Summary/Conclusion: In conclusion, we found a JAK2-dependent down-regulation of SHP1
expression which is related to the resistance of JAK2V617F cells to the antiproliferative
effect of TGFβ explaining clonal selection of mutant stem cells in myelofibrosis
P981: NEXT GENERATION SEQUENCING (NGS): AN IMPORTANT TOOL TO CHARACTERIZE MYELOPROLIFERATIVE
DISEASES IN CHILDREN
S. Bianchi1,*, G. Palumbo1, V. Filipponi1, N. Monaco1, G. Pileggi1, R. Maglione1,
M. Rousseau1, M. L. Moleti1, F. Giona1
1Translational and Precision Medicine, Sapienza, University of Rome, Rome, Italy
Background: Mutations in the JAK2, MPL and CALR driver genes are reported in over
90% of adults with BCR-ABL1-negative chronic myeloproliferative neoplasms (MPNs) and
in 22-40% of children, where inherited forms, such as familial erythrocytosis (FE)
and hereditary thrombocytosis (HT), are common. Next Generation Sequencing (NGS) is
useful to identify clonal markers, other than those found using standardized methods.
Aims: This study was carried out in order to: a) confirm the results obtained in our
preliminary experience, using NGS in a larger number of children and adolescents with
myeloproliferative diseases (MPDs); b) identify non-canonical driver and/or non-driver
mutations in patients (pts) with MPDs; c) evaluate long-term outcome of disease in
different subgroups of pts.
Methods: 86 pts (46 male, 41 female; median age at diagnosis: 15 years) with a BM
evaluation, molecular analysis for driver mutations for MPN (JAK2V617F, JAK2 exon
12, CALR, MPL), and genes involved in the FE (HIF2α) and HT (MPL), were studied in
NGS.
Results: Driver mutations were found in 52/87 pts (60%) (16 JAK2V617F, 10 CALR, 21
MPLS505N, 2 MPLV501A, and 3 HIF2α). A 44-NGS gene panel providing diagnostic information
in myeloid malignancies and in rare inherited erythrocytosis/thrombocytosis were performed
in 71/86 pts (82.5%), 28 of them (39.5%) without any mutations. The 44-NGS gene panel
detected non canonical driver mutations in 5/28 (18%) triple negative pts, and additional
non canonical driver mutations in 2 JAK2V617F and CALR type 2 pts. Overall, driver
mutations were found in 61/86 (71%) of pts. The NGS panel revealed 28 additional functional
non-driver mutations (12 mutations in 5 genes of the signaling group, 5 mutations
in 2 genes of the mRNA splicing group, 6 mutations involving the ASXL1 gene of the
histone modification group, and 5 mutations involving 2 genes of the DNA methylation
group) in 29 pts, 16 of them with no driver mutations.
Pts with acquired MPNs were retrospectively classified according to the WHO 2016 criteria;
pts with HIF mutations and/or anamnestic criteria of familial erythrocytosis were
considered as FE, pts with MPLS505N, or MPLV501A mutations were defined as HT (Table1).
One or more non-driver mutations were found in 17/37 (46%) TE, 4/15 (26.5%) PV and
mPV, 6/9 (66.6%) FE pts, and in 0/23 HT pts. During follow-up, 20 pts (12 ET, 5 HT,
2 PV and 1 pre-PMF) presented progressive splenomegaly, after a median of 120 months;
in addition, 10 (6 ET, 3 HT, and 1 pre-PMF) of them developed a ≥2 BM fibrosis, after
a median of 188 months. Considering driver mutations, 8/20 pts were JAK2V617Fmutated,
5 had CALR mutations, and 5 were MPLS505N mutated; 2 pts were wild type. One or more
additional non-driver mutations were present in 3/5 CALR, and in 3/8 JAK2V617Fmutated
pts. Overt MFI developed in 3 pts, 2 of them needed hematopoietic stem cell transplantation.
Two pts developed a malignant neoplasia. Six (7%) pts, 3 MPLS505N, 2 JAK2V617F, and
one wild type, without thrombophilic abnormalities, experienced a thrombotic event.
All pts are alive after a median time of 15 years from initial diagnosis.
Image:
Summary/Conclusion: In our experience, the use of a 44-gene NGS panel enabled us to
identify non canonical driver, and functional non-driver mutations, in addition to
those detected by conventional methods, in about 90% of pediatric pts with acquired
and familial MPDs. Additional clonal mutations have been identified in two thirds
of FE pts, but none in HT pts. Interestingly, the development of BM fibrosis was observed
in pts with HT, but not in FE group.
P982: MYELOID-DERIVED SUPPRESSOR CELLS: CHARACTERIZATION IN PERIPHERAL BLOOD AND SPLEEN
TISSUE OF PATIENTS WITH MYELOFIBROSIS
R. Campanelli1,*, A. Bonometti2,3, M. Massa4, C. Abbà1, L. Villani1, A. Carolei1,
P. Catarsi1, M. Paulli3, G. Barosi1, E. Boveri3, V. Rosti1
1Center for the Study of Myelofibrosis, General Medicine 2—Center for Systemic Amyloidosis
and High-Complexity Diseases, IRCCS Policlinico San Matteo Foundation, Pavia; 2IRCCS
Humanitas Research Hospital, Milan; 3Department of Diagnostic Medicine, Anatomic Pathology
Unit; 4General Medicine 2—Center for Systemic Amyloidosis and High-Complexity Diseases,
IRCCS Policlinico San Matteo Foundation, Pavia, Italy
Background: Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells that
accumulate in patients with malignancies, sepsis or chronic inflammation. Features
of MDSCs are: myeloid origin, immature state, strong ability to reduce cytotoxic functions
of T/NK cells, potential to differentiate into endothelial cells favoring neoangiogenesis.
MDSCs are conventionally divided into polymorphonuclear (PMN)- and monocytic (M)-MDSCs.
PMN-MDSCs are characterized by the production of reactive oxygen species and arginase-1
(Arg-1), that directly inhibits T-cell proliferation and activation, while M-MDSCs
predominantly express inducible nitric oxide synthase and produce nitric oxide.
Myelofibrosis (MF) is characterized by clonal expansion of a hematopoietic stem cell,
extramedullary hematopoiesis, bone marrow (BM) fibrosis, splenomegaly, BM and splenic
neoangiogenesis and an acquired mutation of JAK2, CALR or MPL gene. Inflammation plays
a relevant role in MF pathogenesis, as proven by high levels of inflammatory cytokines
with prognostic significance and by a state of chronic oxidative stress.
Aims: To characterize circulating MDSCs and assess their localization and quantification
in spleen samples from MF patients.
Methods: Peripheral blood mononuclear cells (PBMCs) from MF patients (n=15) and healthy
subjects (HDs; n=10) were stained with surface antibodies (CD11b, CD14, HLA-DR, CD15,
CD33) and intracellular Arg-1 and analysed by flow cytometry. Formalin-fixed paraffin-embedded
samples of spleen specimens (MF n=41; HDs n=20) were immunostained with anti-Arg-1
antibody using the automated platform Dako Omnis Envision Flex with the EnVision FLEX,
High pH, HRP Rabbit/Mouse High pH [GV800] revelation kit.
Results: In PBMCs, the percentage of PMN-MDSCs (identified as CD11b+HLA-DRlow/-CD33dimCD14-CD15+)
was higher (p=0) in patients (median 9.2, range 1-59) than HDs (median 1.7, range
0.1-4.5) and directly correlated with the allelic burden (R=0.86, p=0.006) in JAK2-mutated
patients. Patients in pre-fibrotic phase (n=7) had levels of PMN-MDSCs (median 9.2,
range 1-31.4) comparable to HDs; on the contrary, the percentage of PMN-MDSCs in patients
with BM fibrosis (n=8; median 13.3, range 3.5-59) was higher (p=0) than in HDs. All
PMN-MDSCs were Arg-1+, both in MF and in HDs. Gene expression studies are ongoing.
The expression of the CXCR4 receptor on PMN-MDSCs was lower, although not statistically
significant, in MF (median 1.1, range 0-9.3) than in HDs (median 2.5, range 0-11.9).
PMN-MDSCs in MF inversely correlated (R=-0.89 p=0.007) with CXCR4 expressed on CD34+
cells.
The percentage of M-MDSCs (identified as CD11b+HLA-DRlow/-CD33hiCD14+CD15-) was higher
(p=0.015) in HDs (median 0.49, range 0.25-1.5) than in MF (median 0.29, range 0.02-0.76).
Immunohistochemical staining of spleen sections showed a higher (p=0) percentage of
Arg-1+ cells (calculated on total spleen cells) in MF (median 20, range 5-60) than
in HDs (median 7, range 2-20); Arg-1+ cells were present in all patients and predominantly
located in the red pulp in MF patients, while in HDs were detected in the white pulp.
Summary/Conclusion: The identification of MDSCs and their relation with the mutational
status suggest a new mechanism of MF pathogenesis, either related to the chronic inflammatory
status of patients or as players of the neoangiogenesis that characterizes the disease.
Moreover, the low expression of CXCR4 on PMN-MDSCs could be, on one hand, related
to the low expression of this receptor on CD34+ cells or suggest a MF-specific recruitment
mechanism, other than that observed in solid tumors.
P983: SINGLE CELL ANALYSIS ALLOWS THE EARLY DETECTION OF LEUKEMIC CLONES IN MPN PATIENTS
C. Carretta1,*, S. Parenti1, S. Mallia1, S. Rontauroli1, C. Chiereghin2, S. Castellano1,
E. Bianchi1, E. Genovese1, S. Sartini1, L. Tavernari1, M. Mirabile1, M. G. Della Porta2,3,
R. Manfredini1
1Center for Regenerative Medicine “S. Ferrari”, University of Modena and Reggio Emilia,
Modena; 2Humanitas Clinical and Research Center -IRCCS, Rozzano; 3Department of Biomedical
Sciences, Humanitas Unviersity, Milan, Italy
Background: Myeloproliferative neoplasms (MPNs) are a group of hematopoietic stem
cell disorders resulting in the overproduction of myeloid differentiated cells. Primary
Myelofibrosis (PMF) is characterized by the worst prognosis and 15-20% of cases develop
secondary Acute Myeloid Leukemia (AML). MPNs driver mutations affect JAK2, CALR or
MPL genes. Moreover, mutations in epigenetic regulators can exacerbate the disease
and alter response to treatment. Our group recently demonstrated through single cell
analysis that MPNs progression is due to increased genetic heterogeneity, loss of
heterozygosity and parallel AML evolution.
Aims: In this work we sought to define the genomic architecture of MPN patients during
disease evolution, and gain insight into the chromatin and transcriptional perturbations
induced by epigenetic modifiers’ mutations.
Methods: In order to describe MPNs clonal hierarchy at the single cell level, we employed
Mission Bio Tapestri platform. We analyzed the CD34+ compartment of 3 patients during
the chronic phase of the disease and after leukemic transformation through a custom
panel comprising 29 genes frequently mutated in myeloid neoplasms. CNV assessment
was performed on the same data through Mosaic algorithm.
On one patient, single nucleus RNA (snRNA-seq) and ATAC-seq were then conducted through
10x genomics instrument on CD34+ cells from the chronic and blast phase. Data were
analyzed through Seurat R package.
Results: Genomic analysis was performed on 20.652 single cells coming from 7 samples.
The analyzed patients suffered from Essential Thrombocythemia (n=2) or PMF (n=1);
2 patients harbored CALR type 1 driver mutation, while one patient carried JAK2V617F
variant. In all patients, the first mutational hit occurred on epigenetic remodeler
genes (i.e. TET2, ASXL1). Driver mutations’ allele frequency remained stable during
disease progression and did not seem to drive leukemic transformation; in one case,
JAK2V617F variant was lost in blast phase. In one patient, single cell analysis revealed
the acquisition of 3 mutually exclusive pathogenic variants in RAS pathway (two NRAS
mutations and a KRAS mutation). Notably, leukemic driver mutations, affecting genes
such as IDH2, TP53 and KRAS, were already traceable at very low allele frequency in
the chronic phase of the disease of all patients, despite being undetectable by bulk
diagnostic NGS analysis. For all patients, CNV analysis highlighted a higher gene
dosage imbalance in the leukemic clones when compared with those in the chronic phase.
Preliminary analysis of single cell ATAC+RNAseq data showed that the leukemic sample
is enriched in more primitive cell types (e.g. multipotent progenitors). Moreover,
genes found to be upregulated and more accessible in blast phase erythroid progenitors
and HSC clusters are associated with AML poor prognosis.
Summary/Conclusion: Altogether this analysis suggests that MPNs’ first mutational
hit frequently occurs in chromatin remodeler genes and affects a large fraction of
neoplastic cells, confirming their impact on MPNs pathogenesis. Single cell multiomic
analysis suggests that epigenetic alterations contribute to alter CD34+ cells differentiation
state and activate the expression of pro-leukemic genes. Moreover, genomic analysis
highlighted that clones carrying driver mutations remain stable during time and do
not seem to drive leukemic transformation. On the other hand, genetic alterations,
such as SNVs and CNVs, driving AML evolution are early identified by single cell analysis
despite being undetectable by bulk sequencing.
P984: MUTATIONAL PATTERNS IN PH-NEGATIVE MYELOPROLIFERATIVE NEOPLASMS (MPN)
I. C. Casetti1,*, D. Pietra2, V. V. Ferretti2, A. De Silvestri2, O. Borsani1, D. Vanni1,
C. Trotti1, S. Catricalà2, C. Favaron1, M. Cazzola1, L. Arcaini1,2, E. Rumi1,2
1University of Pavia; 2Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
Background: According to the 2016 WHO classification, Ph-negative MPN include essential
thrombocythemia (ET), polycythemia vera (PV), prefibrotic myelofibrosis (prePMF) and
overt fibrotic myelofibrosis (PMF). The molecular basis of MPN was clarified with
the discovery of driver mutations in JAK2, MPL and CALR genes, but the biological
landscape is more complex than initially assumed. Subclonal somatic mutations, also
found in other myeloid neoplasms (MN), have been described and are associated with
disease progression and poor prognosis. Those mutations target epigenetic regulators
of DNA (DNMT3A, TET2, IDH1/IDH2), genes involved in the chromatin structure (ASXL1,
EZH2), spliceosome genes (SF3B1, SRSF2), transcription factors and tumor suppressor
genes (RUNX1, TP53).
Aims: To correlate driver mutations, subclonal variants, clinical data and diagnosis
in 509 MPN patients (236 ET, 91 prePMF, 182 PMF) and to define a diagnostic and prognostic
role of these variants.
Methods: We studied DNA variants with the Illumina Nextera Rapid Capture Custom Enrichment
Kit and HiSeq2500 platform. We selected a panel of 81 genes based on prior implication
in the pathogenesis of MN. Inclusion criteria were (i) a diagnosis of ET, prePMF or
overt PMF according to the 2016 WHO criteria, (ii) a peripheral blood sampling at
diagnosis or before therapy administration.
Results: Overall, we detected 598 additional somatic variants. PMF showed a larger
proportion of patients (pts) with at least one additional variant compared with prePMF
and ET (86.3% vs 42.9% vs 34.8%, p<0.001) and a higher average number of variants
per pts. These findings suggest that PMF is not an advanced stage of prePMF, but a
different clinical entity with high tendency in variants accumulation.
Considering the driver mutation, the number of pts with at least one additional variant
was significantly different among the molecular subgroups (CALR 46.5%, JAK2 58.9%,
MPL 82.1% TN 31.1%, p<0.001).
Somatic mutations were grouped based on their role in the cell cycle. In ET the most
commonly mutated genes belong to the DNA methylation regulation and the most frequently
involved gene was TET2. In PMF, chromatin structure, RNA splicing and DNA methylation
were the most recurrently involved pathway and ASXL1 and TET2 were the most frequently
mutated genes. In prePMF most of the variants impaired DNA methylation and RNA splicing
and TET2 and SF3B1 were the most frequently mutated genes. RNA splicing and DNA methylation
are often involved in myelodisplastic syndromes (MDS). Thus, PMF and prePMF share
molecular signatures with MDS, suggesting that MN are part of a continuum spectrum
of diseases.
There was no association between driver mutations and additional variants, except
for the spliceosome genes. The rate of splicing factors alterations in CALR mutants
was significantly lower compared with JAK2 or MPL mutants (3.9% vs 22.3% vs 35.7%,
p<0.001).
We finally correlated additional mutations and clinical data (blood count, splenomegaly,
LDH, CD34+ circulating cells), overall survival (OS) and progression to blast phase
(BP). We focused on the most commonly mutated genes, defined as mutations detected
in at least 10 pts (>5%): ASXL1, DNMT3A, TET2, EZH2, SRSF2. ASXL1 was associated with
anemia and splenomegaly, a higher risk of leukemic evolution (sHR=4.5, 95%CI:1.7-11.8,
p=0.003) and shorter OS (HR=6.2 (4.3-9.0) p<0.001); SRSF2 had an impact on BP evolution
and OS, while EZH2 showed an impact on OS.
Image:
Summary/Conclusion: We suggest that not only histological and clinical criteria, but
also distinct mutational patterns might differentiate ET, prePMF and overt PMF.
P985: THE ALLELIC RATIO OF DRIVER AND ASXL1 MUTATIONS IS PROGNOSTICALLY RELEVANT IN
PMF
G. Coltro1,2,*, G. Rotunno1,2, F. Mannelli1,2, G. G. Loscocco1,2, N. Bartalucci1,2,
C. Mannarelli1,2, C. Maccari1,2, P. Guglielmelli1,2, A. M. Vannucchi1,2
1Department of Experimental and Clinical Medicine, University of Florence; 2CRIMM,
Center for Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria
Careggi, Florence, Italy
Background: Primary myelofibrosis (PMF) is a clonal disorder driven by mutations in
JAK2, CALR, and MPL. Somatic mutations in myeloid-associated genes were shown to impact
prognosis of PMF patients (pts). Among these, ASXL1 mutations (ASXL1
mt) are by far the most frequent and prognostically relevant, in fact they are included
in the category of “high molecular risk” (HMR), along with EZH2, IDH1/2, SRSF2, and
U2AF1
Q157 mutations.
Aims: To investigate the phenotypic and prognostic implications of ASXL1
mt and variant allele frequency (VAF) in PMF.
Methods: After IRB approval, consecutive pts with WHO-defined PMF were included in
the study. Mutational analysis by targeted NGS was performed by previously described
methods (Guglielmelli P et al., JCO 2017).
Results: The study enrolled a total of 384 pts, including 190 (49%) prefibrotic and
194 (51%) overt PMF. Median age was 60 (18-90) years, 236 (61%) were male. JAK2, CALR
and MPL mutations were found in 255 (66%), 83 (22%), and 17 (4%) pts, respectively;
36 (9%) were triple negative (TN). Among HMR mutations, ASXL1 was mutated in 88 (23%)
pts, SRSF2 in 35 (9%), EZH2 in 32 (8%), U2AF1 in 15 (5%), and IDH1/2 in 11 (3%). VAF
distribution of driver and HMR mutations was as reported in Fig.1A.
We did not find any significant correlation of ASXL1
mt VAF with other clinical and molecular characteristics, including PMF subtype, gender,
age, hemoglobin, leukocyte, platelet and blast counts, splenomegaly, BM fibrosis grade,
cytogenetics, mutational status for driver and other HMR genes and their VAFs. The
only exception was the association of CALR-mutated PMF with lower ASXL1 VAF (median
28% vs 40%, p=.0332).
Median overall survival (OS) was 120 (102-151) months. In univariate Cox analysis,
ASXL1
mt VAF did not correlate with OS, nor did other single driver and HMR mutations. When
considered as a whole (for the purpose of this study, TN pts were considered has having
driver VAF equal to 0), the VAF of driver mutations inversely correlated with survival
(HR 0.4 [0.2-0.9], p=.0219). We next investigated the allelic ratio between driver
and ASXL1 mutations (d/Aratio) and its prognostic correlates. Median (range) of d/Aratio
was 1.24 (0-13), and ROC analysis with death as an endpoint identified 1.31 as the
optimal cut-off value. In univariate analysis, pts with a d/Aratio <1.31 had a significantly
worse OS compared to those with a ratio ≥1.31 (median 45 vs 104 months, p=.0014; HR
2.4 [1.4-4.1]) (Fig.1B). When TN pts with ASXL1
mt were considered apart, having a d/Aratio <1.31 retained its inferior prognostic
impact compared to the d/Aratio ≥1.31 group (median OS 57 vs 104 months, p<0.0453;
HR 1.8 [1-3.2]), with TN/ASXL1
mt pts being associated with the worst outcome (median OS 24 months) (Fig.1C).
In a multivariate Cox model including VAF-adjusted ASXL1 mutant status (ASXL1
wt
vs ASXL1
mt with d/Aratio ≥1.3 vs ASXL1
mt with d/Aratio <1.3) and other HMR mutations, the former and mutated SRSF2 were
confirmed to be independent predictors of inferior OS. Both d/Aratio <1.31 and ≥1.31
remained significant compared to ASXL1
wt with respective HRs of 3.4 (2.3-5.1; p<.0001) and 1.6 (1-2.6; p=.0342). Notably,
d/Aratio <1.31 still retained its inferior prognostic impact compared to d/Aratio
≥1.31 (HR 2 [1.2-3.5]; p=.0095).
Image:
Summary/Conclusion: This study explores the implications of ASXL1
mt VAF and its interplay with driver mutant burden. The adverse prognosis of pts with
a d/Aratio <1.31 suggests that this disease entity is predominantly driven by ASXL1
mt-clones characterized by a more aggressive biology. Further research is needed.
P986: CYTOGENETIC DIAGNOSIS WITH NEXT-GENERATION CYTOGENETICS BY OPTICAL GENOME MAPPING
IN PATIENTS WITH MYELOFIBROSIS.
Á. Díaz González1,*, G. Avetisyán1, E. Mora1, M. Santiago2, A. Liquori1, S. Furió1,
S. García1, A. Villalba1, Á. López1, J. V. Gil1, C. García1, E. González1, C. Martínez1,
B. Fernández1, M. Guaita1, B. Martín1, L. Cordon1, E. Barragán1, J. de la Rubia1,
J. Cervera2, E. Such1
1Hematology; 2Genetics Unit, Hospital Universitario y Politécnico La Fe, Valencia,
Spain
Background: The prognosis of myelofibrosis patients is highly variable. Due to this
heterogeneity, it is important to have prognostic tools that correctly stratify the
patients. The most current prognostic scales such as the MIPSS70+ v2.0 or the GIPSS
require cytogenetic data. The main limitation in classifying these patients is the
lack of information on cytogenetic alterations because of bone marrow fibrosis and
absence of metaphases.
Recently, a new strategy has emerged that characterize all cytogenetic alterations
using Optical Genome Mapping considered Next Generation Cytogenetics which overcomes
the limitations of conventional cytogenetics by using ultra high molecular weight
DNA instead of metaphases. This DNA can be obtained from peripheral blood or bone
marrow samples.
Aims: The main objective of this project is to perform a complete genomic characterization
of 10 patients with myelofibrosis by karyotype, FISH, Next Generation Sequencing (NGS)
and Optical Genome Mapping (OGM).
Methods: We have performed the karyotype staining the chromosomes by G-banding and
the results were reported according to the International System for Human Cytogenetic
Nomenclature (ISCN, 2020). FISH was performed to discard rearrangements of PDGFRA,
PDGFRB, FGFR1 and JAK2. NGS libraries were prepared using a panel SOPHiA Myeloid Solution™
kit which includes SNV and CNV of 30 genes related to myeloproliferative neoplasms
and leukemia. For study by OGM we obtained ultra-high molecular weight (UHMW) DNA
from peripheral blood samples which are labeled with DLGreen fluorophores using Enzyme
1 (DLE-1) reactions following the manufacturer’s protocols (Bionano Genomics). Labeled
DNA was loaded on a Saphyr chip and run on a Saphyr instrument (Bionano Genomics).
The novo genome map assembly will be performed using BionanoSolve™ and structural
variants will be called against the human reference hg38 assembly. Data was analyzed
with Bionano Access™ (Bionano Genomics).
Results: Preliminary results were obtained:
- Patient 1 had unsuccessful karyotype and JAK2 (p.Val617Phe, VAF 13%) mutation by
NGS.
- Karyotype result of patient 2 suggest that is normal: 46,XY[6]. NGS detected pathogenic
mutations in JAK2 (p.Val617Phe, VAF 41%), SRSF2 (p.Pro95His, VAF 42%) and ETV6 (p.Arg105*,
VAF 41%) and likely pathogenic mutation in ASXL1 (p.?; c.1720-2A>G, VAF 45%).
- Patient 3 had a normal karyotype: 46,XX[18] with CALR type 1 mutation (p.Leu367Thrfs*?,
VAF 37%) by NGS.
We have not detected rearrangements by FISH in none of the 3 patients. 1500 Gbp of
DNA were collected to perform the OGM for each patient and we selected structural
variants that were present in less than or equal to 0,5 % of the control samples with
the same enzyme. Patients 1 and 2 who had unsuccessful karyotype could be characterized
by OGM. We have found by OGM a median of 1 insertion (range 0-4), 8 deletions (4-10)
and 2 duplications (0-3). No inversion or translocation were detected. No cytogenetic
alterations classified as very high-risk karyotype in MIPSS70+ v2.0 scale were detected.
After an exhaustive analysis of these structural alterations initially detected, all
alterations were ruled out as they are present in general population and therefore
correspond to a normal karyotype.
Summary/Conclusion: Optical Genome Mapping suggest to be very promising diagnostic
tool that can complete the study of patients with myelofibrosis, especially in those
with non-assessable karyotype. The extension to a larger number of patients will confirm
the results obtained and detect variables that remain cryptic to the karyotype.
P987: A DIFFERENT BALANCE IN OXIDATIVE STRESS RESPONSE IN CALR AND JAK2 MUTATED MYELOFIBROSIS
PATIENTS CORRELATES WITH CLINICAL OUTCOME
E. Genovese1,*, M. Mirabile1, S. Rontauroli1, S. Sartini1, S. Fantini1, L. Tavernari1,
M. Maccaferri2, P. Guglielmelli3, E. Bianchi1, S. Parenti1, C. Carretta1, S. Mallia1,
S. Castellano4,5,6, C. Colasante5, M. Balliu3, N. Bartalucci3, R. Palmieri7, T. Ottone7,8,
B. Mora9, L. Potenza5, F. Passamonti9, M. T. Voso7,8, M. Luppi5, A. M. Vannucchi3,
E. Tagliafico4,5, R. Manfredini1
1Life Sciences Department, Centre for Regenerative Medicine, University of Modena
and Reggio Emilia; 2Department of Laboratory Medicine and Pathology, Diagnostic Hematology
and Clinical Genomics, AUSL/AOU Policlinico, Modena; 3Department of Experimental and
Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms
(CRIMM), University of Florence, Careggi University Hospital, Florence; 4Center for
Genome Research, University of Modena and Reggio Emilia; 5Department of Medical and
Surgical Sciences, University of Modena and Reggio Emilia, AUSL/AOU Policlinico; 6PhD
Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia,
Modena; 7Department of Biomedicine and Prevention, University of Tor Vergata; 8Santa
Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, Rome; 9Division of Hematology,
Ospedale ASST Sette Laghi, University of Insubria, Varese, Italy
Background: Myelofibrosis (MF) is the Philadelphia-negative myeloproliferative neoplasm
characterized by the worst prognosis and poor response to conventional therapy. Driver
mutations in JAK2 and CALR impact on JAK-STAT pathway activation but also on the production
of reactive oxygen species (ROS). ROS play a pivotal role in inflammation-induced
oxidative damage to cellular components including DNA, that leads to greater genomic
instability and promotes cell transformation.
Aims: In order to unveil the role of driver mutations in oxidative stress response,
we evaluated oxidative stress markers levels in CD34+ hematopoietic stem/progenitor
cells and plasma from MF patients.
Methods: Firstly, we have measured the levels of ROS, 8-hydroxy-2-deoxy-guanosine
(8-OHdG) and activity of superoxide dismutase (SOD) in CD34+ cells from 20 JAK2 and
14 CALR myelofibrosis (MF) patients compared with 17 healthy donors (HD). In particular,
the redox-sensitive CM-H2DCFDA was used to measure the intracellular ROS. The levels
of 8-OHdG were detected by enzyme-linked immunosorbent assay (ELISA). Moreover, SOD
activity was measured by means of colorimetric assay. Subsequently, we evaluated whether
a different oxidative status could be detected in 129 plasma samples of MF patients
(n=86 JAK2, n=43 CALR), as the level of total antioxidant capacity (TAC), an analyte
frequently used to assess the antioxidant status of biological samples. Finally, we
evaluated the association between TAC plasma levels and overall survival (OS) by means
of multivariate analysis.
Results: Our results demonstrated that ROS production in CD34+ cells from CALR-mutated
MF patients is strongly increased compared with patients harboring JAK2 mutation,
leading to increased oxidative DNA damage. Moreover, CALR-mutated cells show less
SOD antioxidant activity than JAK2-mutated ones. Subsequently, we found that high
TAC plasma levels correlate with detrimental clinical features, such as high levels
of lactate dehydrogenase (LDH) and circulating CD34+ cells. Moreover, high TAC plasma
levels are also associated with a poor OS in JAK2-mutated patients. Multivariate analysis
demonstrated that high plasmatic TAC is an independent prognostic factor for OS that
allows the identification of patients with inferior OS in both DIPSS lowest and highest
categories.
Summary/Conclusion: Altogether, our results confirmed that CALR mutation has a higher
impact on the oxidative stress status in MF cells compared to JAK2 variant. Furthermore,
plasma samples from CALR mutated patients have significantly lower TAC levels leading
to a lower responsiveness to oxidative injury. On the other hand, increased TAC levels
correlate with the presence of JAK2 mutation and several detrimental clinical features.
MF patients with high plasmatic TAC display inferior survival and multivariate analysis
demonstrated that increased TAC activity might represent a novel prognostic biomarker
independent from DIPSS classification. We speculated that the high increase in oxidative
stress in CALRmutated patients could be involved in the activation of protective mechanisms
which ultimately promote cell death. On the contrary, in JAK2 mutated patients, the
slight increase in oxidative stress can determine the persistence of cells with damaged
DNA where the accumulation of mutations promotes the disease progression.
P988: LACTATE RESHAPES TUMOR MICROENVIRONMENT AND METABOLIC PROFILE IN MYELOFIBROSIS
S. Giallongo1,*, D. Tibullo1, M. Spampinato1, C. Giallongo2, E. La Spina3, L. Longhitano1,
A. Romano3, I. Dulcamare3, A. Barbato3, G. Scandura3, A. M. Amorini1, G. Lazzarino1,
T. Zuppelli2, R. Caltabiano2, G. Li Volti1, F. Di Raimondo3, G. A. M. Palumbo2
1BIOMETEC; 2Department of Medical, Surgical Sciences and Advanced Technologies G.F.
Ingrassia, A.O.U. “Policlinico-Vittorio Emanuele; 3Department of General Surgery and
Medical-Surgical Specialties, Division of Hematology, A.O.U. “Policlinico-Vittorio
Emanuele”, University of Catania, CATANIA, Italy
Background: Primary myelofibrosis (PMF) is myeloproliferative neoplasm depicted by
bone marrow (BM) fibrosis, ineffective clonal hematopoiesis, and splenomegaly. These
outcomes are related with changes in bone marrow microenvironment, sustaining cancer
glycolytic metabolism, fibrosis, immune evasion, and thrombotic events. Lactate, a
glycolysis byproduct, may represent the driving-force towards the acquisition of these
features. For this reason, we investigated PMF CD34+ cells lactate production as factor
prompting the metabolic rewire of BM niche.
Aims: In this work we aimed to target lactate metabolism as stricking factor improving
PMF proliferative features.
Methods: Primary mesenchymal stem cells (MSCs) and HS-5 proliferation assays were
performed in vitro. Protein expression level was assessed using immunoblot and immunohistochemical
assays. Moreover, Cytokines were also investigated ex vivo, using Multiplex immunobead
assay technology. These results were also mirrored in vivo on TPOhigh Zebrafish (Danio
rerio) model.
Results: The monocarboxylate transporters (MCT-1 and -4), orchestrating lactate intake,
were significatively upregulated in CD34+ cells and sera derived from MF-patients
compared to healthy donor. MF-patients are characterized by increased percentage of
circulating immunosuppressive cells. Incubation of peripheral blood mononucleated
cells (PBMNCs) with MF-sera increased Treg and Myeloid-derived suppressor cells (M-MDSCs)
expansion. This effect was disrupted by addition of MCT1 inhibitor AZD3965. The role
of lactate on BM niche was evaluated by culturing healthy primary MSCs. Lactate (20 mM)
and MF-sera exposition induced extracellular matrix (ECM) reorganization as supported
by increased level of metalloproteases (MMP9, MMP2) together with reticulin and collagen
deposition. Moreover, lactate induces calcium accumulation and soluble osteogenic
molecular signals production (i.e. osteoprotegerin, osteonectin). On the epigenetic
side, lactate treatment reshapes chromatin architecture eventually affecting histones
lactylation, acetylation and methylation levels. These features were rescued upon
AZD3965 treatment. Cancer-associated fibroblast (CAF) play an outstanding role in
cancer development by ECM remodeling.Upon lactate exposure, expression of CAF markers
α-SMA, FAP1 and TGFβ were upregulated in MSCs. The effect was reverted by AZD3965
treatment. The data achieved in vivo, and ex vivo were corroborated in a new pre-clinical
TPOhigh Zebrafish model eventually showing increased Whole Kidney Marrow lactate.
Summary/Conclusion: In this work we report MCT1 inhibition as a strategy to treat
invasive and metastatic cancers. Our results corroborate the idea that targeting lactate
metabolism may represent a promising path to counteract inflammation, osteosclerosis
and fibrosis in PMF patients.
P989: DYNAMIC CHANGES IN THE HISTONE H3 LYSINE 27 TRIMETHYLATION EPIGENETIC LANDSCAPE
FOLLOWING RUXOLITINIB ADMINISTRATION
G. Greenfield1,*, M. F. McMullin2, K. Mills3
1PGJCCR; 2CME, Queen’s University Belfast; 3PGJCCR, Queen’s Universtiy Belfast, Belfast,
United Kingdom
Background: Dysregulation of normal epigenetic processes is increasingly recognised
as a feature of myeloid malignancy generally and implicated in the pathogenesis of
MPN. Epigenetic changes may directly result from the mutation of key epigenetic regulators
including TET2, ASXL1 and EZH2 or indirectly from the activation of intracellular
signalling cascades. We have previously observed that ruxolitinib, a JAK1/2 inhibitor,
may alter the epigenetic landscape of histone modifications in MPN. CUT&RUN is a novel
experimental approach providing high resolution profiling of epigenetic modifications
and transcription factor binding.
Aims: To comprehensively profile the H3K27me3 epigenetic landscape of JAK2 V617F positive
cells resulting from JAK inhibition and determine the dynamic changes occurring between
short term and persistent ruxolitinib therapy to identify potential genetic contributors
to pathogenesis and options for future therapeutics.
Methods:
JAK2 V617F positive UKE1 cells were cultured in duplicate in the presence of 1000nM
ruxolitinib or DMSO 0.1% vehicle control continuously for approximately five weeks.
Samples were harvested at 37 days for CUT&RUN analysis using H3K27me3, EZH2 and IgG
(negative control) antibodies. CUT&RUN performed using Epicypher CUTANA Kit according
to manufacturer instructions. Libraries prepared using NEBNext® Ultra™ II DNA Library
Prep kit with 50 base pair, paired end sequencing undertaken on Illumina NovaSeq 6000.
Resulting FASTQ files were trimmed using Trim Galore! and aligned to reference genome
hg38 using Bowtie2. Duplicate reads were removed using SAMtools rmdup and peaks called
with MACS2 callpeak on the usegalaxy.eu platform. Differential peak analysis undertaken
with Diffbind package in R. Samples were normalised to library size prior to differential
analysis with DESeq2 methodology. Bigwig files generated using deepTools bamcoverage
for visualisation.
Results: UKE1 cells remain sensitive to ruxolitinib after a period of treatment with
high dose ruxolitinib lasting over 5 weeks. CUT&RUN libraries were sequenced to a
minimum depth of 3.6 million unique reads per sample. Initial comparisons between
Day 37 high dose treated cells and vehicle control revealed 1004 differentially enriched
H3K27me3 peaks. Of these, 545 (54.2%) peaks were increased in the ruxolitinib treated
cells and 459 (45.7%) peaks were increased in the control cells. Analysis of the difference
between day 37 and day 1 high dose ruxolitinib treated cells revealed 290 peaks differentially
enriched. Of these peaks, 279 (96.2%) were increased and 11 (3.8%) were decreased
at day 37. Analysis of the overlap between these results identified 33 peaks increased
at D37 in comparison to DMSO control and day 1 treatment samples.
Analysis of the differential binding patterns and direct visualisation of the results
in a genome browser revealed particular genetic locations of interest including the
promoter region of SPRY1, a potential regulator of erythropoiesis and JAK signalling,
UGCG and BTBD11. Figure 1 shows an example. Differential H3K27me3 was also noted at
regions associated with PLXNA1 and NRP2 genes which act as co-receptors for class
III semaphorin signalling.
Image:
Summary/Conclusion: The dynamic changes in the H3K27me3 epigenetic landscape of JAK2
V617F positive cells following persistent high dose ruxolitinib administration has
potential therapeutic implications and may sensitise cells to epigenetic therapies.
Specific gene loci identified demonstrating differential H3K27me3 may offer some insight
into JAK2 V617F mediated pathology and require further investigation.
P990: T CELL RESPONSE AND OMICRON VARIANT NEUTRALISATION FOLLOWING VACCINATION AGAINST
SARS-COV-2 IN PATIENTS WITH CHRONIC MYELOID DISORDERS
P. Harrington1,2,*, J. Saunders1, C. Saha1, A. Sheikh1, R. Dillon1, K. Raj1, L. Cadman-Davies1,
T. Lechmere3, H. Khan3, C. Woodley1, S. Asirvatham1, N. Curto-Garcia1, J. O’Sullivan1,
S. Kordasti1, D. Radia1, D. McLornan1, M. Malim3, K. Doores3, C. Harrison1, H. de
Lavallade1,2
1Clinical Haematology, Guy’s & St Thomas’ NHS Foundation Trust; 2School of Cancer
& Pharmaceutical Science; 3Infectious Diseases, King’s College London, London, United
Kingdom
Background: Concerns remain over the response to vaccination against SARS-CoV-2 in
patients with haematological malignancy.
Aims: Herein, we report a comprehensive immunological evaluation of the response to
SARS-CoV-2 vaccination in patients with chronic myeloid neoplasms (MPN).
Methods: Antibody response was assessed using anti-Spike (anti-S) IgG with anti-nucleocapsid
(anti-N) IgG used to determine previous infection. Neutralising antibody analysis
was performed assessing inhibitory effect of plasma on entry of HIV-1 particles expressing
Wuhan and omicron variant spike proteins into cells expressing ACE-2 receptor.
T cell response was assessed using flow cytometric evaluation of intracellular pro-inflammatory
cytokines and surface exhaustion markers upon re-exposure to S peptides (Miltenyi)
in T cell subsets. Subsequently, T cell response was evaluated using a fluorospot
assay assessing IFNg/IL-2 secretion upon re-exposure to S peptides (Mabtech). Plates
were analysed using the IRIS reader providing both spot-forming unit (SFU) frequency
and relative spot volume (RSV).
Results: Samples were collected in 102 patients with MPN or post allo-SCT. Testing
was performed in 37 MPN patients after one dose, 61 after 2 doses and 30 after 3 doses
to date.
An anti-S IgG response was observed in 81.1% (30/37) of MPN patients after a first
dose, increasing after two doses to 91.7% (55/60). Those with previous infection had
higher anti-S IgG and neutralising antibody levels following two doses of vaccine
(p=0.0002/p<0.0001). MPN patients receiving the BNT162b2 vaccine had higher anti-S
IgG and neutralising antibody levels compared with those receiving ChAdOx-1 (p=0.05/p=0.04).
The proportion of patients on ruxolitinib (rux) with a negative or borderline response
after two doses was higher than observed in other patients (5/10 vs 5/47, p=0.01).
Preliminary analysis in 20 patients showed reduced neutralisation of omicron compared
with the Wuhan spike, with neutralising antibody ID50 of 93 vs 218 (p=0.06, Fig. 1a).
Using fluorospot analysis after two doses, an overall T-cell response was observed
in 88.3% (53/60) patients. Patients on rux had lower SFUs for IFN and IL-2 and amongst
those on treatment, RSV of IFNg was again lowest in patients on rux, with mean RSV
of 2942 vs 7990 in patients on hydroxycarbamide and 6687 in peg-IFNa (p=0.016/p=0.0008).
T cells expressed increased exhaustion markers upon re-exposure to S peptides in patients
with CML following vaccination.
After a third dose, T cell response was assessed in 30 patients with a positive response
in 90% (27/30). IFNg SFUs and RSV were significantly higher after 3 vs 2 doses at
242 and 9184 vs 72 and 5031 (p=0.0004/<0.0001, Fig. 1b,c). Patients on rux after 3
doses had lower SFU and RSV of IFNg than other patients at 79 and 7690 vs 302 and
9727 (p=0.035/0.04 Fig 1d,e). Patients on rux were more likely to have a negative
T cell response than other patients (4/8 vs 20/22, p=0.0294). Patients with a diagnosis
of MF had lower SFU and RSV for IFNg than patients with CML, PV and ET (SFU: 93 vs
294, 224 and 446, p=0.09/0.002/0.15, RSV: 7532 vs 9528, 10499 and 10630, p=0.08/0.001/0.01,
Fig. 1f)
Image:
Summary/Conclusion: We show overall high frequency of seroconversion and memory T
cell responses but demonstrate the need for alternative strategies in some groups,
including patients taking ruxolitinib. The increase in T cell reactivity after 3 doses
is of particular significance in view of the reduced neutralisation of the now dominant
omicron variant, with studies showing T cell functionality is maintained against omicron.
Updated results will be presented.
P991: IMPAIRMENT OF TCR SIGNALLING IN PATIENTS WITH MYELOFIBROSIS TREATED WITH RUXOLITINIB
P. Harrington1,2,*, R. Dillon1, D. Radia1, D. McLornan1, N. Curto-Garcia1, C. Woodley1,
S. Asirvatham1, S. Kordasti1,2, C. Harrison1,2, H. de Lavallade1,2
1Clinical Haematology, Guy’s & St Thomas’ NHS Foundation Trust; 2School of Cancer
& Pharmaceutical Science, King’s College London, London, United Kingdom
Background: Ruxolitinib (rux) is a JAK1/JAK2 inhibitor with varied immune effects
including on T cell, NK cell and dendritic cell function. Therapeutic effects are
largely attributed to marked reduction in pro-inflammatory cytokines.
Aims: We hypothesised that impairment of T cell receptor (TCR) signalling through
off-target binding of Src kinases, including Lck, would be an additional mechanism
of immune dysfunction.
Methods: We performed phosphoflow cytometry in Tregs, T effectors and NK cells to
assess the effect on signalling downstream from the TCR and activating NK cell receptors.
11-colour flow cytometry was performed after cells were activated with H2O2 for 15
minutes, due to its activity as a potent phosphatase inhibitor and cells were analysed
for phosphorylation of ZAP70 as a surrogate of TCR signalling and STAT5. A gating
strategy of CD4+/CD25+/FOXP3+/CD127lo cells was used for identification of Tregs.
Results: Phosphoflow analysis was performed in 7 patients with a diagnosis of MF managed
with rux and compared with 7 healthy controls (HC). 4 (57%) of patients were male
and mean age was 61 (range 44-76). Patients were taking a median rux dosage of 30mg
daily (10-50). Three patients had a DIPPS+ score of Int-2, 2 Int-1 and 2 low-risk.
Results of phosphoflow analysis are expressed as relative fluorescence intensity (RFI)
defined as MFI in H2O2 stimulated sample/MFI in unstimulated sample. As expected,
patients on rux had significantly reduced phosphorylation of pSTAT5 when compared
with HC in all cell subsets evaluated. The mean RFI of pSTAT5 in CD4+ and CD8+ cells
in HC was 21.9 and 31.1 respectively, compared with 6 and 8.5 in rux patients (p=0.001/p=0.002).
In CD56+ cells this was 46.2 vs 7.7 (p=0.005) and in Tregs this was 21.9 vs 6.1 (p=0.019).
Of note, a difference in phosphorylation was also observed for pZAP70 indicative of
inhibition of TCR proximal signalling. For pZAP70 mean RFI in CD4+ cells was 8.8 in
controls compared with 2.2 in rux patients (p=0.05). In CD8+ cells mean RFI was 11.4
in controls and 2.4 in rux patients (p=0.04). In CD56+ cells mean RFI was 17.8 vs
2.4 (p=0.02), whilst in Tregs, significance was not reached with mean RFI of 6.3 vs
2.2 (p=0.13). Univariate analysis of rux patients showed no effect of age, comorbidities,
DIPPS+ score or rux dosage on TCR or STAT5 signalling.
Findings were compared with those observed in 4 patients with CML and deep molecular
response on nilotinib, in view of the highly specific ABL1 binding properties of this
TKI. As expected, patients on nilotinib had significantly higher RFI for pSTAT5 compared
with rux patients, at 23, 32.8, 21.2 and 46.5 in CD4+, CD8+, Treg and NK cells respectively
(p=0.002/p=0.002/p=0.006/p=0.003). Interestingly, patients on nilotinib also had significantly
higher RFI for pZAP70 in CD4+, CD8+ and NK cells at 6.7, 7.4 and 8.1 compared with
rux patients (p=0.035/p=0.025/p=0.037).
Image:
Summary/Conclusion: Studies have shown that rux inhibits T cell proliferation and
reduces CD4+ cell cytokine secretion. Previous in vitro analysis of rux on CD4+ cells
failed to identify an effect on TCR signalling after CD3/CD28 stimulation. In our
ex-vivo analysis, using H2O2 stimulation, we observe TCR signalling inhibition in
CD4+, CD8+ and NK cells, although reduced compared with effects on STAT5. It has been
shown that rux inhibits Lck with an IC50 of 3.6uM. Whilst this is significantly less
than the inhibitory effect on JAK kinases, there is ~34% sequency homology between
Src and JAK family kinases in the kinase domain, suggesting off target kinase inhibition
is implicated in the impairment of TCR signalling.
P992: OUTCOMES OF GLOBAL COAGULATION ASSAYS IN PATIENTS WITH PHILADELPHIA-NEGATIVE
MYELOPROLIFERATIVE NEOPLASMS WITH RESPECT TO THEIR CLINICAL AND GENETIC DETERMINANTS
OF CLONAL EVOLUTION
A. Lucchesi1,*, R. Napolitano2, M. T. Bochicchio2, G. Simonetti2, G. Micucci1, M.
Poggiaspalla1, V. Di Battista1, G. Musuraca1, F. Foca3, G. Giordano4, L. Catani5,
M. Napolitano6
1Hematology Unit; 2Biosciences Laboratory; 3Unit of Biostatistics and Clinical Trials,
IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola;
4Internal Medicine Division, Hematology Service, Regional Hospital “A. Cardarelli”,
Campobasso; 5IRCCS Azienda Ospedaliero-Universitaria di Bologna, Department of Experimental,
Diagnostic and Specialty Medicine, School of Medicine, Institute of Hematology “Seràgnoli”,
University of Bologna, Bologna; 6Department of Health Promotion, Mother and Child
Care, Internal Medicine and Medical Specialties and Haematology Unit, University Hospital
“P. Giaccone”, Palermo, Italy
Background: Classical Myeloproliferative Neoplasms (MPNs) are hematopoietic stem cell
diseases characterized by inflammation, promotion of atherosclerosis, hypercoagulability,
fibrosis, and clonal evolution. Our previous studies in cytofluorimetry have shown
that the reduced expression of platelet fibrinogen receptor (PFR) - common to all
MPNs - can be due to the hyperactivation of plasma-dependent mechanisms, such as tissue
factor (TF) release, an unbalanced thrombin generation and the marked involvement
of Protease Activated Receptors (PARs) and their signaling crosstalk with TGF-ß. Acetylsalicylic
acid (ASA) seemed to be able to restore the expression of PFRs.
Aims: The aim of the present study is to verify the existence of imbalances in hemostatic
function in rotational thromboelastometry (ROTEM), with respect to the different classical
forms of MPN, their cell counts, the presence of genetic mutations in Next Generation
Sequencing (NGS), PFR expression, the use of ASA and the history of vascular events.
Methods: We enrolled 53 patients affected by MPNs, naïve from any therapy, with the
only exception of ASA (taken by 50% of patients). We used the samples to run assays
of genetics (30-gene panel in NGS - SOPHiA Myeloid Solution™, SOPHiA Genetics, Switzerland),
cytofluorimetric determination of PFRs, global coagulation by rotational tromboelastometry
(ROTEM® Delta, Werfen, Spain) for INTEM, EXTEM, FIBTEM parameters.
Results: ROTEM parameters appear to exhibit substantial variations, depending on the
type of disease under investigation, cell counts, and selected mutations. Since most
of the values are in the normal range, the results should be read in terms of “efficiency”
or “preferential use” of one pathway over another. In particular, essential thrombocythemia
(ET) and CALR mutation seem to be related to an increased efficiency of both classical
coagulation pathways, with significantly more contracted clot formation times (CFT).
In contrast, primary myelofibrosis (PMF) and polycythemia vera (PV) show greater imbalances
in the hemostatic system. PV patients have longer CFT (EXTEM 114 sec, p= 0.008, INTEM
82 sec, p=0.027) and at the same time a selective contraction of the parameters in
INTEM for higher platelet (Plt), white blood cells (WBC) and hemoglobin (Hb) levels.
PMF - on the contrary - seems to exploit more the extrinsic pathway (EXTEM) - figure
A-B. In multivariate analysis, the major contribution to the alterations is credited
to Hb values for PV, while only Plt retains statistical significance for PMF and ET.
Selected mutations are associated with changes in ROTEM parameters. Among the DTA
mutations, the presence of DNMT3A shows a significant reduction in clotting time (CT)
in EXTEM, while ASXL1 is associated with reduced maximum lysis (ML). EZH2 could be
responsible for CFT elongations in the INTEM assay.
In addition, increased expression of PFRs is associated with bleeding history and
sustained CT time at FIBTEM under ASA prophylaxis.
Image:
Summary/Conclusion: In the most indolent MPNs (such as ET, especially if CALR mutated)
the balance and efficiency of the classical pathways is preserved. In PMF, the hemostatic
system is unbalanced towards the extrinsic pathway, confirming the connection between
increasing fibrosis and plasma-dependent hypercoagulable states. In contrast, rheological
characteristics and endothelial stress in PV seem to result in a greater involvement
of the intrinsic pathway. ROTEM and PFR expression confirm the existence of dynamic
states of hypercoagulability in MPNs and could be used to assess the efficacy of prophylaxis.
P993: SETD2 IS A BONA FIDE TUMOR SUPPRESSOR IN SYSTEMIC MASTOCYTOSIS
M. Mancini1,*, C. Monaldi2, S. De Santis2, C. Papayannidis1, M. Rondoni3, C. Sartor1,
S. Bruno2, A. Curti1, R. Zanotti4, M. Bonifacio4, L. Scaffidi5, L. Pagano5, M. Criscuolo5,
F. Ciceri6, C. Elena7, P. Tosi8, P. Valent9, M. Cavo10, S. Soverini2
1IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”;
2Department of Experimental Diagnostic and Specialty Medicine - DIMES, Institute of
Hematology “L. e A. Seràgnoli”, University of Bologna, Bologna; 3Azienda USL della
Romagna, Ravenna; 4Azienda Ospedaliera di Verona, Verona; 5Fondazione Policlinico
Universitario A. Gemelli, Roma; 6Servizio Immunoematologia Trasfusionale (SIMT), IRCCS
Ospedale San Raffaele, Milano; 7Fondazione I.R.C.S.S. Policlinico San Matteo, Pavia;
8U. O. di Oncologia ed Oncoematologia-Osp. Infermi Azienda Unità Sanitaria-Locale
di Rimini, Rimini, Italy; 9Department of Hematology, Internal Medicine I, Medical
University of Vienna, Vienna, Austria; 10IRCCS Azienda Ospedaliero-Universitaria di
Bologna, Istituto di Ematologia “Seràgnoli”; Department of Experimental Diagnostic
and Specialty Medicine - DIMES, Institute of Hematology “L. e A. Seràgnoli”, University
of Bologna, Bologna, Italy
Background: The trimethylation of histone H3 at lysine 36 (H3K36me3), catalyzed by
the SETD2 methyltransferase, is a post-translational modification involved in the
fidelity of transcription and splicing and in the recruitment of DNA repair machinery.
Recent studies have emphasized the tumor-suppressive role of SETD2, especially in
renal cancer where SETD2 is often deleted or mutated. A novel mechanism of SETD2 non
genomic loss of function due to proteasome-mediated degradation has recently been
reported in advanced systemic mastocytosis (advSM). Proteasome inhibition has been
found to inhibit clonogenic growth and induce apoptosis, but whether this is due to
SETD2 re-expression remains to be demonstrated.
Aims: The aim of our study is to demonstrate unequivocally that forced expression
of SETD2 in a cellular context in which it was lost, may restore proliferation control,
clonogenic capacity and DNA damage repair mechanisms.
Methods: The SETD2-deficient HMC-1.2 mast cell leukemia cell line was used as in vitro
model. Forced SETD2 re-expression was obtained by nucleofection: 106 HMC-1.2 cells
were resuspended in 85μl of cell line Nucleofector Solution V and 2μg of a SETD2 (GFP-tagged)
construct were added. An empty vector coding for GFP was used as negative control.
Cells were transfected by using the Lonza Nucleofector 2b and the Lonza Amaxa Cell
Line Nucleofector Kit V according to manufacturer’s instructions. GFP expression was
assessed using a Cytoflex flow cytometer 24 and 48 hours post transfection; 78% of
fluorescence positivity was observed at 48 hours. Neomycin selection (1mg/ml) was
performed to obtain stable SETD2 expression. One month after selection, SETD2 and
H3K36me3 were assessed by Western Blotting (WB). Clonogenic capacity was tested by
clonogenic assays. Immunofluorescence experiments were performed to assess if SETD2
forced expression was able to restore DNA damage repair by using p-H2AX (S139), RAD51,
MSH6 and THEX1 antibodies.
Results: We previously characterized the HMC-1.2 cell line as deficient for SETD2/H3K36me3,
as virtually all patients with advSM. To investigate whether SETD2 may indeed play
a tumor suppressor role in SM, we transfected HMC-1.2 cells with an ectopic SETD2
plasmid. After transfection, the morphology of cells dramatically changed; moreover,
HMC-1.2tsSETD2 showed a 70% increase in doubling time. Co-immunoprecipitation demonstrated
that tsSETD2 was able to interact with p53 and to restore its expression and activity.
We thus observed an increase in p21 and p27 associated with an accumulation of cells
at the G1/S checkpoint. Moreover, SETD2 stable transfection restored DNA damage responses,
as demonstrated by an increase in H2AX phosphorylation and RAD51 (HR), THEX1 (DNA
replication) and MSH6 (MMR) expression after UV exposure. Finally, clonogenic assays
in control and HMC-1.2tsSETD2 cells showed that: 1) SETD2 re-expression restores cell
proliferation control; 2) reduction of clonogenic growth observed after proteasome
inhibition is indeed SETD2-dependent.
Summary/Conclusion: In advSM, SETD2 is a bona fide tumor suppressor and its loss impairs
proliferation control and DNA damage responses. Its overexpression restores cell proliferation
control by stabilizing p53 activity, and DNA damage repair by rescuing the H3K36me3
mark. This validates the therapeutic potential of interfering with the mechanisms
responsible for SETD2 loss. Supported by AIRC IG 2019 grant (23001).
P994: HEDGEHOG PATHWAY INVOLVEMENT IN MASTOCYTOSIS DEVELOPMENT
L. Polivka1,2, V. Parietti3, J. Bruneau4, E. Soucie5, P. Dubreuil5, C. Bodemer2, O.
Hermine6, L. Maouche Chretien7,*
1IMAGINE Institute, Paris-Centre University, INSERM U1163; 2Department of Dermatology,
Hôpital Necker-Enfants Malades; 3Department of animal experimentations, Institut Universitaire
d’Hématologie, Université Paris Diderot, Sorbonne Paris Cité; 4Department of Pathology,
Necker-Enfants Malades Hospital, Paris; 5Centre de Recherche en Cancérologie de Marseille,
INSERM U1068, Marseille; 6Department of Hematology, Necker-Enfants Malades Hospital;
7Imagine Institute, Paris-Centre University, INSERM 1163, Paris, France
Background: Mastocytosis is a rare and heterogenous disease characterized by abnormal
accumulation and activation of mast cells in one or several organs. The clinical spectrum
of the pathology varies from relatively mild forms with isolated skin lesions to very
aggressive forms with wide systemic involvement, often fatal. The disease can affect
both children and adults. While most pediatric forms resolve during or in late puberty,
adult cases are persistent and may evolve into aggressive forms. A somatic KIT D816V
mutation is detected in 85% of patients but all attempts to demonstrate its oncogenic
effect have failed. There is strong evidence that the KIT D816V mutation alone cannot
trigger the disease and that other genes may be involved.
Aims: The French national reference center of mastocytosis (CEREMAST) has recorded
more than 2000 patients with sporadic mastocytosis and 60 familial cases. Since KIT
mutations do not fully explain the pathophysiology of the disease, we sought to identify
other mutations that could act synergistically with KIT D816V to induce the disease.
Methods: We focused on patients presenting with mastocytosis in particular syndromic
contexts within the pediatric cohort followed at Necker hospital; we first investigated
the molecular mechanisms underlying mastocytosis onset in patients with both congenital
mastocytosis and an extremely rare polymalformative syndrome.
Results: From 3 children presenting with both Greig cephalopolysyndactyly syndrome
(GCPS) and congenital mastocytosis, we could demonstrate the implication of the hedgehog
(Hh) pathway in mastocytosis. GCPS is a very rare syndrome resulting from haploinsufficiency
of GLI3, the major repressor gene of the hedgehog (Hh) family. We showed that the
Hh pathway is barely active in normal primary mast cells (MCs) and overactive in neoplastic
MCs. Using a GCPS mouse model, we demonstrated that mutations in GLI3 and KIT have
a synergistic, tumorigenic effect leading to mastocytosis onset. We also showed that
Hh inhibitors suppress abnormal mast cell proliferation, in vitro, and extend the
survival of mice with aggressive systemic mastocytosis (ASM).
Summary/Conclusion: Thus, for the first time, we revealed the involvement of Hh signaling
pathway in the pathophysiology of mastocytosis and demonstrated the cooperative effects
of the KIT and Hh oncogenic pathways in ASM. Finally, we provided the first evidence
that Hh inhibitors represent a promising new therapeutic target.
P995: MYELOID NEOPLASMS-ASSOCIATED GENE VARIANTS IN 639 PATIENTS WITH POST-POLYCYTHEMIA
VERA AND POST-ESSENTIAL THROMBOCYTHEMIA MYELOFIBROSIS: AN ANALYSIS OF THE MYSEC COHORT
B. Mora1,2,*, P. Guglielmelli3, A. Kuykendall4, M. Maffioli1, G. Rotunno3, R. S. Komrokji4,
F. Palandri5, J.-J. Kiladjian6, A. Iurlo7, G. Auteri5, D. Cattaneo7, V. De Stefano8,
S. Salmoiraghi9,10, T. Devos11, F. Cervantes12, M. Merli1, A. Campagna13, G. Benevolo14,
M. Brociner1, F. Albano15, J. Gotlib16, M. Caramella17, M. Ruggeri18, D. M. Ross19,
F. Orsini20, C. Pessina20, I. Colugnat20, F. Pallotti2,20, T. Barbui10, L. Bertù2,
M. G. Della Porta13, A. M. Vannucchi3, F. Passamonti1,2
1Hematology, Ospedale di Circolo, ASST Sette Laghi; 2Department of Medicine and Surgery,
University of Insubria, Varese; 3Center for Research and Innovation of Myeloproliferative
Neoplasms, University of Florence, Florence, Italy; 4Malignant Hematology Department,
Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute,
Tampa, Florida, United States of America; 5Institute of Hematology, Azienda Ospedaliero-Universitaria
S.Orsola-Malpighi, Bologna, Italy; 6Centre d’Investigations Cliniques, Hôpital Saint-Louis,
Paris, France; 7UOC Hematology, Foundation IRCCS Ca’Granda Ospedale Maggiore Policlinico,
Milan; 8Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università
Cattolica del Sacro Cuore, Rome; 9Hematology and Bone Marrow Transplant Unit; 10FROM
Research Foundation, ASST Papa Giovanni XXIII, Bergamo, Italy; 11Hematology, University
Hospitals Leuven, Rega Institute, Leuven, Belgium; 12Hematology, Hospital Clinic,
IDIBAPS, University of Barcelona, Barcelona, Spain; 13Cancer Center, IRCCS Humanitas
Clinical and Research Center, Rozzano; 14Hematology, Azienda Universitario-Ospedaliera
Città della Salute e della Scienza di Torino, Turin; 15Hematology - Dept. of Emergency
and Organ Transplantation, University of Bari, Bari, Italy; 16Division of Hematology,
Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California,
United States of America; 17Hematology, ASST Grande Ospedale Metropolitano Niguarda,
Milan; 18Hematology, San Bortolo Hospital, Vicenza, Italy; 19Haematology Directorate,
SA Pathology, Royal Adelaide Hospital and Flinders Medical Centre, Adelaide, Australia;
20Laboratory of Cytogenetics and Molecular Biology, Ospedale di Circolo, ASST Sette
Laghi, Varese, Italy
Background: Polycythemia vera (PV) and essential thrombocythemia (ET) are myeloproliferative
neoplasms evolving to post-PV (PPV-) and post-ET (PET-) myelofibrosis (MF), called
secondary MF (SMF). In primary MF, knowledge on non-driver myeloid neoplasms-associated
gene variants (M-GVs) influences clinical decision-making. In SMF, information on
M-GVs is scant.
Aims: The primary aim was to assess the pattern of distribution of M-GVs, their correlations
with SMF subtype and with driver mutations, in a large cohort of SMF patients studied
by next generation sequencing (NGS).
Methods: The study involved SMF patients of the MYSEC (Myelofibrosis Secondary to
PV and ET) project. In 97% of cases, NGS were performed within one-year pre/post-SMF
diagnosis. Characteristics of study cohort were described by standard statistic. Associations
were investigated by Chi-square or Fisher exact test.
Results: The clinical features of the 639 NGS-annotated patients entering the analysis
are reported in Table 1.
Table 1.
Characteristics of 639 NGS-annotated SMF.
SMF (639)
PPV-MF (290)
PET-MF (349)
p
Age at SMF (years), mean (SD)
62.2 (11.8)
63.2 (10.7)
61.4 (12.5)
0.17
Male gender, n (%)
323 (51)
151 (52)
172 (49)
0.48
JAK2 mutated, n (%)
477 (75)
290 (100)
187 (53)
CALR mutated, n (%)
125 (19)
125 (36)
MPL mutated, n (%)
24 (4)
24 (7)
Triple negative, n (%)
13 (2)
13 (4)
Hb (g/dl), mean (SD)
11.3 (2.1)
12.0 (2.1)
10.7 (1.9)
<0.0001
WBC (x10^9/l), mean (SD)
13.7 (12.7)
15.8 (13.9)
11.9 (11.3)
<0.0001
PLT (x10^9/l), mean (SD)
368.5 (248.4)
310.6 (211.6)
417.8 (266.4)
<0.0001
Blasts (%), mean (SD)
0.7 (1.9)
0.7 (1.6)
0.7 (2.1)
0.59
Constitut. symptoms, n (%)
259 (41)
134 (46)
125 (36)
0.01
A total of 198 (31%) patients did not harbor any M-GV, with no imbalance as for SMF
subtype. Among the 441 (69%) with M-GVs, 223 (51%) had one, 137 (31%) two, 52 (12%)
three, 23 (5%) four and 6 (1%) five or more. PPV-MF subjects reported more frequently
one M-GV, while those with PET-MF at least three (p=0.02). Mean number of M-GVs was
1.4 per patient (range, 0-7). In detail, it was 1.2 (range, 0-4) and 1.5 (range, 0-7)
per patient in PPV- and in PET-MF, respectively (p=0.01). The most frequent (≥5% of
dataset) M-GVs involved: ASXL1 (n=181, 41%), TET2 (n=145, 33%), DNMT3A (n=49, 11%),
TP53 (n=43, 10%), EZH2 (n=39, 9%), SF3B1 (n=31, 7%), U2AF1 (n=29, ~7%), ZRSR2 (n=27,
6%), CBL and RUNX1 (n=21 each, 5%). In PET-MF there was a significantly higher frequency
of M-GVs in ASXL1 (47% vs 34%, p=0.01), SRSF2 (5% vs 1%, p=0.01), U2AF1 (9% vs 4%,
p=0.04) and CBL (7% vs 2%, p=0.01) compared to PPV-MF. The latter was significantly
associated with ETV6 alterations (5% vs 1% in PET-MF, p=0.04). As regards driver mutations,
we found an association between “triple negative” status (TN) and M-GVs in SETBP1
(38%, p=0.002), IDH2 (25%, p=0.02), EZH2 (25%, p=0.05), and SRSF2 (25%, p=0.01). Figure
1 shows the frequency of M-GVs found in the MYSEC cohort, distinguished by SMF subtype
(a) and driver mutations (b).
Image:
Summary/Conclusion: Among 639 NGS-annotated SMF cases, 69% presented at least one
M-GV with a mean of 1.4 per patient. Overall, the most frequent (≥10%) M-GVs were
in ASXL1, TET2, DNMT3A and TP53 genes. Different pathways of progression among PPV-
and PET-MF have been disclosed. PPV-MF showed an increased rate of ETV6 alterations,
opening the question of possible predisposing genetic factors. TN cases clustered
with specific M-GVs, potentially targetable (IDH2). This is the first study exploring
the mutational landscape of a wide cohort of SMF patients, paving the way for further
investigations on the topic.
P996: UTILITY OF KIT P.D816 IN MYELOID NEOPLASM WITHOUT DOCUMENTED SYSTEMIC MASTOCYTOSIS
TO DETECT HIDDEN MAST CELLS IN BONE MARROW
D. H. Kim1,*, F. Jia1, C. Y. Ok1
1Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, United
States of America
Background:
KIT p.D816 mutation is strongly associated with systemic mastocytosis (SM). Although
it is not specific for SM, it is one of the diagnostic criteria for SM since its presence
in the right morphologic context (i.e. presence of atypical mast cell clusters) is
a strong support for neoplastic nature of mast cells. Next-generation sequencing (NGS)
is now routinely performed in almost all bone marrow sample and KIT mutations are
detected from patients who are not known or suspected to have SM.
Aims: Therefore, we wanted to assess if KIT mutations in this patient population are
associated with unsuspected SM.
Methods: We searched NGS result in our institution between 1/1/2013 and 9/30/2021
with positive result for KIT mutation from patients with known/suspected myeloid neoplasms.
Patients with previously documented history of systemic mastocytosis were excluded.
Bone marrow biopsies from patients with KIT mutation were assessed with immunohistochemical
stains for CD117 and mast cell tryptase (MST).
Results: A total of 49 patients who had KIT mutation and had available formalin-fixed
paraffin embedded blocks were identified. Thirty-eight (77.6%) patients has acute
myeloid leukemia (AML) and 6 patients had chronic myelomonocytic leukemia (CMML, 12.2%).
The remaining patients had myelodysplastic syndrome (n=1), myelodysplastic/myeloproliferative
neoplasm, unclassifiable (n=1), myeloproliferative neoplasm (MPN, n=1), AML in remission
(n=1) and therapy-related myeloid neoplasm (n=1).
A total of 41 patients had a single KIT mutation and 8 patients had 2 or more KIT
mutations. KIT p.D816V mutation was the most common mutation (n=38, 64.4%), followed
by D816Y (n=8, 13.6%), D816H (n=5, 8.5%), Y418_D419insFF (n=2, 3.4%), R815_D816insVL
(n=1, 1.7%), S197L (n=1, 1.7%), K558E (n=1, 1.7%), D419del (n=1, 1.7%), N822K (n=1,
1.7%) and R815_D816insIPP (n=1, 1.7%).
Immunohistochemical stains for CD117 and MST were performed in all 49 patients. The
MST stain was more suitable since 10 patients (20%) had increased blasts with CD117
expression which obscured morphologic review. A total of 4 patient (8.2%) showed mast
cell nodules where spindled shaped mast cells were present, meeting the criteria for
SM. All four patients had KIT p.D816V mutation and had high mutant allelic frequency
(~50%) except one patient (1%). These patients had MPN (n=1), AML (n=1), AML in remission
(n=1) and CMML (n=1), respectively. One (2%) patient has increased (>10%) mast cells
but scattered throughout the bone marrow space without nodules and mast cells were
round-to-ovoid shape. Eleven patients (22.4%) showed rare scattered mast cells and
33 patients (67.3%) did not show mast cells at all.
Summary/Conclusion: We discovered approximately 8% of patients who had myeloid neoplasms
with unexpected KIT mutations are met for systemic mastocytosis after additional immunohistochemical
studies. Our data support that application of additional immunohistochemical studies
are recommended to identify underrecognized SM when KIT mutations are unexpectedly
found by molecular assays.
P997: PROTEOMIC ANALYSIS ON PLATELETS OF ESSENTIAL THROMBOCYTHEMIA PATIENTS UNDERSCORES
THE ROLE OF MITOCHONDRIA IN JAK2 V617F PLATELET REACTIVITY AND FUNCTION
X. Guerrero-Carreño1, S. Smits2, A. Esteban Lasso3, J. A. Escudero García3, M. Samiotaki4,
A. Alvarez-Larrán5, A. Angona6, A. J. Sáen Marín7, V. García Gutiérrez7, D. Dekkers8,
J. Demmers8, C. M. Benavente Cuesta9, F. Ferrer-Marín10, J. C. Hernández-Boluda11,
F. J. Iborra12, I. M. De Cuyper13, P. Vandenberghe14, P. Papadopoulos1,*
1Hematology, IdISSC, Madrid, Spain; 2Human Genetics, KULeuven, Leuven, Belgium; 3Sanidad
Animal and VISAVET, Universidad Complutense de Madrid, Madrid, Spain; 4Institute for
Bioinnovation, BSRC “Al. Fleming”, Vari, Greece; 5Hematology, Hospital Clínic; 6Hematology,
Hospital del Mar, Barcelona; 7Hematology, Hospital Ramón y Cajal, IRICYS, Madrid,
Spain; 8Proteomics Center, ErasmusMC, Rotterdam, Netherlands; 9Hematology, Hospital
Clínico San Carlos, Madrid; 10Hematology, Hospital Morales Meseguer, UCAM, IMIB, Murcia;
11Hematology, Hospital Clínico Universitario, INCLIVA; 12Biological Noise and Cell
Plasticity, CIPF, Centro de Investigación Príncipe Felipe, Valencia, Spain; 13Blood
Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam, Netherlands;
14Hematology, UZLeuven, Leuven, Belgium
Background: Essential thrombocythemia (ET) is a heterogeneous disease subdivided into
five genetic groups according to WHO, based on the common MPN driver mutations (JAK2
V617F, MPL W515K/L, and CALR Type I&II). Current treatment strategies include anti-platelet
(e.g. acetylsalicylic acid (ASA)) or cytoreductive agents (e.g. hydrea (HU)) and target
mainly complications related to platelet dysfunction (i.e. hemorrhage/thrombosis).
Platelet activation is an energy demanding process fueled mainly by a dynamic equilibrium
between mitochondria oxidative phosphorylation and glycolysis1. Importantly, altering
the platelet catabolic response to activation has been shown to prevent thrombus formation2.
JAK2 V617F platelets have been reported to be more activated than CALR mutated platelets3,
as well as JAK2 mutated patients have a greater thrombotic risk in comparison to other
ET groups. However, the cause of this phenotype is not completely understood.
Aims: In this study we aimed to analyze the proteome of ET platelets and to characterize
their functional properties according to the mutational background and treatment regimens.
Methods: 22 healthy donors (HD) and 67 ET patients have been included in the study.
Most of the patients were treated with low dose aspirin (ASA), anagrelide (ANA) or
hydrea (HU) or a combination of these. Specifically, 35 out of the 67 ET platelet
samples and 8 out of 22 HD were subjected to label-free quantitation mass spectrometry
(LFQ-MS). All HD and ET platelets were also analyzed for surface marker expression,
degranulation and aggregation capacity by flow cytometry.
Results: Hierarchical clustering of the mass spectrometry data revealed different
proteomic profiles between HD and ET mutational groups and specifically between JAK2
V617F and CALR I and CALR Type II treated and/or untreated platelets. In general,
HD platelets were more enriched for proteins related to platelet activation, and degranulation
as compared to ET platelets and clustered next to JAK2 V617F ASA-treated platelet
samples (Figure 1). In particular, the JAK2 V617F ASA-treated platelets presented
significant enrichment in platelet activation metabolic and mitochondrial proteins
in contrast to the CALR Type I and Type II ET platelets. Of note, no peptides corresponding
to the mutant CALR were detected using label-free MS methods. Surface marker expression
was variable among ET patients. However, CD49B and CD36 surface markers were the most
affected among the JAK2V617F and CALR Type I and II platelets and their levels (mean
fluorescence intensity-MFI-) were inversely correlated. With regards to platelet aggregation,
JAK2 V617F platelets presented similar or lower levels when compared to HD platelets,
in line with their proteomic profiles. Differences in aggregation and degranulation
levels were also observed between CALR Type I and Type II platelets specifically in
the untreated condition.
Image:
Summary/Conclusion: ET platelets are functional but they show different capacities
to respond upon various stimuli and treatment regimens. JAK2 V61F platelets are more
activated than any other ET group according to their proteome profile, which is not
fully reflected by the functional assays. Mitochondrial activity arises as an important
factor in the control of platelet reactivity and it could be critical in disease management
and treatment strategies, specifically for the JAK2 V617F patients.
References
1. Aibibula, M., et al. J Thromb Haemost. 2018; 16(11):2300-2314,
2. Nayak, M.K., et al. Blood Adv. 2018; 2(15):2029-2038,
3. Hauschner, H., et al. Am J Hematol, 2020. 95(4): p. 379-386.
P998: REGULATION OF BCL-X SPLICING BY SRC KINASES INHIBITORS IN MYELOPROLIFERATIVE
NEOPLASMS
J. Petiti1,*, F. Itri1, C. Fava1, D. Cilloni1
1Dept. of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
Background: Myeloproliferative neoplasms (MPN) are a group of incurable diseases that
include essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis
(PMF). Both ET and PV are associated with prolonged clinical courses, with symptoms
that affect their quality of life, and risk of progression to MF and acute leukemia.
MF is the MPN subtype with the most adverse prognosis and is associated with a high
risk of leukemic transformation. The FDA approved the use of ruxolitinib, a JAK1/2
kinase inhibitor, that proved to be effective in reducing symptoms and spleen volume,
but not in inducing complete molecular remission. Although hematopoietic stem cell
transplantation involves a high risk of mortality, it is nowadays the only curative
treatment for MF. Therefore, it is relevant to identify new therapeutic strategies
to improve the clinical outcome of MPN. Recently, there has been a growing interest
in Bcl‐xL, the long isoform encoded by alternative splicing of the Bcl‐x gene, that
acts as an anti‐apoptotic regulator. Bcl-xL appears to be up-regulated in MPN patients
and we previously showed that its modulation correlates with the clinical severity
of MPN subgroups. We also demonstrated that ABT-737, a specific Bcl-xL inhibitor,
has a synergistic effect in combination with Ruxolitinib in HEL cell lines and MPN
patients’ cells, suggesting that Bcl-xL inhibition could increase the power of treatment
(Petiti et al. J Cell Mol Med. 2020). Unfortunately, ABT-737 is characterized by poor
bioavailability, so a new approach to target Bcl-xL must be investigated. Paronetto
et al. found that tyrosine phosphorylation of Sam68 in cancer cells may protect them
from apoptosis by altering the Bcl-xS/L ratio in favor of Bcl-xL. Intriguingly, c-Src
is one of the tyrosine kinases that phosphorylate Sam68 and several papers indicated
its involvement in the MPN pathogenesis, without explaining the molecular mechanism.
Aims: We aimed to molecular investigate the mechanism linking c-Src kinase activity
with the deregulation of Bcl-x splicing in MPN, intending to consider c-Src kinase
inhibitors, widely bioavailable, as indirect modulators of Bcl-xL to improve MPNs
outcome.
Methods: HEL cell lines, JAK2 V617F-mutated in homozygosis, has been incubated with
different concentrations of ruxolitinib (JAK1/2 inhibitor) and dasatinib (SRC kinases
inhibitor) at different concentrations and time points. Then, proliferation, and apoptosis
were evaluated. Subsequently, the c-Src pathway, Sam68, and Bcl-xL/S ratio were investigated
at both RNA and protein levels.
Results: Performing in vitro study on HEL cell lines, we identified that dasatinib
induced apoptosis and inhibition of proliferation in HEL cell lines, with a synergistic
effect in combination with ruxolitinib. We also showed that dasatinib alone inhibited
c-Src and Sam68 phosphorylation and down-modulated Bclx-L protein expression. Combination
treatment with ruxolitinib and dasatinib increased both pro-apoptotic Cleaved Caspase
3 and BAX protein expression. Furthermore, we noted that treatment with ruxolitinib
alone decreases the mRNA levels of Bclx-L, but also of the Bcl-xS isoform. On the
contrary, combined drug therapy modulated the Bcl-xL/S mRNA ratio in favor of the
pro-apoptotic Bcl-xS isoform.
Summary/Conclusion: Although further studies will be necessary to better understand
the c-Src role in Bcl-x splicing regulation in MPN, our preliminary data suggest that
indirect downmodulation of Bcl-xL through SRC kinases inhibitors in combination with
standard therapy could be useful in the future for the treatment of MPN patients.
P999: ERK1/2 INHIBITION REDUCES OSTEOPONTIN PLASMA LEVELS AND BONE MARROW FIBROSIS
IN A MYELOFIBROSIS MOUSE MODEL
S. Rontauroli1,*, E. Bianchi1, L. Tavernari1, M. Dall’Ora2, G. Grisendi3, M. Mirabile1,
S. Sartini1, E. Genovese1, C. Carretta1, S. Mallia1, S. Parenti1, L. Fabbiani4, N.
Bartalucci5, L. Losi6, M. Dominici3, A. M. Vannucchi5, R. Manfredini1
1Centre for Regenerative Medicine, Life Sciences Department, University of Modena
and Reggio Emilia, Modena; 2Rigenerand S.R.L., Medolla (MO); 3Division of Oncology,
Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences of Children
& Adults; 4Department of Medical and Surgical Sciences of Children & Adults, Pathology
Unit, University of Modena and Reggio Emilia, Modena; 5Department of Experimental
and Clinical Medicine, and Center Research and Innovation of Myeloproliferative Neoplasms
(CRIMM), University of Florence, Careggi University Hospital, Florence; 6Department
of Life Sciences, Pathology Unit, University of Modena and Reggio Emilia, Modena,
Italy
Background: Primary myelofibrosis (PMF) is a stem cell disorder belonging to Philadelphia-negative
myeloproliferative neoplasms (MPNs). The disruption of bone marrow (BM) microenvironment
due to the extensive deposition of extracellular matrix fibers is the distinctive
trait of PMF and is accompanied by hematopoietic stem cells (HSCs) mobilization and
extramedullary hematopoiesis. BM fibrosis is caused by the complex interaction between
stromal and hematopoietic cells belonging to the neoplastic clone, in particular megakaryocytes
and monocytes play a pivotal role through the production of pro-fibrotic cytokines.
Supporting this hypothesis, we have previously demonstrated that osteopontin (OPN)
contribute to the development of BM fibrosis. OPN plasma levels are increased in PMF
patients and correlate with higher BM fibrosis grade, circulating CD34+ cells and
inferior survival. Megakaryocytes and monocytes turned out as the main source for
OPN production that can induce fibroblasts and mesenchymal stromal cells proliferation,
as well as collagen upregulation.
Aims: To determine whether OPN might represent a druggable target in PMF we assessed
the inhibition of signaling pathways responsible for its production. In particular,
we evaluated the effect of ERK1/2 inhibition over OPN expression and myelofibrosis
development in a MF mouse model.
Methods: The activity of inhibitors of signaling pathways affecting OPN expression
was evaluated in vitro in human primary monocytes, given their key role in OPN secretion.
The effects on cell viability were assessed by XTT assay at 72 hours of treatment,
while OPN expression and production were evaluated by qRT-PCR and ELISA, respectively,
at 72 and 96 hours of treatment.
Inhibitors of OPN production selected from the in vitro screening were analyzed for
their effect on OPN plasma levels in a MF mouse model induced by the treatment with
a thrombopoietin receptor agonist Romiplostim (Rom). Hematological parameters and
splenomegaly were monitored over time while BM and spleen fibrosis were evaluated
at sacrifice.
Results: Our in vitro analysis demonstrated that drug inhibitors of ERK1/2, MEK1/2,
p38 and a Ca2+ channel antagonist were able to reduce OPN expression in primary monocytes,
both at RNA and protein levels, without affecting cell viability.
Next, we moved to a MF mouse model obtained through the stimulation of thrombopoietin
receptor that develops bone marrow and spleen fibrosis, together with increased OPN
expression, faster than JAK2V617F knock-in mice. ERK1/2 inhibition by Ulixertinib
did not affect the development of thrombocytosis and splenomegaly but was able to
significantly reduce OPN plasma levels. Interestingly, ERK1/2 inhibition also counteracted
the development of bone marrow fibrosis in Rom-treated mice. The same results were
observed in mice when Ulixertitnib was given together with the JAK inhibitor Ruxolitinib.
Drug combination resulted in a reduction of both OPN plasmatic levels and bone marrow
fibrosis development.
Summary/Conclusion: Our results demonstrated that Ulixertinib treatment reduced OPN
production both in vitro and in vivo. Moreover, in a MF mouse model, the concurrent
inhibition of ERK1/2 and JAK2 signaling pathways displayed synergistic effects by
diminishing OPN plasma levels and constraining BM fibrosis. These results provide
a rational for the development of novel combination therapeutic approach for PMF patients.
P1000: INCREASED PLASMA LEVELS OF LNCRNAS ARE POTENTIAL PROGNOSTIC BIOMARKERS IN MYELOFIBROSIS
S. Sartini1,*, S. Fantini1, S. Rontauroli1, M. Mirabile1, E. Bianchi1, F. Badii1,
M. Maccaferri2, P. Guglielmelli3, T. Ottone4,5, R. Palmieri4, E. Genovese1, C. Carretta1,
S. Parenti1, S. Mallia1, L. Tavernari1, C. Salvadori3, F. Gesullo3, C. Maccari3, M.
Zizza3, A. Grande6, S. Salmoiraghi7, B. Mora8, L. Potenza9, V. Rosti10, F. Passamonti8,
A. Rambaldi7, M. T. Voso4,5, C. Mecucci11, E. Tagliafico9,12, M. Luppi9, A. M. Vannucchi3,
R. Manfredini1
1Center for Regenerative Medicine, Life Sciences Department, University of Modena
and Reggio Emilia; 2Department of Laboratory Medicine and Pathology, Diagnostic Hematology
and Clinical Genomics, AUSL/AOU Policlinico, Modena; 3Department of Experimental and
Clinical Medicine, and Center Research and Innovation of Myeloproliferative Neoplasms
(CRIMM), University of Florence, Careggi University Hospital, Florence; 4Department
of Biomedicine and Prevention, University of Tor Vergata; 5Neuro-Oncohematology, Santa
Lucia Foundation, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.),
Rome; 6Department of Biomedical, Metabolic and Neural Sciences, University of Modena
and Reggio Emilia, Modena; 7Hematology, ASST Papa Giovanni XXIII, Bergamo; 8Division
of Hematology, Ospedale ASST Sette Laghi, University of Insubria, Varese; 9Department
of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico,
Modena; 10Center for the Study of Myelofibrosis, Foundation Policlinico San Matteo,
Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Pavia; 11Department
of Medicine and Surgery, Section of Hematology and Clinical Immunology, University
of Perugia, Perugia; 12Center for Genome Research, University of Modena and Reggio
Emilia, Modena, Italy
Background: Myelofibrosis (MF) displays the worst prognosis among Philadelphia-negative
myeloproliferative neoplasms and is characterized by megakaryocyte hyperplasia, progressive
bone marrow fibrosis, extramedullary hematopoiesis and frequent transformation to
acute myeloid leukemia.
Different therapeutic approaches are being used depending on the severity and the
specific clinical manifestations of the disease in each patient. Unfortunately, no
curative therapy is currently available for MF, except for bone marrow transplantation,
which however has a consistent percentage of failure. There is therefore a great urgency
to identify biomarkers correlated with the different stages of the disease and to
treat patients with a more tailored treatment.
Long non-coding RNAs (lncRNAs) have been recently described as key mediators in the
development of hematological malignancies. Moreover, circulating lncRNAs have already
been proposed as a new class of non-invasive biomarkers for cancer diagnosis and prognosis.
Aims: The aim of this study was to identify circulating lncRNAs whose plasmatic concentration
differs between MF patients and healthy donors (HDs). Subsequently, we evaluated their
potential role as non-invasive disease biomarkers in MF.
Methods: As a preliminary result we analyzed the expression of 38 lncRNAs in CD34+
cells collected from 83 MF patients and 26 HDs leading to the identification of 26
differentially expressed lncRNAs. To confirm these results and to move to a more accessible
sample type we collected plasma from 143 MF patients and 65 HDs. RNA was extracted
from these samples and the relative abundance of lncRNAs, including some of those
deregulated in CD34+ cells or already described as involved in hematological malignancies
and myeloid differentiation, was assessed using qRT-PCR. According to the plasmatic
levels of each lncRNA patients were split into two groups (low- or high-) and this
subdivision was used to unveil the potential correlation between the various lncRNAs
and the clinical features of MF patients.
Results: Our analysis identified 7 lncRNAs significantly upregulated in MF patients’
plasma compared to HDs. Among these, high levels of LINC01268, MALAT1 or GAS5 correlated
with several detrimental clinical features of MF, such as high counts of leukocytes
and CD34+ cells, a severe grade of bone marrow fibrosis and the presence of splenomegaly.
Strikingly, high plasma levels of LINC01268 (Log-rank p-value = 0.0018), GAS5 (Log-rank
p-value = 0.0008) or MALAT1 (Log-rank p-value = 0.0348) were associated with a poor
overall-survival (OS) while high levels of LINC01268 correlated also with a shorter
leukemia-free-survival (LFS). Finally, multivariate analysis demonstrated that a high
plasma concentration of LINC01268 was an independent prognostic variable for both
OS (HR = 2.104; confidence interval (CI) = 1.08–4.12; p = 0.0297) and LFS (HR = 8.190;
CI = 1.02–65.78; p = 0.0479).
Summary/Conclusion: To our knowledge, this is the first study describing the expression
profile of circulating lncRNAs in MF patients’ plasma and focusing on their putative
role as biomarkers in clinical practice. In particular, our results demonstrated that
increased levels of circulating LINC01268, GAS5 or MALAT1 are associated with disease
detrimental features and correlate with an inferior OS in MF patients. Notably, multivariate
analysis confirmed that LINC01268 plasma levels might improve the identification of
patients with a poor prognosis. If the prognostic value of this lncRNA will be confirmed
in independent patients’ cohorts it might be used to integrate contemporary prognostic
models.
P1001: DNMT3A/TET2/ASXL1 MUTATIONS DETERMINE THROMBOTIC RISK IN POLYCYTHAEMIA VERA
A. Segura Diaz1, R. Stuckey1,*, Y. Florido Ortega1, M. Sobas2, A. Álvarez Larrán3,
F. Ferrer-Marín4, M. Pérez Encinas5, G. Carreño6, M. Fox7, B. Tazón Vega7, B. Cuevas8,
J. López Rodriguez1, N. Farías Sánchez1, C. Bilbao Sieyro1, M. Gómez Casares1
1H.U.Dr. Negrin de GC., Las Palmas, Spain; 2Wroclaw Medical University, Wrocław, Poland;
3H. Clínic de Barcelona, Barcelona; 4Hospital Morales Messeguer, IMIB, UCAM, Murcia;
5Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela;
6Hospital Universitario 12 de Octubre, Madrid; 7, Hospital Universitari Vall d’Hebron,
Barcelona; 8Hospital Universitario de Burgos, Burgos, Spain
Background: Polycythaemia vera (PV) is the myeloproliferative neoplasm with the highest
incidence of thrombosis, with cardiovascular events being the main cause of death
in these patients. Recent studies have identified clonal haematopoiesis (CHIP) in
healthy individuals, which is associated with older age and an increased risk of developing
both myeloid neoplasms and vascular events.
Aims: In a previous study we saw an association between the presence of mutations
in DTA genes (DNMT3A, TET2 and ASXL1, the most frequently mutated genes in CHIP),
and vascular events in patients with PV. We aimed to confirm this association in a
consecutive series of PV patients from our centre and in an age-matched case control
study with patients from various European centers.
Methods: All patients aged 18 years and above with a confirmed diagnosis of PV in
our hospital were recruited consecutively between 2003 and 2018. NGS was performed
on 200 ng genomic DNA extracted from peripheral blood at diagnosis. Sequencing was
performed using a MiSeq (Illumina) with the 30-gene panel Myeloid Solution (SOPHiA
Genetics). Only variants with an allelic frequency (VAF) ≥ 2% and annotated as pathogenic
or probably pathogenic were considered. Additional mutations are defined as pathogenic
mutations in any non-driver gene from the myeloid panel.
The case-control study included PV patients with a thrombotic event in their patient
history and the results of a myeloid NGS panel available from 9 Spanish and 1 Polish
hospital. Cases were gender- and age-matched with control patients (with a diagnosis
of PV but no thrombotic event) from the consecutive series.
Results: A total of 79 patients with PV were analysed in our consecutive series. An
association was observed between DTA mutation and thrombotic event (OR 4.5; p=0.002;
χ2). An association was also observed between any non driver mutation from the myeloid
panel and thrombotic event (OR 7.7; p=<0.0001; χ2). However, no association was seen
between non-DTA mutation and thrombotic events (p=0.231; χ2). The association between
DTA mutation and thrombotic event was confirmed in multivariate analysis (p=0.021,
Table 1. Thrombotic event was associated with age (p=0.020) and marginally with leukocyte
count at diagnosis (p=0.065) but not with JAK2 VAF. We saw that when the JAK2 mutation
comes before DTA, there is a tendency for the risk of thrombosis to be higher than
for DTA first.
We also observed a positive association between cardiovascular risk factors (CVRF)
and thrombotic events (Table 1), as expected. However, its significance was lost in
the multivariate analysis. The DTA and CVRF variables are closely related, with a
positive association between them (OR 6.77; p=0.009; χ2). Fifty eight of 79 patients
(73.4%) were hypertensive; however, an association was still found between thrombotic
event and DTA mutation in this group of hypertensive patients (p=0.033).
The association between DTA and thrombotic events in the uni and multivariate analyses
was confirmed by the case-control study of 47 cases and 47 gender- and age-matched
controls (OR 2.7; p=0.036; χ2).
Image:
Summary/Conclusion: We were able to confirm the previously observed association in
PV between the presence of DTA mutations and higher risk of developing a vascular
event. Thus, detection of DTA mutation by NGS could help predict thrombotic risk in
PV patients, including those with pre-existing CVRF.
P1002: ANTI-FIBROTIC ACTIVITY OF BMP2 IN BONE MARROW-DERIVED MESENCHYMAL STROMAL CELLS
OF MYELOPROLIFERATIVE NEOPLASMS
T. Subotički1,*, E. Živković1, O. Mitrović Ajtić1, M. Vukotić1, D. Đikić1, T. Dragojević1,
D. Šefer2, S. Bižić2, J. F. Santibanez1, M. Gotić3, V. P. Čokić1
1Department for Molecular Oncology, Institute for Medical Research, National Institute
of Republic of Serbia; 2Clinic of Hematology, University Clinical Center of Serbia;
3Faculty of Medicine, University of Belgrade, Belgrade, Serbia
Background: Myeloproliferative neoplasms (MPN) are clonal hematopoietic disorders
that include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis
(PMF). Bone marrow fibrosis (BMF) is a shared feature of all MPN, although it is most
pronounced in PMF and represents a major diagnostic criteria. Studies suggest a correlation
between the grade of BMF and prognosis of MPN, with more fibrosis associated with
worse outcome. A large body of evidence has suggested that transforming growth factor
beta (TGF-β) is among the most prominent inducers of fibrotic processes. Bone morphogenetic
proteins (BMPs) are major regulators of cell fate in tissue homeostasis. In MPN, bone
marrow-derived mesenchymal stromal cells (BM-MSC) are identified as a major cellular
source of fibrosis, but exact molecular mechanism involved have not been identified
so far.
Aims: In addition to apoptosis and prolifration, we analyzed the effect of BMP2 and
TGF- β / SMAD signaling pathway on the fibrotic phenotype of BM-MSC isolated from
MPN patients and healthy donors.
Methods: Bone marrow aspirates from 5 newly diagnosed MPN patients (3 PMF and 2 PV
patients) and 3 healthy donors were analyzed by immunofluorescence expression of fibronectin
and alpha smooth muscle Actin (αSMA), after treatment with TGF-β and / or BMP2. Using
immunocytochemistry, we analyzed HEL 92.1.7 cells with a homozygous expression of
JAK2V617F for proliferation (Ki67) and apoptosis (ssDNA) during exposure to BMP2 and
selective BMP signaling inhibitor LDN-193189.
Results: Our results showed that TGF-β significantly increased fibronectin expression,
in contrast to BMP2, in BM-MSC of healthy donors. Also, the joint treatment of TGF-β
and BMP2 reduced the level of fibronectin expression relative to TGF-β. In addition,
TGF-β increased αSMA expression in BM-MSC from healthy donors. In contrast, BMP2 reduces
the expression of αSMA and fibronectin in BM-MSC of healthy donors.BMP2 significantly
reduced fibronectin expression in BM-MSC compared to untreated cells of patients with
MPN. BMP2 dose dependently and significantly (p<0.01) increased the proliferation
of HEL 92.1.7 cells, while the BMP signaling inhibitor LDN-193189 also demonstrated
dose dependence in stimulation of proliferation (p<0.001). Apoptosis of HEL 92.1.7
cells was slightly affected by BMP2 and LDN-193189.
Summary/Conclusion: Our results show that BMP2 has anti-fibrotic activity that is
antagonistic to TGF-β. This indicate that BMP2 may modify the TGF-β signaling pathway.
P1003: PERIPHERAL BLOOD CYTOTOXIC T CELLS SHOW EARLY EXHAUSTED FEATURES IN MYELOFIBROSIS
PATIENTS
L. Tavernari1,*, S. Rontauroli1, M. Maccaferri2, B. Mora3, E. Bianchi1, S. Parenti1,
E. Genovese1, P. Guglielmelli4, C. Carretta1, S. Mallia1, M. Mirabile1, S. Sartini1,
C. Colasante5, L. Potenza5, F. Passamonti3, E. Tagliafico5,6, M. Luppi5, A. M. Vannucchi4,
R. Manfredini1
1Centre for Regenerative Medicine, Life Sciences Department, University of Modena
and Reggio Emilia; 2Department of Laboratory Medicine and Pathology, Diagnostic Hematology
and Clinical Genomics, AUSL/AOU Policlinico, Modena; 3Division of Hematology, Ospedale
ASST Sette Laghi, University of Insubria, Varese; 4Department of Experimental and
Clinical Medicine, and Center Research and Innovation of Myeloproliferative Neoplasms
(CRIMM), University of Florence, Careggi University Hospital, Firenze; 5Department
of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Policlinico;
6Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
Background: Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) characterized
by megakaryocyte hyperplasia, bone marrow fibrosis and extramedullary hematopoiesis.
Compelling evidence are suggesting that persistent antigen stimulation in the tumor
microenvironment can differentiate T effector cells into terminally exhausted T cells,
a functional state characterized by decrease in proliferation, cytotoxicity, and cytokine
production. Cutting-edge therapies are focused on the reversion of exhausted state
through immune checkpoint inhibition to recover the immune response against the tumor.
Aims: Despite MPNs are myeloid neoplasms, T cells are showing dysfunctional features.
To assess if immune checkpoint inhibition could be a new effective therapy in combination
with those currently approved, we investigated T cell exhaustion in MF.
Methods: Peripheral blood mononuclear cells (PBMCs) were isolated from 38 MF patients
and 20 healthy donors (HD). The expression profile of the following inhibitory receptors
(IRs): PD1, CD244, CD160, TIM3, LAG3, CTLA4 was characterized by flow cytometry in
CD3+CD8+ T cells. Results were correlated with clinical features such as Dynamic International
Prognostic Scoring System (DIPSS) classification, splenomegaly, hemoglobin level and
other negative prognostic factors. For T cell activation assays we stimulated PBMCs
overnight in the presence of coated anti-CD3, anti-CD28 and Brefeldin A, whilst cytokine
production was then assessed by intracellular flow cytometry staining. Chi-squared
and Mann Whitney tests were used for statistical analysis. Total cellular RNA was
isolated from peripheral blood granulocytes of 141 primary and secondary MF patients
and 28 HDs, and we performed Gene expression profiling (GEPs) by microarray platform.
Results: We reported expansion of PD1+CD244+, PD1+CD160+, PD1+CD57- cytotoxic T cell
populations from MF patients, together with increased expression of all the IRs assessed
on both total CD3+CD8+ T cells and PD1+CD57- T cells fraction. Furthermore, we observed
decreased secretion of IFNɣ and TNFɑ by CD3+CD8+ T cells of MF patients, while significant
reduction of Granzyme B production was observed mainly in patients with expansion
of PD1+CD57- T cell population. GEPs analysis highlighted increased expression of
IR-ligands like CD274, CEACAM1, CD48 on granulocytes from MF patients compared to
HDs. The correlation analysis of IRs expression with patients’ clinical features showed
that higher levels of PD1, CD244, CD160, CTLA4, LAG3 or TIM3 associates with detrimental
features like splenomegaly, low hemoglobin levels, HMR mutation number and more severe
DIPSS classification.
Summary/Conclusion: Our data evidenced in MF patients the presence of an impaired
population of peripheral blood cytotoxic T cells expressing multiple IRs and with
reduced cytokine production after in vitro activation. Moreover, granulocytes from
MF patients display higher level of IR-ligands transcripts hinting an immunosuppressive
interplay between the myeloid neoplastic clone and T cells. Lastly, clinical correlations
suggest a more severe disease in patients with higher IRs expression on cytotoxic
T cells. Taken together this data highlights an early exhausted state likely implicated
in immune escape which will be investigated for immune checkpoint inhibition.
P1004: EFFECT AND MOLECULAR MECHANISM OF TQ05105, A NOVEL SMALL MOLECULE INHIBITOR
OF JAK2 IN MYELOPROLIFERATIVE NEOPLASM
W. Zhang1,2,3,4,*, J. Liu1,2,3,4, Z. Shi4, L. Yang1,2,3,4, B. Li1,2,3,4, Z. Xiao1,2,3,4
1State Key Laboratory of Experimental Hematology; 2National Clinical Research Center
for Blood Diseases; 3Institute of Hematology & Blood Diseases Hospital, Tianjin; 4Chinese
Academy of Medical Science & Peking Union Medical College, Beijing, China
Background: The JAK2V617F gene mutation leads to upregulation of the JAK/STAT signaling
pathway, which causes excessive bone marrow cell proliferation, elevated inflammatory
cytokines, constitutional symptoms and shortened survival in myeloproliferative neoplasms
(MPN) patients. The JAK1/2 kinase inhibitor ruxolitinib can dramatically improve disease-related
symptoms, but side effect such as anemia and thrombocytopenia limit its use in a substantial
proportion of patients. Several JAK2 kinase inhibitor III Clinical trials have yielded
unsatisfactory outcomes recently. TQ05105 is a new small molecule inhibitor that targets
JAK2 kinase.
Aims: We aim to investigate the effect and molecular mechanism of TQ05105 on MPN cell
lines and mouse models.
Methods: We systematically measured the function of TQ05105 in MPN cell lines, including
UKE1 and SET2 cells, by performing proliferation, apoptosis and colony-forming assays.
RT-PCR and inflammatory factor multi-assay kit was used to detect the secretion of
inflammatory factors such as TNF-α, IL-1β, MCP-1 in mRNA level and protein level respectively.
CFU-E and BFU-E assays were performed on MPN cell lines and primary cells. In vivo,
TQ05105 was evaluated in JAK2V617F Jak2V617F transplant mouse model and MPL515 retroviral
mouse model. Mice were orally administrated with placebo, 25 mg/kg (bid) and 50 mg/kg
(bid) every day for 4 weeks. Flow cytometry analysis was performed to determine the
proportion of different hematopoietic progenitor cell populations. In addition, HE
staining and Gomori staining was used to detect the changes in the spleen and bone
marrow.
Results: In UKE1 and SET2 cell lines, TQ05105 can inhibit the proliferation and induced
cell apoptosis, notably reduce the expression of cytokines, including TNF-α, IL-1
and MCP-1. According to Western blot results, the phosphorylation of JAK/STAT signaling
pathway associated proteins JAK2, STAT3 and STAT5 were significantly suppressed by
TQ05105. Furthermore, compared to the control group, TQ05105 can strongly limit the
ability of UKE1 cell lines, SET2 cell lines and primary cells from MPN patients (PMF,
PV, ET) to form BFU-E and CFU-GM.
TQ05105 can striking reduce mouse spleen volume, along with favorable impact on leukocyte
and erythrocyte counts in Jak2V617F transplant mouse model. The proportion of stem
and progenitor cells, especially CMPs and MEPs, was considerably reduced in mice after
treatment with TQ05105. HE staining showed remarkably Spleen response such as recovering
structure, reduced infiltration of erythrocytes and suppressed proliferation of megakaryocytes.
MPLW515L retroviral mouse model recapitulated the feature of myelofibrosis, application
of TQ05105 in this model resulted in reduction of the spleen volume, extramedullary
hematopoiesis, leukocyte and platelets count, bone marrow fibrosis and decreased fraction
of CMPs and MEPs.
Summary/Conclusion: TQ05105, as a novel small molecule JAK2 inhibitor, can inhibit
cell proliferation, induce cell apoptosis and decrease inflammatory cytokines by affecting
the JAK/STAT signaling pathway. In vivo experiments further proved that TQ05105 can
reduce spleen size and improve disease-related symptoms of MPN, indicating that it
is a promising therapeutic drug for MPN.
P1005: A PHASE 1, OPEN-LABEL, DOSE-ESCALATION STUDY OF SELINEXOR PLUS RUXOLITINIB
IN PATIENTS WITH TREATMENT-NAÏVE MYELOFIBROSIS
H. Ali1,*, A. Kishtagari2, K. Maher3, S. Mohan2, A. Mazumder4, K. Chamoun5, I. Karasik5,
E. Sbar5, L. Dugom5, S. Tamir5, X. Wang5, J. Prchal6, S. Tantravahi6
1City of Hope, Duarte; 2Vanderbilt Ingram Cancer Center, Nashville; 3VCU Massey Cancer
Center, Richmond; 4The Oncology Institute of Hope & Innovation, St. Petersburg; 5Karyopharm
Therapeutics, Newton; 6Division of Hematology and Hematologic Malignancies, Huntsman
Cancer Institute, University of Utah, Salt Lake City, United States of America
Background: Myelofibrosis (MF) is a myeloproliferative neoplasm commonly associated
with gene mutations in JAK2, CALR, or MPL caused by the unregulated proliferation
of clonal myeloid precursors in the bone marrow. The JAK 1/2 inhibitor, ruxolitinib
(RUX), has shown reductions in spleen volume and improvement in MF-related symptoms
when used in the frontline. Despite the significant improvements of RUX, most patients
(pts) eventually progress and lose response to treatment over time. Therefore, novel
combinations are critical to improve responses and delay progression. Selinexor (SEL)
is an oral selective inhibitor of nuclear export (SINE) compound, specifically inhibiting
exportin-1 (XPO1), that has been approved for use in multiple myeloma and diffuse
large B-cell lymphoma. Preclinical studies of the combination SEL and RUX have demonstrated
significant activity. In clinical studies of MF refractory to JAK inhibitors, SEL
monotherapy has exhibited robust clinical activity with a tolerable safety profile
(NCT03627403).
Aims: Here, we present the initial results of a phase 1 dose escalation study to determine
the optimal dose and preliminary efficacy of once weekly (QW) SEL in combination with
RUX in pts with treatment-naïve MF.
Methods: In the ongoing multicenter, open-label, Phase 1/2 study (NCT04562389) using
a 3 + 3 design, two dose levels of SEL were evaluated, 40 mg and 60 mg QW plus RUX
twice daily (BID) as per label in 28-day cycles. For nausea prophylaxis, all pts received
a 5-HT3 antagonist. Primary study objectives include safety, maximum tolerated dose
(MTD), recommended Phase 2 dose (RP2D), and preliminary efficacy. Secondary objectives
include spleen volume, symptom and anemia response, and overall survival (OS).
Results: As of 24 Feb 2022, 10 pts have been dosed in 2 dose levels 40 mg (n=3), and
60 mg (n=7) SEL QW plus RUX. The starting dose of RUX was 20 mg in 8 pts, 15 mg in
one patient and 10 mg in one patient. The median age was 64 (range 45-76) and 7 pts
had primary MF and 3 had post-essential thrombocythemia (ET) MF. The Dynamic International
Prognostic Scoring System (DIPSS) risk category was int-1 (n=4), int-2 (n=4) and high
risk (n=2). There were no dose limiting toxicities reported for either SEL dose levels.
Due to dizziness, one patient had a dose interruption and after 5 months of therapy
discontinued treatment from new onset of atrial fibrillation and pulmonary hypertension
(unrelated to SEL and RUX). Currently, all other pts remain on study. Hemoglobin levels
were maintained without significant worsening in majority of patients. Low grade nausea
(30%) was the most common treatment-emergent adverse event. All pts experienced an
improvement in their white blood cell count. Of the 6 evaluable pts, 5 had a ≥35%
spleen volume reduction at week 12.
Summary/Conclusion: In pts with treatment-naïve MF, QW SEL in combination with RUX
is well tolerated with a manageable side effect profile. Based on current data there
have been no observed dose limiting toxicities in cohort 1 of QW oral SEL 40 and 60 mg
with RUX.
P1006: NEXT GENERATION SEQUENCING (NGS) IN PEDIATRIC MASTOCYTOSIS
A. Angi1,*, S. Bianchi1, G. Palumbo1, V. Filipponi1, M. Rousseau1, M. L. Moleti1,
F. Giona1
1EMATOLOGIA PEDIATRICA, Policlinico Umberto I Università Sapienza Roma, Roma, Italy
Background: Mastocytosis is a clonal disorder characterized by the accumulation of
mast cells in the skin and, less frequently, in other organs. The disease is limited
to the skin in children and may occur in one of three cutaneous variants: maculopapular
cutaneous mastocytosis (MPCM), diffuse cutaneous mastocytosis (DCM) and mastocytoma.
Systemic mastocytosis (SM) typically occurs in adults with KIT D816V mutation (3).
Additional genetic mutations (TET2, N-RAS, SF3B1, ASXL1, etc.) have been detected
using NGS in adults with mastocytosis (4). Currently, there is no data concerning
the use of NGS in pediatric mastocytosis.
Aims: The aims of the monocentric study, named MAS_PED1, that was approved by Ethics
Committee, are to: a) identify patients (pts) with different cutaneous forms at risk
of developing aggressive systemic disease; b) detect the known KIT D816V mutation
using RT-PCR; c) identify different KIT, and/or additional gene mutations using NGS;
and d) evaluate potential role of different mutations in the outcome of pts with mastocytosis,
aged < 18 years at lesion onset.
Methods: Pts with pediatric onset of mastocytosis, followed at the study Center were
included. Peripheral blood samples were collected to detect KITD816V mutation, using
both RT-PCR and ddPCR techniques, and to investigate other molecular mutations using
NGS panels for rare and myeloid genes.
Results: All 36 pts included in this study, 19 male and 17 female, with median age
of lesion onset ranged from birth to 17.81 years (median 4.74 months), had a cutaneous
mastocytosis (CM). Twenty-one of the 36 pts (58%) underwent cutaneous biopsy after
a median from lesion onset of 3.77 months (range: 2.49 months – 11.6 years). According
to the type of cutaneous lesions at diagnosis, pts were classified as: 20 (55%) MPCM,
10 (28%) DCM and 6 (17%) mastocytoma. The median tryptase value at the onset was 5 ng/ml,
higher in MPCM (range: 1.2 - 141 ng/ml), than in DCM (range: 2.71 - 19.4 ng/ml), and
in mastocytoma (range: 3.8 - 7.3 ng/ml). Two of the MPCM pts developed indolent SM
(ISM) after 10 and 20 years from the onset disease. RT-PCR and NGS assessment were
performed in 17 pts (47%), 15 CM and 2 ISM. RT-PCR identified KIT D816V mutation in
4 pts (2 MPCM, 1 DCM, 1 ISM) while NGS revealed KIT D816V in 3 pts and other KIT mutations,
KIT D816Y and KIT Y553C, in 2 pts. An additional 10 myeloid gene mutations were detected
by NGS technique: 5 were already known (ASXL1 G1397S; JAK2 L393V; KITD816Y; LNK E208Q;
TET2 Y867H) while the other 5 have not been previously described (ETV6A215P; KIT Y553C;
NFE2 1291T; SH2B3 G382D; SH2B3 L438V). Mutations in myeloid genes were prevalent in
the signalling functional group. A single mutation was found in 7 pts (3MPCM, 3 DCM,
1ISM), while two or more mutations were found in 3 DCM pts, 2 of them had a spontaneous
regression. Two different mutations (KIT Y553C and SH2B3 L438V) were found in two
sisters. Overall, 9/36 pts (5 DCM, 3 MPCM, 1 mastocytoma) presented spontaneous complete
regression of cutaneous lesions after a median time of 25 months (range: 17 months
- 25 years).
Summary/Conclusion: KIT mutations resulted in 35% of the children tested. The RT-PCR
technique resulted more sensitive in finding KIT D816V, while NGS in detecting other
mutations. ASXL1, JAK2 and TET2 mutations, detected in our population, are different
to those reported in adult pts with SM. The presence of multiple mutations in the
DCM form, does not appear to influence the evolution to a systemic form, unlike in
adults.
P1007: PHASE 2, OPEN-LABEL, MULTICENTER, SINGLE-ARM STUDY INVESTIGATING THE EFFICACY
AND SAFETY OF ROPEGINTERFERON ALFA-2B IN JAPANESE PATIENTS WITH POLYCYTHEMIA VERA
K. Kirito1,*, Y. Edahiro2,3,4, K. Ohishi5, A. Gotoh6, K. Takenaka7, H. Shibayama8,
T. Shimizu9, K. Usuki10, K. Shimoda11, M. Ito12, S. A. VanWart13, O. Zagrijtschuk14,
A. Qin15, H. Kawase16, N. Miyachi16, T. Sato16, N. Komatsu2,3,4,16
1Department of Hematology and Oncology, University of Yamanashi, Chuo-shi, Yamanashi;
2Department of Hematology; 3Laboratory for the Development of Therapeutics Against
MPN; 4Department of Advanced Hematology, Juntendo University Graduate School of Medicine,
Bunkyo-ku, Tokyo; 5Department of Transfusion Medicine and Cell Therapy, Mie University
Hospital, Tsu-shi, Mie; 6Department of Hematology, Tokyo Medical University, Shinjuku-ku,
Tokyo; 7Department of Hematology, Clinical Immunology, and Infectious Diseases, Ehime
University Graduate School of Medicine, Toon-shi, Ehime; 8Department of Hematology,
National Hospital Organization Osaka National Hospital, Chuo-ku, Osaka; 9Division
of Hematology, Department of Medicine, Keio University School of Medicine, Shinjuku-ku,
Tokyo; 10Department of Hematology, NTT Medical Center Tokyo, Shinagawa-ku, Tokyo;
11Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine,
Faculty of Medicine, University of Miyazaki, Kiyotake-cho, Miyazaki; 12Department
of Pathology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya-shi,
Aichi, Japan; 13Enhanced Pharmacodynamics, LLC, NY; 14PharmaEssentia Corporation USA,
MA, United States of America; 15PharmaEssentia Corporation, Taipei, Taiwan; 16PharmaEssentia
Japan K.K., Minato-ku, Tokyo, Japan
Background: Polycythemia vera (PV) is a BCR–ABL-negative myeloproliferative neoplasms
with a JAK2 gene mutation, that can be complicated by thrombosis and hemorrhage. It
may also evolve to secondary acute myeloid leukemia, which has a poor prognosis and
is potentially fatal. Ropeginterferon alfa-2b, a peg-proline-interferon alfa-2b with
improved pharmacokinetic properties, allowing for less frequent administration and
better tolerability, has been shown in clinical studies (PEGINVERA, PROUD-PV, CONTINUATION-PV)
to be efficacious and have a favorable safety profile. However, its efficacy and safety
have not been evaluated in Japanese PV patients.
Aims: The aim of this phase 2, single-arm study (NCT04182100) was to investigate the
efficacy and safety of ropeginterferon alfa-2b in Japanese patients with PV to assess
the clinical utility of this cytoreductive therapy as a treatment option.
Methods: Japanese patients aged ≥20 years with a PV diagnosis according to World Health
Organization 2008/2016 criteria and who were unsuitable candidates for current standard
treatment were included, and patients being refractory to hydroxyurea (HU), symptomatic
splenomegaly, or previous treatment with interferon alfa were excluded. Ropeginterferon
alfa-2b was administered subcutaneously every 2 weeks for 12 months (M) at a starting
dose of 100μg (50μg in patients receiving HU). This was increased by 50μg every 2
weeks until patients had achieved complete hematological response (CHR): hematocrit
<45% without phlebotomy in the preceding 3M, platelets ≤400×109/L, leukocytes ≤10×109
/L, or reached the maximum recommended single dose (500μg). The primary endpoint was
phlebotomy-free CHR at 9M and 12M (end of treatment [EOT]). Secondary endpoints were
change in hematologic parameters, CHR in patients with or without prior HU use, JAK2V617F
allele burden, molecular response, and frequency of phlebotomy. Safety endpoints were
incidence of treatment-emergent adverse events (TEAEs) and AEs of special interest.
Results: A total of 29 patients comprised the intent-to-treat population (median age,
54 years; mean hematocrit, 46.85%; mean platelets, 747.3×109/L; mean leukocytes, 17.07×109/L
at baseline). Twenty-seven patients had a JAK2V617F mutation (mean allele burden,
72.19%). Two patients discontinued the study (one withdrew consent, the other due
to an AE [silent thyroiditis]). The primary outcome of durable CHR without phlebotomy
at 9M and EOT was achieved by 8/29 (27.6%) patients. Hematocrit, platelets, and leukocytes
decreased over time (mean±SD change from baseline to EOT: –5.45±6.66%, –493.6±374.9×109/L,
–11.71±8.38×109/L, respectively), and all parameters improved to their target value
range. The proportion of patients with CHR at EOT was similar in patients with or
without prior HU use (Table). JAK2V617F allele burden decreased over time (change
from baseline to EOT: mean ±SD, –19.17±22.64; Median [min, max], -11.62 [-74.80,33.80];
n=26). Molecular response increased over time. Eight (27.6%) patients did not require
phlebotomy. TEAEs occurred in all patients; however, no event of grade ≥ 3 occurred.
The most common TEAEs were alopecia (55.2%), fatigue (27.6%) and influenza-like illness
(27.6%). AEs of special interest, which could be both disease and treatment-related,
occurred in 9 (31.0%) of the patients and were related to the study treatment in 5
(17.2%) of the patients.
Image:
Summary/Conclusion: Ropeginterferon alfa-2b is a safe and efficacious treatment option
in Japanese patients with polycythemia vera.
P1008: ASSOCIATION BETWEEN FRAILTY AND CLINICAL OUTCOMES IN MYELOPROLIFERATIVE NEOPLAMS:
A POPULATION-BASED STUDY FROM ONTARIO, CANADA
A. Bankar1,*, W. Chan2, N. Liu2, M. Cheung3, S. Alibhai4, V. Gupta1
1Medical Oncology and Hematology, Princess Margaret Cancer Center, University Health
Network; 2ICES; 3Sunnybrook Health Sciences Centre; 4General Internal Medicine, Toronto
General Hospital- University Health Network, Toronto, Canada
Background: Frailty independently predicts adverse outcomes in several different cancers
and community-dwelling older adults. Its impact on clinical outcomes in myeloproliferative
neoplasms (MPN) is unknown.
Aims: To measure the association between frailty and all-cause mortality and thrombosis
in MPN patients.
Methods: Method: A retrospective, population-based study using province-wide administrative
databases of Ontario.
Study population: We included MPN patients in the Ontario Cancer Registry from 2004
to 2019 ((Total n= 10,336: ET, n=5,108; PV, n=3,843; MF, n=1,385). Baseline frailty
was measured during two years prior to date of MPN diagnosis using either (i) the
Johns Hopkins Adjusted Clinical Groups® frailty indicator (ACG-F), categorized as
fit or frail if any of the ten frailty-defining diagnoses were present or (ii) the
McIsaac’s cumulative deficit frailty index (mFI), categorized as fit, prefrail, or
frail if mFI <0.10, 0.10-0.19, > 0.19 respectively.
Main outcome measures: Cox proportional hazard model was used to generate the Hazard
Ratios (HRs) with 95% confidence interval (CI) for all-cause mortality comparing frail
vs. prefrail vs. fit patients. Subdistribution hazard ratios (SHR) using the Fine-Gray
models were used to evaluate the effect of frailty on development of thrombosis, taking
death as the competing event. Analyses were adjusted for age, comorbidities, prior
thrombosis, and level of marginalization.
Results: Results: The mean duration of follow-up for ET, PV and MF was 3.8, 4.0 and
2.9 years, respectively. 16% MPN cases were categorized as mFI-frail and 51% as mFI-prefrail.
Patient with MF were more likely to be mFI-frail or mFI-prefrail compared to ET and
PV (34%, 23%, and 20% respectively, p<0.001). In all MPN subtypes, frailty was independently
associated with increased risk of mortality after adjusting for age, sex, and comorbidities.
Compared to fit patients, the HRs for all-cause mortality for prefrail and frail patients
were: 1.6 (1.3-1.9), and 3.6 (2.9-4.4) in ET; 1.3 (1.1-1.5) and 2.7 (2.1-3.4) in PV,
and 1.2 (1.0-1.5) and 2.0 (1.5-2.7) in MF. Other predictors associated with increased
all-cause mortality were advanced age (compared to age <40 years, HRs for 40-65 years,
65-75 years, >75 years: ET: 3.3 (2.0-5.5), 5.34 (3.2-8.7), 12.9 (7.9-21.2); for PV:
3.2 (1.6-6.7), 7.36 (3.6-15.0), 17.3 (8.5-35.2), for MF: 1.8 (0.8-3.8), 3.0 (1.4-6.5),
4.8 (2.2-10.4)) and comorbidities (1 and ≥ 2 comorbidities vs no comorbidities: ET:
1.3 (1.1-1.5), 2.4 (2.0-2.8); for PV: 1.0 (0.8-1.2), 1.2 (1.0-1.5); for MF: 1.2 (1.0-1.5),
1.4 (1.1-1.8)). With ACG-F, frailty was noted in 11% in MPN cases and was associated
with increased mortality after adjusting for comorbidities and advanced age in all
MFN subtypes [HRs in ET 2.5 (2.2-2.8), PV: 2.1 (1.8-2.4), and MF: 1.8 (1.5-2.2)].
In multivariable Fine-Gray regression, neither mFI-prefrail nor mFI-frail patients
had an increased risk of thrombosis in all MPN subtypes. Compared to fit patients,
frail patients using ACG-F had lower SHRs of thrombosis (ET: 0.6 (0.4-0.8), PV: 0.6
(0.4-0.8), MF: 0.2 (0.05-0.8), but higher SHRs for the competing event of death (ET:
3.0 (2.6-3.5), PV: 2.3 (1.9-2.9), MF: 1.9 (1.5-2.4)).
Image:
Summary/Conclusion: Conclusions: Using large population-based databases, we found
that a significant proportion of MPN patients are frail or prefrail at diagnosis despite
younger age or less comorbidity burden. After adjusting for confounding from advanced
age and increasing co-morbidity burden, frailty independently predicts increased risk
of all-cause mortality in ET, PV, and MF.
P1009: PREDICTORS OF HOSPITALIZATION AND SEVERE OUTCOMES IN PATIENTS WITH MPN AND
COVID-19
T. Barbui1,*, A. Carobbio1, A. Masciulli1, A. Iurlo2, M. A. Sobas3, E. M. Elli4, E.
Rumi5, V. De Stefano6, F. Lunghi7, M. Marchetti8, R. Daffini9, M. Gasior Kabat10,
B. Cuevas11, M. L. Fox12, M. M. Andrade Campos13, F. Palandri14, P. Guglielmelli15,
G. Benevolo16, C. Harrison17, M. A. Foncillas18, M. Bonifacio19, A. Alvarez-Larran20,
J.-J. Kiladjian21, E. Bolanos Calderon22, A. Patriarca23, K. S. Quiroz Cervantes24,
M. Griesshammer25, V. Garcia-Gutierrez26, A. Marin Sanchez27, E. Magro Mazo28, M.
Ruggeri29, J. C. Hernandez-Boluda30, S. Osorio31, G. Carreno-Tarragona32, M. Sagues
Serrano33, R. Kusec34, B. Navas Elorza35, A. Angona36, B. Xicoy Cirici37, E. Lopez
Abadia38, S. Koschmieder39, E. Cichocka40, A. Kulikowska de Nałęcz41, M. Bellini42,
D. Cattaneo2,43, C. Bucelli2, F. Cavalca4, O. Borsani5, S. Betti6, N. Curto-Garcia17,
S. Carbonell20, L. Benajiba21, A. Rambaldi42,43, A. M. Vannucchi15
1FROM Research Foundation, Bergamo; 2Hematology division, Foundation IRCCS Ca’ Granda
Ospedale Maggiore Policlinico, Milan, Italy; 3Department of Hematology, Blood neoplasms
and Bone marrow transplantation, Wroclaw Medical University, Wroclaw, Poland; 4Hematology
division and Bone marrow transplant unit, San Gerardo Hospital, ASST Monza, Monza;
5Department of Molecular medicine, University of Pavia, Pavia; 6Fondazione Policlinico
“A. Gemelli” IRCCS, Rome; 7IRCCS Ospedale San Raffaele, Milan; 8AOU SS. Antonio e
Biagio e C. Arrigo, Alessandria; 9ASST Spedali Civili, Brescia, Italy; 10Hospital
Univeristario La Paz, Madrid; 11Hospital Universitario de Burgos, Burgos; 12Department
of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Hospital
Universitari, Vall d’Hebron Barcelona Hospital Campus; 13Hospital del Mar, Barcelona,
Spain; 14IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna; 15Center Research
and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental
and Clinical Medicine, Azienda Ospedaliera Universitaria Careggi, University of Florence,
Florence; 16AOU Città della Salute e della Scienza di Torino, Torino, Italy; 17Guy’s
and St. Thomas’ NHS Foundation Trust, London, United Kingdom; 18Hospital Universitario
Infanta Leonor, Madrid, Spain; 19Ospedale Policlinico “G.B. Rossi”, Borgo Roma, Verona,
Italy; 20Hospital Clinic de Barcelona, Barcelona, Spain; 21Hospital Saint-Louis, Paris,
France; 22Hospital Clinico San Carlos, Madrid, Spain; 23AOU Maggiore della Carità,
Novara, Italy; 24Hospital Universitario de Mostoles, Madrid, Spain; 25University Clinic
for Hematology, Oncology, Hemostaseology and Palliative Care, Johannes Wesling Medical
Center, Minden, Germany; 26Hospital Ramon y Cajal, IRYCIS, Madrid; 27Hospital General
Universitario de Albacete, Albacete; 28Hospital Universitario Principe de Asturias,
Alcalà de Henares, Madrid, Spain; 29Ospedale San Bortolo, Vicenza, Italy; 30Hospital
Clinico Universitario, INCLIVA, Valencia; 31Hospital Gregorio Maranon; 32Hospital
Universitario 12 de Octubre, Madrid; 33ICO L’Hospitalet-Hospital Moises Broggi, Sant
Joan Despì, Barcelona, Spain; 34Department of haematology, Clinic of internal medicine,
University Hospital Dubrava-School of Medicine University of Zagreb, Zagreb, Croatia;
35Hospital Moncloa, Madrid; 36ICO Girona Hospital Josep Trueta, Girona; 37ICO Hospital
Germans Trias i Pujol, Badalona (Barcelona); 38Hospital General de Elche, Elche (Alicante),
Spain; 39Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation,
Faculty of Medicine, RWTH Aachen University, Aachen, Germany; 40Department of Hematology
and Bone Marrow Transplantation, Nicolaus Copernicus Hospital, Torun; 41Department
of Hematology and Internal Diseases, State Hospital, Opole, Poland; 42ASST Papa Giovanni
XXIII, Bergamo; 43Università degli Studi di Milano, Milano, Italy
Background: Risk factors for severe COVID-19 in myeloproliferative neoplasms (MPN)
have been extensively explored. However, no information is available on risk factors
to hospitalization at COVID-19 diagnosis.
Aims: To provide an evidence-based triage and inform for a timely and antiviral therapy
prescription in early phases of viral replication.
Methods: The MPN-COVID study is still enrolling consecutive adult MPN patients with
COVID-19 infection since February 15, 2020. Among 479 patients (ET n=175, PV n=158,
MF n=91, and pre-PMF n=55) with COVID-19 from Feb 2020 to Jun 2021, 248 (52%) were
managed at home and 231 (48%) were hospitalized.
Results: Univariate analysis
Compared to outpatients, those admitted to hospital were more likely to be men (58.9%
vs. 45.2%, p=0.003), older than 70 years (61.3% vs. 29.0%, p<.001), with at least
one comorbidity (79.7% vs. 55.5%, p<.001) and a history of thrombosis (26.5% vs. 16.6%,
p=0.008). Overt myelofibrosis (MF) cases were more frequent in hospital (38.5% vs.
18.1%, p<.001) than PV, ET or pre-PMF. Ruxolitinib was more frequently used in patients
who underwent to hospital than managed at home. (25.7% vs. 12.1%, p<.001). In comparison
with outpatients, hospitalized cases had a significantly lower median values of hemoglobin
(12.1 vs. 13.3 g/dL, p<.001), platelets (250 vs. 390 x109/L, p<.001), absolute lymphocytes
(0.8 vs. 1.4 x109/L, p<.001) and higher neutrophil counts (5.1 vs. 4.5 x109/L, p=0.022),
leading to a significant increase of neutrophil to lymphocyte ratio (NLR) (6.6 vs.
3.2, p<.001). Compared to lymphocytopenia, the best sensitivity and specificity was
found for NLR, whose AUC was 77.28% by ROC analysis.
Multivariate analysis
By adjusting for sex, comorbidity, fever, systemic symptoms, O2 saturation, previous
thrombosis, MPN type, ruxolitinib exposure, and first vs. subsequent waves and vaccination
period, three factors emerged as independent predictors of hospitalization: age over
70 years, (OR=3.02, p=0.038), dyspnea (OR=7.23, p<.001) and NLR ≥4, (OR=6.94, p<.001).
Interaction model of risk factors
In a model fitted to test the interaction terms of the three significant variables,
we evaluated the marginal effect of NLR and dyspnea across different age classes (Figure)
and found that in younger patients (i.e., from 50 to 70 years) dyspnea was the stronger
predictor than increased NLR; conversely, both dyspnea and NLR showed a high and comparable
marginal effect in age >80 years. Remarkably, the probability of hospitalization consistently
exceeded 90% for any age group when dyspnea and NLR were concomitantly present, and
their combination was more prevalent in MF (42%) than in the other phenotypes (24%,
25% and 29% in pre-PMF, PV and ET patients, respectively).
In addition to predict hospitalization, dyspnea and NLR≥4 were also associated with
severity of COVID-19 illness in terms of respiratory support at hospitalization (OR=2.44,
p=0.023). Of note, only age >70 years and NLR higher than 6 were predictors of survival
in hospitalized patients (OR=3.24, p=0.007 and OR=5.41, p=0.041, respectively).
Image:
Summary/Conclusion: For triage purposes of MPN patients tested positive for COVID-19,
dyspnea, age and NLR are powerful predictors of hospitalization and identify patients
at higher probability of invasive or non-invasive respiratory support and survival,
particularly in overt MF. This MPN subgroup at high risk for progression to severe
COVID-19 disease should be prioritized for antiviral therapy, even in the ambulatory
setting.
P1010: RUXOLITINIB IN MYELODEPLETIVE MYELOFIBROSIS: RESPONSE, TOXICITY, AND OUTCOME
F. Palandri1,*, D. Bartoletti1,2, M. Breccia3, G. Auteri1,2, E. M. Elli4, M. M. Trawinska5,
N. Polverelli6, M. Tiribelli7, G. Benevolo8, A. Iurlo9, A. Tieghi10, F. H. Heidel11,
G. Caocci12, E. Beggiato13, G. Binotto14, F. Cavazzini15, M. Miglino16,17, C. Bosi18,
M. Crugnola19, M. Bocchia20, B. Martino21, N. Pugliese22, A. D. Romagnoli1,2, C. Mazzoni1,2,
L. Scaffidi23, A. Isidori24, D. Cattaneo9, M. Krampera23, F. Pane22, D. Cilloni25,26,
G. Semenzato14, R. M. Lemoli16,17, A. Cuneo15, E. Abruzzese5, N. Vianelli1, M. Cavo1,2,
M. Bonifacio23, G. A. Palumbo27
1IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”;
2Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di
Bologna, Bologna; 3A.O.U. Policlinico Umberto I, Università degli Studi di Roma “La
Sapienza”, Rome; 4Ospedale San Gerardo, ASST Monza, Monza; 5Ospedale S. Eugenio, Università
Tor Vergata, Rome; 6ASST Spedali Civili di Brescia, Brescia; 7A.O.U. Integrata di
Udine, Udine; 8A.O.U. Città della Salute e della Scienza, Torino; 9Foundation IRCCS
Ca’ Granda, Ospedale Maggiore Policlinico, Milano; 10Azienda USL - IRCCS di Reggio
Emilia, Reggio Emilia, Italy; 11Innere Medicine C, Universitätsmedizin Greifswald,
Greifswald, Germany; 12Polo oncologico “A. Businco”, Università degli studi di Cagliari,
Cagliari; 13Dipartimento di Oncologia, Università di Torino, Torino; 14A.O.U. di Padova,
Padova; 15A.O.U. Arcispedale S. Anna, Ferrara; 16IRCCS Policlinico San Martino; 17Dipartimento
di Medicina interna e Specialità mediche, Università di Genova, Genova; 18AUSL di
Piacenza, Piacenza; 19Azienda Ospedaliero-Universitaria di Parma, Parma; 20Policlinico
S. Maria alle Scotte, AOU Senese, Siena; 21Division of Hematology, Azienda Ospedaliera
‘Bianchi Melacrino Morelli’, Reggio Calabria; 22Dipartimento di Medicina clinica e
Chirurgia, Università degli Studi di Napoli Federico II, Napoli; 23A.O.U. Integrata
Verona, Verona; 24A.O. Ospedali Riuniti Marche Nord (AORMN), A.O. San Salvatore, Pesaro;
25A.O. Ordine Mauriziano di Torino; 26AOU San Luigi Gonzaga, Torino; 27Dipartimento
di Scienze Mediche, Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia”, Università
di Catania, Catania, Italy
Background: Around 30% of Myelofibrosis (MF) either primary (PMF) or secondary to
polycythemia vera/essential thrombocythemia (SMF) may present a myelodepletive phenotype
(MyD) (ie, thrombocytopenia, leukopenia, anemia). Pts with MyD MF represent a challenging
population, as prognosis is poorer compared to pts with myeloproliferative (MyP) MF
and therapeutic options, including the JAK1/2 inhibitor ruxolitinib (RUX), are limited
or must be given at reduced doses.
Aims: In light of the upcoming new drugs that may be used in MyD MF, we explored prognostic
correlates of MyD phenotype in RUX-treated MF pts.
Methods: After IRB approval, the “RUX-MF” retrospective real-world study collected
801 chronic phase MF pts treated with RUX in 26 Hematology Centers. MyD was defined
as: WBC <4×109/L and/or Hb <11/<10 g/dL (males/females) and/or PLT <100×109/L with
no increase of other blood cells (WBC >15×109/L, Hb >16.5/>16 g/dL in males/females,
Plt >450×109/L). 219 (27.3%) had a MyD MF, including 140 (17.5%) PMF and 79 (9.8%)
SMF. Spleen and symptoms response (SR/SyR) were defined according to IWG-MRT criteria.
NGS mutational analysis was available for 167 pts.
Results: In multivariable analysis (MVA), PMF diagnosis (p=0.001) and unfavorable
karyotype (p=0.01) confirmed their significant association with MyD. In PMF pts, MyD
was due to leukopenia, anemia and thrombocytopenia in 7.1%, 52.2% and 9.3%, respectively;
in SMF, corresponding figures were 5%, 51.9% and 11.4%. Two or more cytopenias were
found in 31.4% and 31.7% of PMF and SMF patients, respectively.
In MVA, lower peripheral blast count (p=0.03), higher TSS (p=0.04) and BM fibrosis
grade ≥2 (p=0.03) confirmed their association with MyD in PMF pts. In univariate/MVA,
MyD SMF patients were more likely to have higher peripheral blast count (p=0.003/p=0.04),
a higher MYSEC-PM risk (p<0.001/p=0.001), and to be triple negative (p=0.005/0.03).
RUX starting, median at 3 months, and median overall dose was more frequently ≤10 mg
BID in MyD than in MyP pts (44.9% vs 67.5%, p<0.001; 39.5% vs 59.9%, p<0.001; 34.9%
vs 57.3%, p<0.001, respectively). This was confirmed also in PMF and SMF separately.
The rate of SR was comparable in MyD and MyP patients. However, SR at 3 and 6 months
was lower in pts with PLT<100 x 109/l (p=0.02). In SMF, MyD pts had lower rates of
SR (10.8% vs 28.0% at 3 mos, p=0.004; 20.0% vs 33.1% at 6 mos, p=0.05).
SyR was significantly lower in MyD MF (51.9% vs 62.5% in MyP at 3 mos, p=0.01; 59.8%
vs 71.0% at 6 mos, p=0.008). In particular, anemia and thrombocytopenia were significantly
associated with lower SyR.
After a median RUX exposure of 2.3 yrs (0.1-12.6), 364 (45.4%) pts stopped RUX, 110
(13.7%) had a blast phase and 366 (45.7%) died. After competing risk analysis, the
cumulative incidence of RUX discontinuation was higher in MyD MF patients overall
(p<0.001), only PMF (p=0.03) and only SMF (p<0.001) (Fig.1a). Incidence of RUX stop
was significantly higher in MyD patients with ≥2 cytopenias (p=0.03).
Leukemia-free survival was not influenced by MyD/MyP phenotype (Fig.1b).
In Cox regression analysis adjusted for DIPSS score, OS was significantly shorter
in MyD vs MyP MF (median, 4.5 vs 5.7 yrs; p=0.03) (Fig.1c). This was confirmed considering
only SMF patients (p=0.02).
Image:
Summary/Conclusion: MyD phenotype is associated with baseline high-risk clinical and
molecular features, with lower responses to RUX, particularly in case of low PLT count,
and higher risk of drug discontinuation and death. Newer strategies are warranted
in this setting.
P1011: PREDICTORS OF COVID-19 DISEASE AND SURVIVAL TO COVID-19 IN MPN PATIENTS TREATED
WITH RUXOLITINIB
F. Palandri1,*, D. Bartoletti1,2, E. M. Elli3, G. Auteri1,2, M. Bonifacio4, G. Benevolo5,
F. Heidel6, M. M. Trawinska7, E. Rossi8,9, C. Bosi10, A. Tieghi11, M. Tiribelli12,
A. Iurlo13, N. Polverelli14, G. Caocci15, G. Binotto16, F. Cavazzini17, E. Beggiato18,
D. Cilloni19, C. Tatarelli20, F. Mendicino21, M. Miglino22,23, M. Bocchia24, M. Crugnola25,
C. Mazzoni1,2, A. D. Romagnoli1,2, G. Rindone3, S. Ceglie8, A. D’Addio26, E. Santoni4,
D. Cattaneo13, R. M. Lemoli22,23, M. Krampera4, A. Cuneo17, G. Semenzato27, R. Latagliata28,
E. Abruzzese7, N. Vianelli1, M. Cavo1,2, A. Andriani29, V. De Stefano8,9, G. Palumbo30,
M. Breccia31
1IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”;
2Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di
Bologna, Bologna; 3Ospedale San Gerardo, ASST Monza, Monza; 4A.O.U. Integrata Verona,
Verona; 5A.O.U. Città della Salute e della Scienza, Torino, Italy; 6Innere Medicine
C, Universitätsmedizin Greifswald, Greifswald, Germany; 7Ospedale S. Eugenio, Università
Tor Vergata; 8Section of Hematology, Department of Radiological and Hematological
Sciences, Catholic University School of Medicine; 9Fondazione Policlinico Universitario
A. Gemelli IRCCS, Rome; 10AUSL di Piacenza, Piacenza; 11Azienda USL - IRCCS di Reggio
Emilia, Reggio Emilia; 12A.O.U. Integrata di Udine, Udine; 13Foundation IRCCS Ca’
Granda, Ospedale Maggiore Policlinico, Milano; 14ASST Spedali Civili di Brescia, Brescia;
15Polo oncologico “A. Businco”, Università degli studi di Cagliari, Cagliari; 16A.O.U.
di Padova, Padova; 17A.O.U. Arcispedale S. Anna, Ferrara; 18Dipartimento di Oncologia,
Università di Torino, Torino; 19A.O. Ordine Mauriziano di Torino, Torino; 20Azienda
Ospedaliera Universitaria Sant’Andrea di Roma, Rome; 21Unit of Hematology, Hospital
of Cosenza, Cosenza; 22IRCCS Policlinico San Martino; 23Dipartimento di Medicina interna
e Specialità mediche, Università di Genova, Genova; 24Policlinico S. Maria alle Scotte,
AOU Senese, Siena; 25Azienda Ospedaliero-Universitaria di Parma, Parma; 26Division
of Hematology, Onco-hematologic Department, AUSL della Romagna, Ravenna; 27Unit of
Hematology and Clinical Immunology, University of Padova, Padova; 28Hematology Unit,
Ospedale Belcolle, Viterbo; 29Hematology, Fabrizio Spaziani Hospital, Frosinone; 30Department
of Scienze Mediche, Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia”, University
of Catania, Catania; 31Division of Cellular Biotechnologies and Hematology, University
Sapienza, Rome, Italy
Background: Ruxolitinib (RUX) use and discontinuation are risk factors for severe
COVID-19 and death in MPN patients (pts). In pts on RUX therapy, predictors for COVID-19
disease and survival (OS) to COVID-19 are unknown.
Aims: The aims of this study were to distinguish RUX-treated pts at higher risk of
COVID-19 and to assess prognostic factors for OS.
Methods: We performed a sub-analysis of the RUX-MF and the PV-ARC observational studies
that include consecutive adult pts with myelofibrosis (MF) and polycythemia vera (PV),
respectively. Overall, 815 MF and 172 PV pts treated with RUX outside clinical trials
have been registered. At pandemic start, 494 pts (359 MF and 135 PV) on RUX were included
in this analysis.
Results: Among 66 (13.6%) pts (PV n=11, MF n=55) with COVID-19 from Feb 2020 to Jan
2022, 1 (1.5%), 14 (21.2%), 9 (13.6%), 17 (25.8%), 4 (6.1%) and 21 (31.8%) pts had
an asymptomatic, mild, moderate, severe, critical, and fatal infection, respectively;
42 (63.7%) were hospitalized.
Overall, 14, 38 and 14 infections were observed during the 1st (Feb-Jun 2020), 2nd
(Jul 2020-Jun2021) and 3rd (Jul 2021-Jan 2022) wave of the pandemic, with an overall
incidence rate of 10.2 per 100 pt-yrs. Incidence rates in the 3 waves were 8, 10.2
and 7 per 100 pt-yrs respectively. Hospitalized cases were significantly less frequent
during the 3rd wave (35.7% vs 64.3%/73.7% in the 1st/2nd wave, p=0.04).
Overall, 283/390 evaluable pts (72.6%) received ≥1 dose of Comirnaty vaccine (19/66
COVID-19 pts; 5, 7 and 7 pts had received 1, 2 or 3 vaccine doses, respectively).
At COVID-19 diagnosis, RUX was reduced in 10 (15.1%) pts and discontinued in 9 (13.6%)
pts, comparably in MF and PV.
In the total cohort, COVID-19 infection was more frequent in pts with MF (15.3% vs.
8.2% PV pts, p=0.04), with ≥1 comorbidity (15% vs. 8.7%, p=0.04). Also, COVID-19 infections
after vaccine availability were more frequent in unvaccinated pts (37.4 vs. 6.3%,
p<0.001).
COVID-19 requiring hospitalization was more frequently observed in pts ≥70 yrs (12.2%
vs. 6.8% in pts <70 yrs, p=0.04), and without COVID-19 vaccine (32.4% vs. 2.9%, p<0.001).
No additional predictors for COVID-19 were noted analyzing MF and PV separately.
In COVID-19 pts, hospitalized cases had a significantly lower median platelet count
(275 vs. 168 x109/L, p=0.02), were receiving lower RUX doses (33.3% <10 mg BID vs.
8.3%, p=0.02) and more frequently presented comorbidities (40.5% vs. 13.6%, p=0.03)
compared to outpatients. MF vs. PV, median hemoglobin levels, age≥70 yrs and sex were
not associated with hospitalization. MF pts who were not in spleen response at COVID-19
infection had higher risk of hospitalization (73% vs. 44.4% in responders, p=0.04).
After multivariable Cox analysis including previous anti-SARS-Cov-2 vaccine, need
for hospitalization, age≥70 and male sex, OS to COVID-19 was significantly improved
in pts who had previously received anti-SARS-Cov-2 vaccine (HR=0.10, p=0.02) (Fig.1),
in pts with COVID-19 not requiring hospitalization (HR=0.19, p=0.03) and in patients
<70 yrs (HR=0.38, p=0.03). The COVID-19 wave did not impact OS (p=0.53).
Image:
Summary/Conclusion: Among RUX-treated pts, lower RUX doses, comorbidities and no spleen
response are significant predictors of hospitalization. Vaccine was the most protective
factor against COVID-19 disease, hospitalization, and mortality. RUX-treated pts,
regardless of MPN type, should be sensitized to adherence to the vaccine program and
prioritized for antiviral therapy in case of infection.
P1012: A NUMBER OF CONGENITAL ERYTHROCYTOSIS ARE MULTIGENIC
A. Benetti1,*, G. Biagetti1, E. Cosi1, I. Bertozzi1, G. Ceolotto1, M. L. Randi1
1Department of Medicine, University of Padua, Padua, Italy
Background: Imbalance in erythropoiesis and oxygen homeostasis could lead to the onset
of Erythrocytosis, a clinical condition characterized by persistently raised hemoglobin
(Hb) and hematocrit (Ht) levels.
Nowadays, a high number of patients are classified as “Idiopathic Erythrocytosis”
(IE) meaning that it is not possible to identify the cause of hematocrit increase.
Recently, HFE mutations has been found to be often present in idiopathic erythrocytic
patients.
The HFE mutations could facilitate the increase in red cell mass, and this indicates
the possible involvement of Iron Metabolism in causes of IE.
Aims: To identify the possible mutations of IE patients using our gene panel for Next
Generation Sequencing (NGS).
Methods: In 118 patients with IE, regularly followed in our surgery, we used a gene
panel for NGS composed by fifteen genes: JAK2, EGLN1 (PHD2), EPO-R, FTL, FTH, ASXL1,
HFE, HFE2, TFR2, HAMP, SLC40A1, SLC11A2, VHL, BPMG, EPAS1 (HIF-2α). We analyzed all
the exons parts of these genes. The approach used for the library preparation was
a multiplexing PCR and data were analyzed by bioinformatics tools. In all patients,
the mutations found were confirmed with Sanger Sequencing.
Results: In 78 out of the 118 patients (66%) evaluated, we found one 1 mutated gene
in 55 patients (7 EGLN1, 1 VHL, 34 HFE, 6 TFR2, 3 JAK2 and 4 EPO-R), 2 mutated genes
in 18 patients (1 EGLN1/JAK2, 4 EGLN1/HFE, 2 EPAS1/HFE, 1 EPAS1/JAK2, 1 EPAS1/EGLN1,
2 EPO-R/HFE, 2 HFE/JAK2, 1 JAK2/TFR2, 2 with TFR2/EGLN1, 1 VHL/HFE, 1 TFR2/HFE) and
3 mutated genes in 5 patients (1 EGLN1/EPO-R/HFE, 1 VHL/HFE/EGLN1, 1 EPAS1/EPO-R/HFE,
1 EPAS1/HFE/EGLN1 and HFE/TFR2/JAK2). All the mutations found were germline. Interestingly,
21 out of the 27 (78%) patients carrying HFE mutation displayed 2 or more mutations.
Summary/Conclusion: The use of NGS panels in various clinical conditions has frequently
demonstrated that multiple mutations coexist in various patients and that the monogenic
diseases are quite rare. This study is the first at our best knowledge that gives
evidence of more than one mutated gene in erythrocytotic patients, and this is the
results of the use of an appropriate NGS panel.
The data here reported suggest that we can confirm that HFE mutations are common in
IE patients, in some cases associated with other mutations, but the present data ask
some questions: HFE mutations alone can induce erythrocytosis? or other genetic imbalances
are needed? The presence of multiple mutations could lead to a greater severity of
the disease? A significant number of IE patients displayed TFR2 mutations, giving
more relevance to the hypothesis of the iron metabolism implication in IE genesis.
Finally, the presence of 11 patients carrying germlines JAK2 mutations is surprising,
and we suppose it will be necessary to strictly watch these patients in the suspicion
of a future appearance of a polycythemia vera.
The present study demonstrates the need to extend the biomolecular knowledge in patients
with idiopathic erythrocytosis.
P1013: OVERALL SURVIVAL IN PATIENTS WITH SYSTEMIC MASTOCYTOSIS WITH ASSOCIATED HEMATOLOGIC
NEOPLASM TREATED WITH AVAPRITINIB VERSUS BEST AVAILABLE THERAPY
A. Reiter1,*, J. Gotlib2, I. Álvarez-Twose3, D. H. Radia4, J. Luebke1, P. J. Bobbili5,
A. Wang5, C. Norregaard6, S. Dimitrijević7, E. Sullivan6, M. Louie-Gao6, J. Schwaab1,
I. A. Galinsky8, C. Perkins2, W. R. Sperr9, P. Sriskandarajah4, A. Chin5, S. R. Sendhil5,
M. S. Duh5, P. Valent9, D. J. DeAngelo8
1Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany;
2Stanford Cancer Institute/Stanford University School of Medicine, Stanford, United
States of America; 3Institute of Mastocytosis Studies of Castilla La Mancha (CLMast)
─ Spanish Reference Center (CSUR) for Mastocytosis and CIBERONC, Virgen del Valle
Hospital, Toledo, Spain; 4Guy’s & St Thomas’ NHS Foundation Trust, Guy’s Hospital,
London, United Kingdom; 5Analysis Group, Inc., Boston; 6Blueprint Medicines Corporation,
Cambridge, United States of America; 7Blueprint Medicines Corporation, Zug, Switzerland;
8Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, United States
of America; 9Department of Internal Medicine I, Division of Hematology and Hemostaseology,
and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of
Vienna, Vienna, Austria
Background: Avapritinib, a selective inhibitor of KIT D816V, was approved in the United
States (US) for treatment of adults with advanced systemic mastocytosis (AdvSM), a
rare myeloid neoplasm with poor overall survival (OS), based on data from the Phase
1 EXPLORER (NCT02561988) and Phase 2 PATHFINDER (NCT03580655) single-arm trials. Systemic
mastocytosis with an associated hematologic neoplasm (SM-AHN) is both the most common
and most heterogenous subtype of AdvSM. No randomized control trial (RCT) has been
conducted comparing efficacy of avapritinib with best available therapy (BAT) in SM-AHN.
Aims: This study (NCT04695431) compared OS between SM-AHN patients treated with avapritinib
in the EXPLORER and PATHFINDER trials versus SM-AHN patients treated with BAT in standard
clinical practice.
Methods: A multi-center, global, observational, retrospective chart review study was
conducted at 6 study sites (4 European, 2 US) to identify and collect data from AdvSM
patients who received BAT. SM-AHN patients were identified using inclusion/exclusion
criteria similar to the EXPLORER and PATHFINDER trials. Patients receiving BAT could
contribute data on multiple lines of therapy (LOTs) to the analysis; these data were
compared with patient-level data from the EXPLORER and PATHFINDER trials. OS was defined
as the time from avapritinib or BAT initiation to death from any cause; patients were
censored at date of last follow-up if alive. Kaplan-Meier analysis was used to assess
OS. Inverse probability of treatment weighting (IPTW) was used to adjust for differences
in key variables between the treatment cohorts, including age, sex, region, anemia,
thrombocytopenia, leukocyte count, skin involvement, number and type of prior treatments,
performance status, serum tryptase level, and presence of SRSF2/ASXL1/RUNX1 mutations.
An IPTW-weighted Cox proportional hazards model, adjusted for variables that remained
unbalanced after weighting, was used to compare OS between treatment groups.
Results: This analysis included 119 avapritinib patients and 83 BAT patients with
SM-AHN (the latter contributed data on 121 LOTs). Median (range) age of the avapritinib
and BAT cohorts was 70 (45−88) and 71 (38−88) years, and 61% vs. 76% of patients were
male, respectively. Prior systemic therapy was received by 58% of avapritinib and
44% of BAT patients. In the avapritinib cohort, 76% of patients from both trials were
dosed at ≤200mg and 24% of patients, all from EXPLORER, were dosed at ≥300mg. BAT
patients were most frequently treated with tyrosine kinase inhibitors (73 out of 121
LOTs, 60%) or cytoreductive therapies (46 out of 121 LOTs, 38%). Mean durations of
follow-up were 17.6 and 18.1 months, with 29 (24%) and 56 (68%) deaths in avapritinib
and BAT cohorts, respectively. Median OS for avapritinib was 46.9 months (95% CI:
44.9, not estimable) and 18.0 months (95% CI: 13.0, 26.8) for BAT (Figure 1). IPTW-adjusted
median OS was 46.9 months (95% CI: 21.4, 49.0) for avapritinib and 19.5 months (95%
CI: 13.0, 32.2) for BAT. Weighted Cox analysis showed that OS was significantly improved
for avapritinib versus BAT (hazard ratio [95% CI]: 0.42 [0.24, 0.74]; P<0.001), even
with further adjustment for unbalanced variables.
Image:
Summary/Conclusion: The results of this analysis indicate that patients with SM-AHN
treated with avapritinib in clinical trials had significantly longer OS compared to
patients treated with BAT. With no RCTs, these data offer crucial insights into the
survival benefit of SM-AHN patients treated with avapritinib compared to other treatments
for AdvSM.
P1014: OVERALL SURVIVAL IN PATIENTS WITH ADVANCED SYSTEMIC MASTOCYTOSIS RECEIVING
AVAPRITINIB VERSUS MIDOSTAURIN OR CLADRIBINE
A. Reiter1,*, J. Gotlib2, I. Álvarez-Twose3, D. H. Radia4, J. Luebke1, P. J. Bobbili5,
A. Wang5, C. Norregaard6, S. Dimitrijević7, E. Sullivan6, M. Louie-Gao6, J. Schwaab1,
I. A. Galinsky8, C. Perkins2, W. R. Sperr9, P. Sriskandarajah4, A. Chin5, S. R. Sendhil5,
M. S. Duh5, P. Valent9, D. J. DeAngelo8
1Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany;
2Stanford Cancer Institute/Stanford University School of Medicine, Stanford, United
States of America; 3Institute of Mastocytosis Studies of Castilla La Mancha (CLMast)
─ Spanish Reference Center (CSUR) for Mastocytosis and CIBERONC, Virgen del Valle
Hospital, Toledo, Spain; 4Guy’s & St Thomas’ NHS Foundation Trust, Guy’s Hospital,
London, United Kingdom; 5Analysis Group, Inc., Boston; 6Blueprint Medicines Corporation,
Cambridge, United States of America; 7Blueprint Medicines Corporation, Zug, Switzerland;
8Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, United States
of America; 9Department of Internal Medicine I, Division of Hematology and Hemostaseology,
and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of
Vienna, Vienna, Austria
Background: Avapritinib, a selective KIT D816V inhibitor, was approved for the treatment
of adults with advanced systemic mastocytosis (AdvSM) in the United States (US), based
on two single-arm trials: Phase 1 EXPLORER (NCT02561988) and Phase 2 PATHFINDER (NCT03580655).
No randomized controlled trial (RCT) has been conducted comparing efficacy of avapritinib
versus alternative therapies for AdvSM.
Aims: This study (NCT04695431) compared overall survival (OS) between AdvSM patients
treated with avapritinib in the EXPLORER and PATHFINDER trials and those treated with
midostaurin or cladribine in real-world clinical practice.
Methods: Pooled data from the EXPLORER and PATHFINDER trials were compared with data
for patients with AdvSM obtained through a multi-center, global, observational, retrospective
chart review study conducted at 6 study sites (4 European, 2 US). Patients treated
with midostaurin or cladribine were identified using inclusion/exclusion criteria
similar to the trials and could contribute multiple lines of therapy (LOTs) to the
analysis. OS, assessed using Kaplan-Meier analysis, was defined as time from initiation
of avapritinib, midostaurin, or cladribine to death from any cause; patients were
censored at date of last follow-up if alive. Inverse probability of treatment weighting
(IPTW) was used to adjust for differences in key variables between treatment cohorts,
including age, sex, AdvSM subtype, anemia, thrombocytopenia, leukocyte count, skin
involvement, number and type of prior treatments, performance status, serum tryptase
level, and presence of SRSF2/ASXL1/RUNX1 mutations. IPTW-weighted Cox proportional
hazards models, adjusted for variables that remained unbalanced after weighting, were
used to compare OS between cohorts.
Results: This analysis included 176 patients treated with avapritinib, 94 treated
with midostaurin (LOT, n=99), and 44 treated with cladribine (LOT, n=49). Median (range)
age was 68 (31−88) for avapritinib, 69 (26−87) for midostaurin, and 66 (45−88) years
for cladribine patients. 103 (59%) avapritinib patients were male, versus 64 (68%)
in the midostaurin and 27 (61%) in the cladribine cohort. In the avapritinib cohort,
136 (77%) patients from both trials were dosed at ≤200mg and 40 (23%) patients, all
from EXPLORER, were dosed at ≥300mg.
In unweighted analysis of avapritinib versus midostaurin, mean follow-up durations
were 17.9 and 27.9 months, respectively, during which 34 (19%) avapritinib patients
and 56 (60%) midostaurin patients died. Median OS was not reached (NR) (95% CI: 46.9,
not estimable) in the avapritinib cohort and was 28.6 months (95% CI: 18.2, 44.6)
in the midostaurin cohort (Figure 1). In weighted Cox analysis, OS was significantly
improved in the avapritinib versus midostaurin cohort (hazard ratio [HR] [95% CI]:
0.59 [0.36, 0.97]; P<0.001).
The mean-follow-up duration for the cladribine cohort was 24.2 months, during which
29 (66%) patients died. The median OS in the cladribine cohort was 23.4 months (95%
CI: 14.8, 40.6). Weighted Cox analysis showed that OS was significantly improved in
the avapritinib versus cladribine cohort (HR [95% CI]: 0.32 [0.15, 0.67]; P=0.003).
Image:
Summary/Conclusion: The results from this study indicate that AdvSM patients treated
with avapritinib in clinical trials experienced significantly improved survival compared
with patients treated with midostaurin or cladribine in the real world. Given the
lack of RCTs, these data offer essential insights into the improved survival of patients
treated with avapritinib compared to alternative therapies for AdvSM.
P1015: DURATION OF TREATMENT AND REDUCTION IN SERUM TRYPTASE LEVELS IN PATIENTS WITH
ADVANCED SYSTEMIC MASTOCYTOSIS TREATED WITH AVAPRITINIB VERSUS BEST AVAILABLE THERAPY
A. Reiter1,*, J. Gotlib2, I. Álvarez-Twose3, D. H. Radia4, J. Luebke1, P. J. Bobbili5,
A. Wang5, C. Norregaard6, S. Dimitrijević7, E. Sullivan6, M. Louie-Gao6, J. Schwaab1,
I. A. Galinsky8, C. Perkins2, W. R. Sperr9, P. Sriskandarajah4, A. Chin5, S. R. Sendhil5,
M. S. Duh5, P. Valent9, D. J. DeAngelo8
1Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany;
2Stanford Cancer Institute/Stanford University School of Medicine, Stanford, United
States of America; 3Institute of Mastocytosis Studies of Castilla La Mancha (CLMast)
─ Spanish Reference Center (CSUR) for Mastocytosis and CIBERONC, Virgen del Valle
Hospital, Toledo, Spain; 4Guy’s & St Thomas’ NHS Foundation Trust, Guy’s Hospital,
London, United Kingdom; 5Analysis Group, Inc., Boston; 6Blueprint Medicines Corporation,
Cambridge, United States of America; 7Blueprint Medicines Corporation, Zug, Switzerland;
8Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, United States
of America; 9Department of Internal Medicine I, Division of Hematology and Hemostaseology,
and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of
Vienna, Vienna, Austria
Background: Avapritinib, a selective and highly potent KIT D816V inhibitor, was approved
in the United States (US) for treatment of adults with advanced systemic mastocytosis
(AdvSM) based on results from two single-arm trials: EXPLORER (Phase 1; NCT02561988)
and PATHFINDER (Phase 2; NCT03580655). There is no comparative response data for avapritinib
versus best available therapy (BAT) for AdvSM.
Aims: This study (NCT04695431) compared duration of treatment (DOT) and maximum reduction
in serum tryptase levels between patients treated with avapritinib at a starting dose
≤200mg versus BAT.
Methods: A multi-center, global, observational, retrospective chart review study was
conducted at 6 study sites (4 European, 2 US) to identify and collect data from AdvSM
patients who received BAT. Patients were identified using inclusion/exclusion criteria
similar to EXPLORER and PATHFINDER trials. AdvSM patients receiving BAT could contribute
data on multiple lines of therapy (LOT); these data were compared with patient data
from the trials. DOT was defined as time from initiation to discontinuation of each
LOT. Maximum reduction in serum tryptase on treatment was defined during each LOT.
DOT was compared between cohorts using a Cox proportional hazards model and reduction
in serum tryptase levels was compared using a generalized estimating equation linear
model. Both models were weighted by inverse probability of treatment weights (IPTW)
to adjust for confounding by key variables: age, sex, region, AdvSM subtype, anemia,
thrombocytopenia, leukocyte count, skin involvement, number and type of prior treatments,
performance status, serum tryptase level, and presence of SRSF2/ASXL1/RUNX1 mutations.
Both models further adjusted for key covariates that remained unbalanced after weighting.
Subgroup analyses were conducted in patients who received ≥1 prior LOT (2L+), and
a 200mg starting dose of avapritinib.
Results: The DOT analysis included 136 avapritinib patients and 137 BAT patients.
Median (range) age for avapritinib and BAT patients was 68 (31−88) and 69 (21−88)
years, and 59% versus 66% of patients were male, respectively. In the avapritinib
group, 67%, 17%, and 16% of patients were diagnosed with SM with an associated hematological
neoplasm (SM-AHN), aggressive SM (ASM), and mast cell leukemia (MCL), respectively,
while in the BAT group, 55%, 29%, and 16% were diagnosed with SM-AHN, ASM, and MCL.
Among 188 BAT LOTs with agent-level data available, most common treatments were midostaurin
(53%) and cladribine (24%). Median DOT was 32.1 months (95% CI: 23.8, not estimable)
for avapritinib and 5.5 months (95% CI: 5.1, 7.0) for BAT. In IPTW-weighted Cox analysis,
DOT was significantly longer for avapritinib versus BAT (hazard ratio [95% CI]: 0.35
[0.23, 0.51]; P<0.001).
The analysis of reduction in serum tryptase included 135 avapritinib and 116 BAT patients.
Unadjusted maximum percentage reduction in serum tryptase levels was -84.8% (standard
deviation [SD]: 19.9%) for avapritinib versus -9.2% (SD: 161.4%) for BAT. The adjusted
mean difference in maximum percentage reduction in serum tryptase comparing avapritinib
to BAT was -69.8% (95% CI: -89.4%, -50.2%; P<0.001).
Results were similar in subgroups (Table 1).
Image:
Summary/Conclusion: AdvSM patients treated with avapritinib experienced significantly
longer DOT and greater reduction in serum tryptase levels compared to patients treated
with BAT. In the absence of RCTs, these data offer important and meaningful insight
into the improved effectiveness of avapritinib compared to alternative therapies for
AdvSM.
P1016: POLYCYTHEMIA VERA AND IMMUNE THROMBOGENESIS
R. Cacciola1,*, V. Vecchio2, E. Gentilini Cacciola3, E. CACCIOLA4
1Experimental and Clinical Medicine, Haemostasis Unit - University of Catania; 2University
of Catania, University of Catania School of Medicine, Catania; 3Public Health and
Infectious Diseases, Sapienza University, Roma; 4Medical, Surgical Sciences and Advanced
Technologies “G.F. Ingrassia”, Haemostasi Unit, Catania, Italy
Background: Polycythemia Vera (PV) is a Ph1-negative myeloproliferative neoplasm characterized
by vascular thrombosis and poor survival. The polycythemic thrombogenesis is associated
with erythrocytosis and red cell adhesiveness angiopathy. Recent studies reported
a JAK/STAT-mediated reduction of the regulatory T cells (Tregs) (CD4+, CD25high, CD127low,
FoxP3+) and loss of self-tolerance.
Aims: We investigated Tregs, anti-endothelial cell antibodies (AECA), and endothelial,
platelet and coagulation activation, in Polycythemia Vera (PV) and thrombosis.
Methods: We enrolled 60WHO-defined PV patients (30 men, 30 women; mean age 45±10 years)
without cardiovascular risk factors, autoimmune disease or thrombotic history. Of
PV patients, 40/60 had thromosis includingmyocardial infarction (MI) (10/60) according
to the WHO critera, deep vein thrombosis (20/60) and pulmonary embolism (10/60) on
lower-limb ultrasonography and computed tomography angiography, respectively. All
patients were evaluated for JAK2V617F allele burden, Tregs, AECA,Endothelial Leukocyte
Adhesion Molecule-1 (ELAM-1), Intercellular Adhesion Molecule-1 (ICAM-1),prothrombin
time (PT), activated partial thromboplastin time (APTT), fibrinogen (Fib) and D-dimer
(DD). JAK2V617F allele burden were analyzed by Polymerase chain reaction, Tregs were
measured by flow cytometry,AECA, ELAM-1 and ICAM-1 by ELISA, PT and APTT by coagulometric
test, Fib using Clauss method, and DD using ELISA.Complete blood hemostasis was studied
by PFA-100 on Collagen/ADP (CT-ADP) and Collagen/Epinephrine (CT-EPI) cartridges and
Thromboelastometry method on Clotting Time (CT), Clotting Formation Time (CFT), Maximum
Clot Firmness (MCF), and clot lysis at 30 minutes (LY-30).
Results: The patients with thrombosis had JAK2V616F allele burden higher (> 50%) compared
to patients without thrombosis (< 50Kroll%), lower Tregs (1,5±0,5% vs 3.5±1%),higher
AECA (200±20% vs 110±10%), ELAM-1 (80±10 ng/ml vs 45±5 ng/ml), and ICAM (170 ng/mL±10
vs 110±10 ng/mL), longer PT (30±10 s vs 20±2 s) and PTT (60±10 s vs 38±5 s), lower
Fib (90±20 mg/dl vs 120±20 mg/dl), higher DD (650±100 mg/l vs 300±50 mg/l) and shorter
C/ADP and C/EPI (C/ADP, n.v. 68-121 s (40±10 s vs 55±20 s) and C/EPI n.v. 84-160 s
(35±5 s vs 60±10 s). The patients with thrombosis had shorter CT (INTEM 35±20 s vs
70±20 s, EXTEM 20±10 s vs 30±5 s), shorter CFT (INTEM 15±10 s vs 25±5 s EXTEM 18±10
s vs 28±5 s), longer MCF (INTEM 130±10 mm vs 90±10 mm, EXTEM 120±10 mm vs 82±10 mm),
and lower LY-30 (INTEM 0.9% vs 15%, EXTEM 0.8% vs 15%). A positive correlation there
was between Tregs and AECA, ELAM-1 and ICAM-1 and thrombosis.
Summary/Conclusion: These findings shed new light on thrombotic pathogenesis in patients
with PV.
P1017: HARBOR: A PHASE 2/3 STUDY OF BLU-263 IN PATIENTS WITH INDOLENT SYSTEMIC MASTOCYTOSIS
AND MONOCLONAL MAST CELL ACTIVATION SYNDROME
M. Castells1,*, R. Scherber2,3, V. Bhavsar2, K. He2, C. Akin4
1Mastocytosis Center, Brigham and Women’s Hospital and Harvard Medical School, Boston;
2Blueprint Medicines Corporation, Cambridge; 3UT Health San Antonio, MD Anderson Cancer
Center, San Antonio; 4University of Michigan, Ann Arbor, United States of America
Background: The KIT D816V mutation plays a key role in the aggregation and accumulation
of aberrant mast cells which characterize systemic mastocytosis (SM), a rare, clonal
mast cell neoplasm. The disease subtype of indolent SM (ISM) is characterized by the
presence of mast cell aggregates in bone marrow and other organs which can lead to
chronic, debilitating, and potentially life-threatening symptoms. Mast cell infiltration
in the skin is common in ISM but is not always present. Similarly, monoclonal mast
cell activation syndrome (mMCAS) is a clonal KIT D816V–positive mast cell disease
without mast cell aggregates which can be accompanied by similar symptoms. Neither
ISM nor mMCAS currently have approved molecularly targeted therapies, and significant
symptom burden exists despite best supportive care (BSC) symptom-based therapy. BLU-263
is a novel, oral, next-generation tyrosine kinase inhibitor, exhibiting potent inhibition
of KIT D816V and an evolved preclinical profile with limited central nervous system
penetration. In our phase 1 study in normal healthy volunteers, BLU-263 was safe,
with linear pharmacokinetics across all tested doses and a half-life allowing once-daily
dosing, and thus supporting continued development for patients with SM.
Aims: HARBOR (NCT04910685) is a randomized, double-blind, placebo-controlled, phase
2/3 study assessing efficacy and safety of orally administered BLU-263 in patients
with ISM whose symptoms are not adequately controlled by standard therapies.
Methods: Part 1 of the study includes 3 dose groups (25 mg, 50 mg, and 100 mg) and
a placebo group combined with BSC therapies to determine the recommended dose (RD)
of BLU-263 in approximately 40 patients with ISM (Figure). After determination of
the RD and analysis of Part 1, Part 2 will open for enrollment of approximately 303
patients undergoing BSC therapies, randomized 2:1 to BLU-263 or placebo. The primary
endpoint of Part 2 is the proportion of patients achieving a ≥30% reduction in total
symptom score as assessed by the Indolent Systemic Mastocytosis Symptom Assessment
Form (ISM-SAF). Patients completing Part 1 or Part 2 will roll over to Part 3 for
open-label long-term evaluation of BLU-263 at the RD. The exploratory, open-label
study Part M will explore BLU-263 at the RD in patients diagnosed with mMCAS. Other
endpoints including but not limited to Mastocytosis Quality of Life scores, KIT D816V
allele burden, and bone marrow mast cell involvement will also be assessed. Two pharmacokinetic
(PK) groups will enroll patients with ISM prior to or concurrent with the Part 1 and
Part 2 study cohorts to better characterize the PK and safety of BLU-263 in specific
patient populations.
Results: Topline results of Part 1 of the study are anticipated in the second half
of 2022.
Image:
Summary/Conclusion: Overall, the goal of HARBOR is to investigate the safety and efficacy
of the selective, targeted KIT inhibitor BLU-263 as a potential treatment option to
reduce symptom burden for patients with ISM.
P1018: TREND OF CIRCULATING CD34+ CELLS IN MYELOFIBROSIS PATIENTS TREATED WITH RUXOLITINIB
D. Cattaneo1,*, N. Galli1, C. Bucelli2, S. Artuso3, A. Iurlo3
1Oncology and Hemato-Oncology, University of Milan; 2Hematology, Foundation IRCCS
Ca’ Granda Ospedale Maggiore Policlinico; 3Hematology, Foundation IRCCS Ca’ Granda
Policlinico, Milano, Italy
Background:
BCR-ABL1-negative myeloproliferative neoplasms (MPNs) are variably characterized by
an increase in peripheral blood (PB) and bone marrow (BM) progenitor cells. More specifically,
in myelofibrosis (MF) patients (pts) it has already been reported that the number
of circulating hematopoietic precursors is consistently high, with a relative circulating
CD34+ cells count of 15 ×106/L as the most frequently used criterion to discriminate
between MF and other MPNs. Nowadays, ruxolitinib (RUX) is used for the treatment of
splenomegaly and related symptoms in MF.
Aims: To evaluate CD34+ cells levels in a series of primary (PMF) and secondary (SMF)
MF pts at baseline and during treatment with RUX and identify any possible correlation
with response to treatment.
Methods: Between Oct 2014 and Dec 2021, CD34+ cells count from 49 consecutive pts
with PMF or SMF (32 males and 17 females; median age at RUX start, 70.6 years) was
retrospectively assessed at baseline and after 3, 6, 12 and 24 months (mts) from RUX
start. All pts were treated with RUX according to current indications, requiring an
IPSS risk of at least intermediate-1. At the time of CD34+ cells measurement, a complete
blood cells count was available for all pts and their spleen measurement [expressed
as the distance from the left costal margin (BCM) in cm] was taken.
Results: 24 pts were classified as having PMF (10 in the pre-fibrotic and 14 in the
overt fibrotic stage) and 25 as having SMF (15 had PPV- and 10 PET-MF).
JAK2V617F mutation was detected in 38 (77.6%) cases, CALR mutations in 8 (16.3%),
and MPL in 1 (2%). The remaining 2 pts (4.1%) were defined as “triple-negative”.
The median absolute number of circulating CD34+ cells in the overall population at
diagnosis was 83.5/mcL (range, 1-1528/mcL), with 31 (63.3%) pts showing >15 CD34+
cells/mcL.
At RUX start (after a median time from MF diagnosis of 33.9 mts), median absolute
number of CD34+ cells was 123/mcL (range, 2-1528/mcL), with 43 (87.7%) cases showing
higher than normal levels.
As expected, spleen measurements progressively decrease during the first mts of RUX
(Fig. 1A): in particular, the spleen was palpable at a median of 10 cm BCM at RUX
start, 5 and 3.5 cm after 3 and 6 mts of therapy, respectively. On the contrary, it
increases up to 5.5 cm after 12 mts, without significant changes after 24 mts.
Interestingly, with the exception of a transient increase after 3 mts of RUX therapy,
a progressive reduction in the absolute number of PB CD34+ cells after 6 and 12 mts
was documented in the whole cohort, with only a slight increase after 24 mts (Fig.
1B). However, considering PMF and SMF separately, a different behavior of CD34+ cells
was detected: in detail, in PMF pts circulating hematopoietic precursors gradually
decreased from 3 to 24 mts of RUX therapy in parallel with the persistence of a reduction
in the spleen size (Fig. 1C). On the contrary, in SMF pts both CD34+ cells and spleen
size still improve up to 6 mts of RUX, while they both regrow after 12 and 24 mts
of treatment (Fig. 1D).
Image:
Summary/Conclusion: Preliminary results of our study confirm that PB CD34+ cells are
increased in the majority of MF pts, both at diagnosis and during follow-up. To the
best of our knowledge, we have first reported the changes in circulating CD34+ cells
count during RUX, showing a parallel decrease in spleen diameter and circulating hematopoietic
precursors.
These findings suggest that this tool could facilitate the assessment of RUX responses
in MF pts, although this needs to be confirmed by further studies.
P1019: A PREDICTION RULE TO GUIDE JAK2 MUTATION TESTING IN PATIENTS WITH SUSPECTED
POLYCYTHEMIA VERA: RESULTS FROM THE JAK2 PREDICTION COHORT (JAKPOT) STUDY
B. Chin-Yee1,*, P. Bhai2, I. Cheong2, M. Matyashin1, C. Hsia1, E. Kawata3, J. Ho1,
M. Levy2, A. Stuart2, H. Lin2, I. Chin-Yee2, M. Kadour2, B. Sadikovic2, A. Lazo-Langner1
1Hematology; 2Pathology and Laboratory Medicine, Western University, London, Canada;
3Hematology and Oncology, Kyoto Prefectural University of Medicine, Kyoto, Japan
Background: The widespread availability of molecular testing for JAK2 mutations in
patients referred for elevated hemoglobin has facilitated the diagnosis of polycythemia
vera (PV) but also raises concerns of test overuse. A prediction rule could be useful
to improve test utilization.
Aims: In this study, we aimed to derive and validate of a simple rule using complete
blood count and white blood cell differential (CBC) parameters to predict the likelihood
of having a JAK2 mutation in patients referred for elevated hemoglobin.
Methods: We examined all adult patients with elevated hemoglobin (≥160 g/L for women,
or ≥165 g/L for men) who underwent JAK2 mutation testing using either quantitative
polymerase chain reaction (qPCR), single nucleotide polymorphism (SNP) allelotyping
or Next Generation Sequencing (NGS) panel between 2015 and 2021 at London Health Sciences
Centre in Ontario, Canada. We extracted data on age, sex, and complete blood count
and white blood cell differential (CBC) at the time of testing. All CBCs were performed
on a Sysmex XN Analyzer. Patients were randomly divided into derivation and validation
cohorts including all methods of JAK2 mutation testing. JAK2-positive and -negative
groups were compared using Student’s t-tests or χ2 tests, as appropriate. Continuous
variables were dichotomized at optimal cut-off points using receiving operating characteristic
curves. Potentially significant predictors were evaluated using multiple variable
stepwise logistic regression analysis with JAK2 positivity as the dependent variable.
A score was derived and internally validated using non-parametric bootstrapping. The
model was tested in the validation cohort and sub-analyses for each method were conducted
in a similar fashion. Test accuracy for the score was evaluated in all cohorts.
Results: The total study cohort included 901 patients (derivation n=616, validation
n=285). Population characteristics are shown in Table 1A. The final model included
1-point for any of the following: erythrocytes >6.45 × 1012/L, platelets >350 × 109/L,
and neutrophils >6.2 × 109/L. Patients with a score of 0 were considered low-risk;
all others were classified as high-risk. The percentage of JAK2 positive patients
in patients with a score of 1-3 was 24% versus 0.8% in patients with a score of 0.
The model had a sensitivity of 94.7% and a negative predictive value of 98.8% in the
derivation cohort; both sensitivity and negative predictive value were 100% in the
validation cohort. The percent of false negatives was 0.6% and 0% in the derivation
and validation cohorts, respectively (Table 1B). The results were consistent for each
testing method.
Image:
Summary/Conclusion: We developed and validated a simple rule using CBC parameters
to predict the likelihood of JAK2 mutation positivity in patients with elevated hemoglobin
with demonstrated high sensitivity and negative predictive value across different
JAK2 mutation testing platforms. In our cohort, the use of this rule to guide molecular
testing would have resulted in over 50% fewer tests. Further studies to prospectively
validate this prediction rule in different populations are planned.
P1020: SCREENING FOR PORTAL HYPERTENSION IN PATIENTS WITH PHILADELPHIA NEGATIVE MYELOPROLIFERATIVE
NEOPLASMS
M. Davidson1,*, F. Wong1, M. Atri1, H. Sibai1, D. Maze1, V. Cheung1, T. Nye1, J. Callum2,3,
E. Atenafu1, V. Gupta1
1Princess Margaret Hospital; 2University of Toronto, Toronto; 3Queen’s Unviversity,
Kingston, Canada
Background: Portal hypertension (PH) is a well-recognized complication of BCR-ABL
negative myeloproliferative neoplasms (MPNs) and is associated with significant morbidity
and mortality. However, the prevalence of PH and its associated risk factors have
not been studied systematically in this patient population. Screening by esophagogastroduodenoscopy
(EGD) can facilitate early diagnosis of PH and opportunity for intervention to prevent
complications.
Aims: To investigate the prevalence of PH and its patient and disease-related risk
factors.
Methods: From May 2013 to March 2019, MPN patients ≥ 18 years old with spleen >5 cm
below the costal margin in myelofibrosis (MF) or any size in Polycythemia Vera (PV)
or Essential Thrombocythemia (ET) were enrolled prospectively (NCT01816256). Patients
with a known history of portal or hepatic vein thrombosis, liver cirrhosis, and esophageal
varices were excluded. Screening for PH was done by EGD to evaluate for esophageal
varices (EV) or gastric varices (GV), and portal hypertensive gastropathy (PHG). Abdominal
Doppler ultrasonography (USG) was performed to evaluate for portal vein thrombosis
and spleen size. PH severity was based on the size of varices or severity of PHG.
For sample-size calculation, a precision approach was used, considering scenarios
where PH prevalence ranged from 5 to 10% and an upper limit for sample size of 100.
At a prevalence estimate of 8%, the 90% confidence interval (CI) was 4-14%, with an
acceptable lower limit for moving forward with a screening program. Prevalence of
PH was calculated as a proportion and CI are exact using Clopper-Pearson method.
Results: Among 102 enrolled patients, EGD and USG were completed in 85 and 84, respectively.
Primary and Post-PV/ET MF comprised the majority of the study population (93% [79/85]).
Evidence of PH was found in 32 patients (38%, [90% CI 29-47]) of whom 12 (14% [90%
CI 9-22]) had moderate to severe PH. EV, GV, and PHG were found in 81% (26/32), 13%
(4/32), and 31% (10/32) of patients, respectively. The prevalence of PH among MF and
PV/ET patients was 35% (28/79) and 67% (4/6), respectively. In MF patients, PH was
moderate-to-severe in 13% (90% CI 8-20). No cases of portal vein thrombosis were identified
on USG. Apart from a palpable liver observed more frequently in non-PH patients, there
were no other significant differences in baseline characteristics among patients with
and without PH. The frequency of JAK2, MPL and CALR mutations among PH patients was
78% (25/32),13% (4/32), and 6% (2/32). There was a trend towards more JAK2 mutations
in PH patients (78% [25/32] vs 64% [34/53], p=0.23), but no difference in DIPSS risk
or frequency of high molecular-risk mutations. Overall survival was similar in both
groups. PH-directed interventions were initiated in 59% (19/32); 28% (9/32) were started
on beta blocker (BB) prophylaxis, 16% (5/32) were treated with BB and variceal banding,
and 16% (5/32) continued on surveillance EGDs alone. Three patients with moderate
PH at baseline developed variceal bleeding (2) and ascites (1). All 3 patients were
on BB prophylaxis, and 1 had prior prophylactic banding. There were no adverse events
related to study procedures EGD or USG.
Image:
Summary/Conclusion: Given the study population, our results are generalizable to MF
patients. Our study shows that clinically significant PH is common in MF patients,
and led to additional clinical interventions in significant number of patients. These
data support the inclusion of asymptomatic PH screening in the work up of MF patients.
P1021: ERYTHROCYTOSIS, THROMBOCYTOSIS AND RATE OF RECURRENT THROMBOEMBOLIC EVENT -
A POPULATION BASED COHORT STUDY
A. Enblom-Larsson1,*, B. Andreasson2, H. Holmberg1, M. Liljeholm3, A. Själander1
1Department of Public Health and Clinical Medicine, Umeå University, Umeå; 2Department
of Hematology, NU Hospital Group, Uddevalla; 3Department of Radiation Sciences, Umeå
University, Umeå, Sweden
Background: The diagnosis and management of myeloproliferative neoplasms (MPN), polycythemia
vera (PV) and essential thrombocythemia (ET) are well established. Other conditions
with elevated hemoglobin (Hb), hematocrit (HCT) or platelets (Plt) are not as studied
and there are no international accepted guidelines of how to use phlebotomy and anticoagulants
to reduce thromboembolic complications.
Aims: To assess the frequency of elevated blood values in patients diagnosed with
a thromboembolic event, defined as myocardial infarction, ischemic stroke, transient
ischemic attack, peripheral arterial thromboembolism, pulmonary embolism, deep venous
thrombosis or abdominal thrombosis, and how many of these patients should be examined
further to find an underlying MPN, according to the 2016 revised WHO classification.
The second aim is to investigate if individuals with elevated Hb, HCT or Plt have
a higher rate of first recurrent thromboembolic event depending on underlying condition.
Methods: All patients over the age of 18 years with a thromboembolic event as defined
above during 2017 and 2018 in the county of Norrbotten (population n=250 230) were
included (n=3931). From medical records data of elevated blood values according to
the 2016 revised WHO criteria of PV and ET (Hb >16.5g/dL in men, Hb >16.0g/dL in women,
HCT >0.49 in men, HCT >0.48 in women or Plt >450 x109) were retrieved, as well as
underlying health conditions, recurrent thromboembolic events and death.
Results: In total 29.6% (1165/3931) had elevated Hb/HCT or Plt. Elevated Hb/HCT was
seen in 12.7% (501/3931), 4% (159/3931) had elevated Hb/HCT at the time of initial
thromboembolic event. Underlying conditions causing secondary erythrocytosis was found
in 25.5% (128/501). A MPN-diagnosis was found in 1.8% (9/501), and 72.7% (364/501)
had an unexplained erythrocytosis.
Elevated Plt were found in 17.3% (680/3931). Of these, 91.0% (619/680) had an obvious
reactive thrombocytosis while 2.4% (16/680) were diagnosed with MPN. The remaining
6.6% (45/680) had unexplained thrombocytosis. Thrombocytosis at the time of initial
thromboembolic event was found in 1.3% (51/3931).
In total 35% (405/1165) of the patients with elevated Hb/HCT or Plt should be investigated
further to rule out MPN according to current guidelines. Of these, 41 patients had
thrombocytosis, 360 erythrocytosis and 4 patients had both.
A recurrent thromboembolic event was found in 11% (434/3931), of these 9.7% (42/434)
had elevated Hb or HCT and 18.7% (81/434) elevated Plt. The highest rate of recurrent
events per 100 years was seen in the group with unexplained elevated platelets (8.11,
95% CI 2.37-13.84).
Patients with secondary erythrocytosis and unexplained thrombocytosis had the most
inferior event-free survival, 2.30 years (95% CI 1.95-2.64) and 2.51 (1.98-3.04).
On the other hand, patients with unexplained erythrocytosis displayed an event free
survival even surpassing that of patients without erythrocytosis or thrombocytosis
3.72 (3.55-3.89) vs 3.27 (3.2-3.3) years respectively.
Image:
Summary/Conclusion: Elevated Hb/HCT or Plt is common in patients with arterial or
venous events, affecting almost one third of all patients (1165/3931), out of these
one third (405/1165) should be investigated further to rule out MPN. Secondary erythrocytosis
and unexplained thrombocytosis were associated with the worst event-free survival
while patients with unexplained erythrocytosis were less prone to a recurrent thromboembolic
event.
These results highlights the importance of finding underlying cause of elevated blood
values to reduce risk of recurrent thromboembolic event.
P1022: OUTCOMES IN MYELOPROLIFERATIVE NEOPLASM PATIENTS WITH OR WITHOUT PRIOR HISTORY
OF CANCER: A POPULATION-BASED STUDY FROM ONTARIO
J. T. England1,*, A. Bankar1, W. C. Chan2, N. Liu2, M. C. Cheung3, V. Gupta1
1Medical Oncology/Hematology, Princess Margaret Cancer Centre; 2ICES; 3Hematology,
Sunnybrook Health Sciences Centre, Toronto, Canada
Background: Therapy-related myeloid neoplasms (tMNs) exist as a distinct diagnostic
entity in the 2016 WHO classification, and include MDS, MDS/MPN overlap, and AML that
develop following iatrogenic exposure to mutagenic agents and ionizing radiation.
Patients with the classical myeloproliferative neoplasms (MPNs), essential thrombocythemia
(ET), polycythemia vera (PV), and myelofibrosis (MF), are excluded from the tMN category.
Prior studies have observed an increased risk of other cancers in patients with MPNs,
possibly from shared risk factors including older age and chronic inflammation. The
impact of prior history of cancer on outcomes in MPN patients is not well understood.
A single centre study of MF (Masarova et al, Blood Advances, 2017) found no survival
difference between patients exposed to chemotherapy/radiation and those managed with
surgery/observation/hormone therapy.
Aims: To evaluate the clinical outcomes of MPN patients with or without prior non-MPN
cancer, and the impact of tMN-implicated therapy on survival following MPN diagnosis.
Methods: We conducted a population-based, retrospective cohort study using the ICES
provincial health databases. Included patients were residents of Ontario aged ≥18
years with an MPN diagnosis from Jan 1, 2004 to Dec 31, 2019. Diagnoses of MPN and
non-MPN (other cancers excluding AML) cancers were based on International Classification
of Diseases for Oncology, third edition (ICD-O-3) codes. Therapy for prior non-MPN
cancer were grouped as: therapy implicated in tMN per 2016 WHO criteria (alkylating
agents, antimetabolites, topoisomerase inhibitors, antitubulin agents, and ionizing
radiation), non-implicated therapies (targeted inhibitors, hormone therapy, other),
and surgery/observation alone. The primary outcome was overall survival (OS) from
the MPN diagnosis, with multivariable Cox regression adjusted for age and comorbidities.
In addition, to understand the patterns of non-MPN cancer type, therapy exposure,
and latency period, we compared the MPN cohort with a control cohort with no MPN history
matched (1:4) for age (±3 years), gender, geographic location, and neighbourhood income
quintile.
Results: A total of 10 336 MPN patients (5108 ET, 3843 PV, and 1385 MF) matched to
41 344 controls were identified in the study. MPN patients more frequently had a history
of non-MPN cancer compared to the control cohort (n=1421[13.5%] vs. n=5509[11.9%],
P<0.001). The most common non-MPN cancers were male genitourinary (24%), breast (18%),
gastrointestinal (18%), urothelial (9%), and melanoma (5%). Comparing MPN patients
to matched controls, there was no difference in the types of non-MPN cancer, treatment
exposures, or latency time.
After adjustment for age and comorbid conditions, the OS from PV/ET diagnosis was
predicted by prior cancer managed with surgery/observation hazard ratio (HR)[95% confidence
interval] 1.36 [1.22-1.52], tMN-implicated therapy HR1.83 [1.55-2.18] and non-implicated
therapy HR1.88 [1.49-2.38] (Fig 1a). In MF patients the OS was predicted by tMN therapy,
HR1.44 [1.04-1.98]; but not surgery/observation, HR1.17 [0.94-1.46], or non-implicated
therapy, HR1.25 [0.83-1.87](Fig 1b). Lack of availability of cytogenetics and mutation
profile in this population based dataset is the main limitation of this study.
Image:
Summary/Conclusion: Compared with matched controls, patients with MPNs are more likely
to have had a history of prior cancer. MPN patients with prior cancer have worse OS;
and those treated with systemic therapies have shorter OS than patients managed with
surgery/observation alone.
P1023: COHORT ANALYSIS OF MPN PATIENTS WITH CONCOMITANT MGUS DIAGNOSIS: A RETROSPECTIVE
STUDY
C. Fatigati1,*, F. Antonucci1, L. De Fazio1, R. Iannotta1, M. Lamagna1, A. Leone1,
F. Muriano1, G. Scairati1, F. Pane1, N. Pugliese1
1University of Naples Federico II, University of Naples Federico II, Napoli, Italy
Background: Patients with a Ph-negative myeloproliferative neoplasm (MPN), including
polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis
(PMF), may harbor or develop plasma-cell dyscrasia such as monoclonal gammopathy of
undetermined significance (MGUS) and multiple myeloma (MM), however, the clinical
and molecular determinants of this co-occurrence are still uncertain.
Aims: The main aim of this study was to evaluate correlation between MGUS and MPN
related features and clinical course.
Methods: We retrospectively analyzed clinical data from 448 patients with a diagnosis
of MPNs according to 2016 WHO criteria, performed from 1989 to 2020 at our Institution.
Here, we described the clinical and genetic features of MPN patients with concomitant
MGUS. Data collection included NGS panel, cytogenetic analysis, clinical history (including
thrombotic events) and pharmacological therapy during the entire MPNs follow up.
Results: Among the 448 consecutive patients analyzed, 33 (7.4%) displayed both MPN
and MGUS.
Twenty-three of them were females while 10 males. MPN diagnosis were ET (12/33, 36%),
MF (11/33, 33%), PV (8/33, 24%), or pre-MF (2/33, 6%) at a median age of 61 years
(28-84); 25 patients showed JAK2 V617F mutation (75.8 %), 3 patients type I CALR mutation
(9.1%), 1 patient harbored W515K MPL mutation (3%), and 4 were triple negative (12.1%).
Half of them incurred a MPN diagnosis antecedent (19/33, 57.6%) or synchronous (11/33,
33%) with MGUS diagnosis, while only 3 patients (9.1%) had MGUS diagnosed before MPN.
Serum M protein consisted in a majority of IgG (84.8%, 14 cases ʎ e 14 ƙ light chain),
2 cases of IgA and 1 case of IgM protein with a median count of 1.04 plasma-cells
in bone marrow aspirate at diagnosis.
Eventually, after a median follow-up of 7 years (range 1-30 years), 5 patients (15.2%)
originated a second hematological malignancy and 2 patients (6.1%) evolved in secondary
myelofibrosis. In particular, 3 patients (10%) developed Non-Hodgkin lymphoma, 1 patient
MM, and 1 patient developed acute myeloid leukemia (AML).
NGS, cytogenetic, clinical history and MPN specific therapies don’t seem to influence
clinical course of analyzed patients.
Summary/Conclusion: In literature there are few publications that highlight the coexistence
of MGUS in MPN patients. Currently little is known about the underlying molecular
mechanisms and there are no guidelines clarifying whether a particular treatment or
follow-up is necessary.
The incidence of MGUS in our MPN cohort is higher than general population matched
for sex and age. Although, this data can be influenced by an accurate hematological
follow-up, we can’t exclude a real increased rate of MGUS in MPN patient.
It is known MPN can predispose to an higher risk of developing second primary malignancies
such as B cellular lymphoproliferative disorders. Interestingly, in our cohort the
percentage of patients with coexisting MGUS and MPN who developed second hematological
malignancies seemed slightly higher than literature data.
Further studies with case-control cohort are necessary to better understand the possible
prognostic impact and mutual influence between MPN and MGUS.
P1024: IMPACT OF TP53 IN MYELOFIBROSIS UNDERGOING STEM CELL TRANSPLANTATION
N. Gagelmann1,*, A. Badbaran1, V. Panagiota2, C. Wolschke1, F. Ayuk1, F. Thol2, M.
Ditschkowski3, B. Cassinat4, M. Robin4, T. Schroeder3, M. Heuser2, H. C. Reinhardt3,
N. Kröger1
1University Medical Center Hamburg-Eppendorf, Hamburg; 2Medical School Hannover, Hannover;
3University Hospital Essen, Essen, Germany; 4APHP, Paris, France
Background: Most of myelofibrosis (MF) cases harbor somatic mutations in the driver
genes JAK2, CALR, or MPL. Other somatic nondriver mutations have been increasingly
detected with the use of high-throughput sequencing. The role of TP53 in prognosis
of MF has long been unclear, but recent first studies suggested worse survival for
patients harboring this mutation.
Aims: While allogeneic stem cell transplantation still remains the only curative treatment
option, outcome of patients with TP53 has not been evaluated yet.
Methods: Here, we analysed in 417 patients with PMF or post ET/PV MF incidence and
impact on otcome of TP53. We detected 46 patients with detectable TP53 mutation using
next-generation sequencing, and compared characteristics and outcome after allograft
with 371 non-TP53 patients with primary or secondary MF. Samples were collected at
time of transplantation. Primary end points were overall survival (OS) and nonrelapse
mortality. Secondary objectives were relapse, relapse-free survival, and graft-versus-host
disease (GVHD).
Results: Median follow-up from transplantation was 6.1 years for the TP53 group and
7.0 years for the non-TP53 group, respectively (P=0.50). Baseline characteristics
were generally balanced, whereas more patients in the TP53 group had Karnofsky performance
score <90 (57% versus 40%; P=0.04). Median OS was 2.1 years (0.1-5.4 years) for the
TP53 group versus 15.4 years (11.5-19.3 years) for the non-TP53 group (P=0.002), and
the 6-year OS was 40% (24-55%) for the TP53 group versus 65% (60-70%) for the non-TP53
group. 6-year nonrelapse mortality was 35% (21-49%) for the TP53 group versus 27%
(22-32%) for the non-TP53 group (P=0.16) and 6-year incidence of relapse was 26% (13-39%)
for the TP53 group versus 18% (14-21%) for the non-TP53 group (P=0.09). 6-year relapse-free
survival was 35% (20-50%) versus 53% (48-58%; P=0.01). Acute GVHD occurred in 63%
versus 57% (P=0.43). Of patients with relapse, 92% in the TP53 group versus 63% in
the non-TP53 group died (P=0.05). Risk for death appeared to be associated with variant
allele frequency (P=0.06).
Summary/Conclusion: MF patients with TP53 appeared to have higher nonrelapse mortality
and relapse incidence but significantly worse relapse-free and overall survival, mainly
because of worse survival after relapse.
P1025: RUXOLITINIB-ASSOCIATED CRYPTOCOCCOSIS: A SYSTEMATIC REVIEW AND META-ANALYSIS
M.-A. Gaman1,2,*
1Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy; 2Hematology
Department, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical
Institute, Bucharest, Romania
Background: The use of the JAK 1/2 inhibitor ruxolitinib (RUX) in myeloproliferative
neoplasms (MPNs), namely primary (PMF) or secondary myelofibrosis (MF) and polycythemia
vera (PV), has been associated with the development of opportunistic infections due
to the immunosuppressive actions of this drug on cellular immunity. However, there
is still controversy regarding the association between ruxolitinib treatment and the
occurrence of cryptococcosis, a fungal infection caused by the yeast Cryptococcus
neoformans or Cryptococcus gattii.
Aims: To report on cryptococcosis cases in RUX-treated MPNs and report on epidemiology,
clinical manifestations, RUX associations, outcomes.
Methods: Systematic search computed in PubMed/Medline, Web of Science and SCOPUS,
from the inception of these databases until 28.02.2022, to identify relevant English-written
articles. Eligibility criteria: 1. Confirmed MPNs diagnosis, 2. Confirmed diagnosis
of cryptococcosis, 3. MPNs patients receiving RUX therapy.
Results: The systematic search yielded 60 potentially relevant papers. After removal
of duplicates (n=20) and screening of abstracts/titles (n=29), 11 manuscripts were
selected for full-text review. After applying the eligibility criteria, 10 articles
were entered into the qualitative and quantitative analysis. In total, 10 case reports
on cryptococcosis occurring in RUX-treated MPNs patients were detected: 80.0% males,
mean age 66.8±8.3 years, range 51-79 years. Most reports (70.0%) came from Asia (3
from Japan, 2 from Thailand, 1 from India and Taiwan each) and from the USA (30.0%)
and were published between 2013 and 2021. In total, 70% were diagnosed with PMF, whereas
cryptococcosis also occurred in PV, post-PV MF or post-ET MF (10.0%, 1 subject each).
The average time from RUX initiation was 22.1±14.5 (6-48) months. HIV status was reported
in 60.0% of cases and all subjects were apparently immunocompentent and had negative
HIV screening. Cryptococcosis was caused by Cryptococcous neoformans only and occurred
most commonly as disseminated infection (40.0%), followed by cryptococcal meningitis
(30.0%) or pulmonary cryptococcosis (30.0%). Cryptococcosis co-occurred with tuberculosis
(20.0%), disseminated histoplasmosis (10.0%) or Mycobacterium haemophilum cellulitis
(10.0%). Most common presentations were fever (70.0%), neurological symptoms/signs
(headache, consciousness/visual disturbances, 40.0%), shortness of breath (30.0%)
or fatigue/lethargy (30.0%). Positive cryptococcosis diagnosis was established based
on serum/cerebrospinal fluid (CSF) cryptococcal antigen positivity (80.0%) and/or
positive CSF, blood or bronchoalveolar fluid cultures (70.0%). Specific treatment
consisted in amphotericin B (50.0%) and/or fluconazole (70.0%). Amphotericin B was
given in combination with flucytosine (20.0%) or fluconazole (20.0%) or in monotherapy
(30.0%). Other combinations or drugs, i.e., isavuconazole, voriconazole, were used
due to renal toxicity related to amphotericin B use. RUX was discontinued in 70.0%
of cases and restarted in 20.0% of subjects. Cryptococcosis-related death occurred
in 30.0% of cases.
Image:
Summary/Conclusion: Cryptococcosis can developed during ongoing RUX therapy in MPNs
subjects, even if apparently immunocompetent, potentially due to the immunosuppressive
actions of this drug on cellular immunity. Screening for Cryptococcus neoformans in
selected individuals might reduce the risk of disseminated infection and death. RUX
discontinuation during antifungal treatment is advised due to reported progression
of lesions and drug-drug interactions with antifungal agents.
P1026: COMPARATIVE GENOMIC PROFILING OF MYELOPROLIFERATIVE NEOPLASMS PRESENTING WITH
AND WITHOUT SPLANCHNIC VEIN THROMBOSIS
M. Garrote1,*, M. López-Guerra1, E. Arellano-Rodrigo2, M. Magaz3, A. Triguero4, S.
Carbonell4, J. R. Alamo1, F. Turon3, V. Hernandez-Gea3, A. Baiges3, D. Colomer1, J.
C. García-Pagán3, F. Cervantes4, A. Alvarez-Larrán4
1Pathology - Hematopathology Unit; 2Hemotherapy and Hemostasis; 3Hepatology; 4Hematology,
Hospital Clínic de Barcelona, Barcelona, Spain
Background: Splanchnic vein thrombosis (SVT) is the initial manifestation in 5% of
myeloproliferative neoplasms (MPN). MPN patients presenting with SVT are usually younger,
have less abnormal blood counts and lower JAK2V617F allele burden than other MPN patients.
However, it is unknown whether they have a different genomic background.
Aims: To describe the clinical and genomic profile of MPN presenting with SVT and
to compare it with a matched group of MPN without SVT.
Methods: Samples of 190 MPN patients were selected: 64 MPN presenting with SVT (SVT+)
and 126 MPN without SVT (SVT-). SVT+ and SVT- were matched according to sex, age,
MPN clinical subtype and driver mutation. Next Generation Sequencing was performed
using Myeloid Solution by Sophia Genetics, which includes 30 genes recurrently mutated
in MPN. Variants were selected using Sophia DDM software and were subsequently classified
into 5 categories: 5 (definitely pathogenic), 4 (likely pathogenic), 3 (uncertain),
2 (likely benign), and 1 (benign). Only variants included into 4 and 5 categories
were taken into account. Patients were classified according to the genomic classification
proposed by Grinfeld and colleagues.
Results: One hundred and three patients were women (54.2%) and 87 patients were men
(45.8%), without significant differences between SVT+ and SVT- groups. Median age
was 44 years (range: 15-85) and 50 years (range: 15-87) in SVT+ and SVT-, respectively
(p not significant). Regarding MPN clinical subtype in SVT+ group, 39 (60.9%), 15
(23.4%) and 10 (15.6%) corresponded to Polycythemia Vera (PV), Essential Thrombocythemia
(ET) and MPN unclassifiable, respectively. In the SVT- group, 75 (59.5%) and 51 (40.5%)
corresponded to PV and ET, respectively.
Genomic classification in the SVT+ and SVT- groups is shown in Table 1. The proportion
of patients with high molecular risk (TP53 disruption/aneuploidy, chromatin/spliceosome
mutation, or homozygous JAK2 mutation) was 14.1% and 26.2% for SVT+ and SVT-, respectively
(p=0.06). Median number of pathogenic mutations was 1 (range: 1-4) in both SVT+ and
SVT- groups. Pathogenic mutations in TET2, DNMT3A and ASXL1 were present in 10%, 3.7%
and 3.7%, respectively, without significant differences between both groups.
With a mean follow-up of 10.1 years for cases and 9.7 years for controls, 27 patients
died (13 SVT+ and 14 SVT-). Median survival was 24 years and not reached for SVT+
and SVT-, respectively (p=0.1). On multivariate analysis, SVT+ group showed a higher
risk of death (HR: 2.4, 95%CI: 1.1-5.5, p=0.03) after correction by age, sex and genomic
classification. Transformation to myelofibrosis and acute leukemia was low and similar
in both groups: 4.7% vs. 5.6% for myelofibrosis in SVT+ and SVT-, respectively (p
not significant); and 0% vs. 2.4% for acute leukemia (p not significant). On multivariate
analysis, patients with TP53 disruption/aneuploidy, chromatin/spliceosome mutation,
or homozygous JAK2 showed a higher risk of disease progression to myelofibrosis or
acute leukemia (HR: 14.5, 95%CI: 3.1-68.7, p=0.001) regardless of the presence of
SVT at diagnosis, age and sex.
Image:
Summary/Conclusion: MPN presenting with SVT and MPN-matched controls showed a low
molecular complexity. Genomic classification was helpful in identifying a small proportion
of MPN patients presenting with SVT who are at high risk of disease progression. SVT
at MPN diagnosis is associated with a higher risk of death independent of sex, age
and genomic classification.
P1027: RESPONSES TO AVAPRITINIB IN PATIENTS WITH ADVANCED SYSTEMIC MASTOCYTOSIS: HISTOPATHOLOGIC
ANALYSES FROM EXPLORER AND PATHFINDER CLINICAL STUDIES
T. George1,*, K. H. Karner1, K. A. Moser1, A. Rets1, A. Reiter2, D. H. Radia3, M.
Deininger4, H.-M. Lin5, S. Dimitrijević6, D. J. DeAngelo7
1Department of Pathology, ARUP Laboratories, University of Utah, School of Medicine,
Salt Lake City, United States of America; 2Department of Hematology and Oncology,
University Hospital Mannheim, Heidelberg University, Mannheim, Germany; 3Guy’s & St
Thomas’ NHS Foundation Trust, London, United Kingdom; 4Versiti Blood Research Institute,
Milwaukee; 5Blueprint Medicines Corporation, Cambridge, United States of America;
6Blueprint Medicines Corporation, Zug, Switzerland; 7Department of Medical Oncology,
Dana-Farber Cancer Institute, Boston, United States of America
Background: Systemic mastocytosis (SM) is a mast cell (MC) neoplasm driven by the
KIT D816V mutation in ~95% of cases. Diagnosis includes evaluation of MC aggregates
in extracutaneous organs, atypical MC morphology, and MC immunophenotype. Avapritinib,
a potent and selective KIT D816V inhibitor, has been approved in the US for patients
with advanced SM (AdvSM) based on the phase 1 EXPLORER and phase 2 PATHFINDER studies.
Aims: Here, we describe a comprehensive evaluation of the effect of avapritinib on
bone marrow (BM) pathology in patients treated in these studies.
Methods: Patients aged ≥18 years with diagnoses of AdvSM per local investigator were
treated with once-daily avapritinib <200 mg (n=17), 200 mg (n=126), and ≥300 mg (n=50).
Bone marrow biopsies (BMBs), aspirates (BMAs), and peripheral blood (PB) smears were
obtained at screening, and after 8 and 24 weeks of treatment with avapritinib. Evaluations
were performed using standard Wright-Giemsa and H&E staining, and IHC was performed
on formalin-fixed EDTA-decalcified BMBs using standard techniques for CD25 and CD30.
Changes in fibrosis were assessed by the European Consensus on grading of BM fibrosis
(MF score).
Results: As of April 2021, 193 patients enrolled in EXPLORER (n=86) and PATHFINDER
(n=107) were included in analyses with centrally confirmed diagnoses of aggressive
SM (n=29), SM with an associated hematologic neoplasm (n=119), mast cell leukemia
(n=28), indolent SM (n=14), smoldering SM (n = 2) or CMML (n=1). Median (range) age
was 67 years (31–88), 56% were male and 88% had a KIT D816V mutation. Following treatment
with avapritinib, a decrease in the proportion of patients with multifocal dense aggregates
of MCs in BMBs was observed from 93% at screening to 50% by Week 8 (Table). Avapritinib
treatment reduced the proportion of aberrant CD25+ and CD30+ MCs in BMBs by Week 8
and Week 24 (Table). In BMAs, avapritinib reduced the mean MC burden from 11% of total
nucleated cells at screening to 2% at Week 8 and by Week 24, with reduced proportions
of immature and spindle-shaped MCs (Table). Of 9 patients with circulating MCs at
screening and post-screening sample measurements (6 with SM with associated hematologic
neoplasm diagnoses and 3 with MCL), 8 had no detectable MCs in PB by Week 8. The proportion
of patients with fibrosis in BMBs decreased from 96% (178/186) at screening to 90%
(142/158) at Week 8 to 84% (103/123) through week 24, and MF scores were reduced during
avapritinib treatment in those patients presenting with increased fibrosis at screening
(Table).
Image:
Summary/Conclusion: Patients treated with avapritinib were observed to have rapid
(Week 8) and marked (Week 24) reductions in neoplastic BM MCs with a return to a normal
morphologic appearance and immunophenotype. This was accompanied by an improvement
of fibrosis and a decrease in circulating MCs. These improvements in disease histopathology
provide further support that treatment with this highly selective and potent KIT D816V
inhibitor is disease modifying in AdvSM.
P1028: PTG-300 (RUSFERTIDE) TREATMENT INTERRUPTION REVERSES HEMATOLOGICAL GAINS AND
RESTORES THERAPEUTIC BENEFIT ON REINITIATION IN SUBJECTS WITH POLYCYTHEMIA VERA
A. Kuykendall1,*, M. Kremyanskaya2, Y. Ginsburg2, N. Pemmaraju3, E. Ritchie4, J. Gotlib5,
F. Valone6, S. Khanna6, S. Gupta6, R. Hoffman2, S. Verstovsek3
1Hematology and Med Oncology, USF, Moffitt Cancer Center, Tampa; 2Hematology, Mt.
sinai, New york; 3Dept of Leukemia, M.D. Anderson, Houston, Tx; 4Hematology, Weill
Cornell medical College, New york; 5Hematology, school of Medicine, Stanford, Palo
Alto, Ca; 6Clinical Research and Development, Protagonist Therapeutics, Newark, United
States of America
Background: Polycythemia vera (PV) patients are treated with periodic therapeutic
phlebotomy (TP) and if needed, cytoreductive therapy to maintain hematocrit (HCT)
<45% to reduce the incidence of thrombosis. Constitutive erythropoiesis and repeated
TP result in iron deficiency which further stimulates iron absorption and availability
for robust erythropoiesis. Iron deficiency contributes to fatigue, which is the most
severe and prevalent symptom for patients with PV. We have previously shown that adding
Rusfertide, a hepcidin mimetic, to existing PV treatment, significantly, and effectively
reverses iron deficiency and essentially eliminates TP requirement for >1 year in
PV patients [Hoffman ASH 2021]. The ongoing PTG-300-04 (NCT04057040) Phase 2 study
of Rusfertide in TP-dependent PV patients experienced an FDA-mandated brief hold which
resulted in dosing interruption of Rusfertide for all patients.
Aims: To examine the impact of temporary rusfertide withdrawal on iron homeostasis
and TP requirements after temporary suspension in the ongoing PTG-300-04 study.
Methods: Eligibility criteria for PTG-300-04 study include PV diagnosis [by 2016 WHO
criteria] and ≥3 phlebotomies with/without concurrent cytoreductive therapy in the
24 weeks before enrollment. Rusfertide 10-120 mg weekly SQ injection was added to
existing cytoreductive treatment and adjusted to maintain HCT <45%. During the brief
clinical hold patients were maintained on their cytoreductive regimens and had TP
for HCT >45%. Most patients returned to Rusfertide add-on treatment 2-3 months after
the dosing interruption and reinititation.
Results: Pre-enrollment, PV patients were treated with TP alone (30) or with TP+cytoreductive
therapy (33). TP requirements in the 24 weeks before enrollment ranged from 3 to 9.
Rusfertide treatment enabled consistent HCT control <45%, essentially eliminated TPs
in all sub-groups, decreasing RBC count and increased MCV and MCH values. Pre-enrollment,
mean iron-related parameters were consistent with systemic iron deficiency. Rusfertide
treatment resulted in progressive normalization of serum ferritin, suggesting a redistribution
of systemic iron. During Rusfertide dosing interruption, all patients had significant
(p<0.01) increase in TPs, HCT, and RBC count; and a simultaneous decrease in MCV and
serum ferritin. Patients also reported increase in PV-related symptoms (i.e., increased
fatigue and worsening concentration). Reinitiating Rusfertide resulted in rapid normalization
of hematologic parameters, elimination of TP, and improvement in symptoms demonstrating
potential effectiveness of this approach. Most adverse events were grade 1-2. The
most frequent AE was transient injection site reaction reported in 59% of patients.
Summary/Conclusion: The current results support Rusfertide as an effective treatment
option in PV, reversing iron deficiency and essentially eliminating TP requirements
in PV patients with/without cytoreductive agents. Reversal of hematological parameters
during dosing interruption further confirms the effect of Rusfertide on both iron
homeostasis and erythrocytosis. Taken together, Rusfertide is a very promising novel
biologic agent in both low and high-risk PV patients. A global Phase 3 trial of rusfertide
in PV, VERIFY, has been initiated.
P1029: MATCHING-ADJUSTED INDIRECT COMPARISON (MAIC) OF PELABRESIB (CPI-0610) IN COMBINATION
WITH RUXOLITINIB VS RUXOLITINIB OR FEDRATINIB MONOTHERAPY IN PATIENTS WITH INTERMEDIATE
OR HIGH-RISK MYELOFIBROSIS
V. Gupta1,*, J. Mascarenhas2, M. Kremyanskaya2, R. K. Rampal3, M. Talpaz4, J.-J. Kiladjian5,
A. Vannucchi6, S. Verstovsek7, G. Colak8, D. Dey9, C. Harrison10
1Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada; 2Tisch Cancer
Institute, Icahn School of Medicine at Mount Sinai; 3Memorial Sloan Kettering Cancer
Center, New York, NY; 4University of Michigan Comprehensive Cancer Center, Ann Arbor,
MI, United States of America; 5Hôpital Saint-Louis, Université de Paris, Paris, France;
6Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy;
7University of Texas MD Anderson Cancer Center, Houston, TX; 8Constellation Pharmaceuticals
a MorphoSys Company, Boston, MA, United States of America; 9MorphoSys AG, Planegg,
Germany; 10Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
Background: Pelabresib (CPI-0610), an investigational product, in combination with
ruxolitinib (ruxo) has shown encouraging responses in terms of ≥35% reduction in spleen
volume from baseline (SVR35) and ≥50% reduction in total symptoms score (TSS) from
baseline (TSS50) in Janus kinase inhibitor (JAKi) treatment-naïve patients with intermediate
or high-risk myelofibrosis (MF) in Arm 3 of the open-label Phase 2 MANIFEST study
(NCT02158858). A cross-trial comparison with ruxo or fedratinib monotherapy has limitations
in the presence of the potential imbalances in baseline characteristics.
Aims: Here we report findings from matching-adjusted indirect comparisons (MAICs),
conducted to correct for potential imbalances in baseline characteristics, comparing
the primary endpoint SVR35 and secondary endpoint TSS50 at Week (Wk) 24 for the combination
of pelabresib and ruxo (MANIFEST Arm 3) with Phase 3 ruxo monotherapy (COMFORT-I and
II, SIMPLIFY-1) and fedratinib monotherapy (JAKARTA) data.
Methods: Individual patient level data were available for MANIFEST Arm 3, while only
published summary data were available for COMFORT-I and II, SIMPLIFY-1 and JAKARTA.
An unanchored MAIC was conducted wherein patients in MANIFEST Arm 3 were reweighted
to adjust for imbalances in the following key prognostic baseline characteristics:
gender, MF subtype, International Prognostic Scoring System risk status, previous
hydroxyurea use, platelet count, hemoglobin levels, spleen volume and JAK2V617F status.
Weighted mean outcomes of the treatment effects were estimated together with their
95% confidence interval (CI) using robust sandwich estimators for variance. The weights
were also used to calculate the Effective Sample Size (ESS). Treatment effect outcomes
for SVR35 and TSS50 at Wk 24 are presented in terms of Response Rates (RR) and Response
Rate Ratio (RRR, defined as RR in MANIFEST Arm 3/RR in comparator arm). An RRR >1
favors the pelabresib + ruxo arm in MANIFEST Arm 3 over the JAKi monotherapy comparator
arm. A limitation of a MAIC is that bias due to potential unmeasured differences in
patient baseline characteristics between trials cannot be excluded.
Results: The MAIC analysis resulted in complete summary-level balance in the weighted
distributions of the prognostic factors in MANIFEST Arm 3 versus comparators. The
figure presents the weighted (or MAIC-adjusted) and unweighted (or unadjusted) treatment
effect comparisons along with their 95% CI for SVR35 and corresponding ESS at Wk 24.
Statistically significant MAIC-adjusted (weighted) RRRs of 1.57 (95% CI 1.10, 2.24;
p-value: 0.012), 1.82 (95% CI: 1.17, 2.83; p-value: 0.008), 2.13 (95% CI: 1.51, 3.02;
p-value: <0.001), 1.76 (95% CI: 1.16, 2.66; p-value: 0.008) and 1.55 (95% CI: 1.06,
2.27; p-value: 0.023) were observed for MANIFEST Arm 3 (pelabresib + ruxo) versus
COMFORT-I and -II and SIMPLIFY-1 (ruxo only) and JAKARTA (fedratinib 400 mg and 500 mg),
respectively. These weighted analyses are consistent with statistically significant
results of unadjusted analyses. RRRs >1 were also observed for TSS50 at Wk 24 for
all the comparisons.
Image:
Summary/Conclusion: The MAIC analysis, adjusting for eight key baseline prognostic
factors, suggests an improvement in SVR35 and TSS50 at Wk 24 for JAKi treatment-naïve
patients with MF treated with a combination of pelabresib and ruxo over ruxo or fedratinib
monotherapy. MANIFEST-2 (NCT04603495), a Phase 3 double-blind, randomized study assessing
the efficacy and safety of pelabresib or placebo in combination with ruxo in patients
with MF, is currently open for enrollment.
P1030: MANIFEST-2, A GLOBAL, PHASE 3, RANDOMIZED, DOUBLE-BLIND, ACTIVE-CONTROL STUDY
OF PELABRESIB (CPI-0610) AND RUXOLITINIB VS PLACEBO AND RUXOLITINIB IN JAK INHIBITOR-NAÏVE
MYELOFIBROSIS PATIENTS
C. Harrison1,*, R. K. Rampal2, V. Gupta3, S. Verstovsek4, M. Talpaz5, J.-J. Kiladjian6,
R. Mesa7, A. Kuykendall8, A. Vannucchi9, F. Palandri10, S. Grosicki11, T. Devos12,
E. Jourdan13, M. J. Wondergem14, H. K. Al-Ali15, V. Buxhofer-Ausch16, A. Alvarez-Larrán17,
S. Akhani18, R. Muñoz-Carerras19, Y. Sheykin19, G. Colak19, M. Harris19, J. Mascarenhas20
1Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom; 2Memorial Sloan-Kettering
Cancer Center, New York, NY, United States of America; 3Princess Margaret Cancer Centre,
University of Toronto, Toronto, Canada; 4Leukemia Department, University of Texas
MD Anderson Cancer Center, Houston, TX; 5University of Michigan Comprehensive Cancer
Center, Ann Arbor, MI, United States of America; 6Hôpital Saint-Louis, Université
de Paris, Paris, France; 7Mays Cancer Center at UT Health San Antonio MD Anderson
Cencer Center, San Antonio, TX; 8Mofitt Cancer Center, Tampa, FL, United States of
America; 9Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence;
10IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”,
Bologna, Italy; 11Medical University of Silesia in Katowice, Katowice, Poland; 12University
Hospitals Leuven and Laboratory of Molecular Immunology (Rega Institute), KU Leuven,
Leuven, Belgium; 13C.H.U, Nîmes, France; 14Amsterdam University Medical Centers, Amsterdam,
Netherlands; 15University Hospital Halle, Halle, Germany; 16KRANKENHAUS DER ELISABETHINEN
Linz GmbH, Linz, Austria; 17Hospital Clínic Barcelona, Barcelona, Spain; 18MorphoSys
AG, Planegg, Germany; 19Constellation Pharmaceuticals a MorphoSys Company, Boston,
MA; 20Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY,
United States of America
Background: Myelofibrosis (MF) is characterized by bone marrow fibrosis, anemia, splenomegaly
and constitutional symptoms. Progressive bone marrow fibrosis results from aberrant
megakaryopoeisis and expression of proinflammatory cytokines, both of which are heavily
influenced by bromodomain and extraterminal domain (BET)-mediated gene regulation
and lead to myeloproliferation and cytopenias. Pelabresib (CPI-0610) is an oral small-molecule
investigational inhibitor of BET protein bromodomains currently being developed for
the treatment of patients with MF. It is designed to downregulate BET target genes
and modify nuclear factor kappa B (NF-κB) signaling. MANIFEST-2 was initiated based
on data from Arm 3 of the ongoing Phase 2 MANIFEST study (NCT02158858), which is evaluating
the combination of pelabresib and ruxolitinib in Janus kinase inhibitor (JAKi) treatment-naïve
patients with MF. Primary endpoint analyses showed splenic and symptom responses in
68% and 56% of 84 enrolled patients, respectively.
Aims: MANIFEST-2 (NCT04603495) is a global, Phase 3, randomized, double-blind, active-control
study of pelabresib and ruxolitinib versus placebo and ruxolitinib in JAKi treatment-naïve
patients with primary MF, post-polycythemia vera MF or post-essential thrombocythemia
MF. The aim of this study is to evaluate the efficacy and safety of pelabresib in
combination with ruxolitinib. Here we report updates from a recent protocol amendment.
Methods: The MANIFEST-2 study schema is shown in Figure 1. Key eligibility criteria
include a Dynamic International Prognostic Scoring System (DIPSS) score of Intermediate-1
or higher, platelet count ≥100 × 109/L, spleen volume ≥450 cc by computerized tomography
or magnetic resonance imaging, ≥2 symptoms with an average score ≥3 or a Total Symptom
Score (TSS) of ≥10 using the Myelofibrosis Symptom Assessment Form v4.0, peripheral
blast count <5% and Eastern Cooperative Oncology Group performance status ≤2. Patient
randomization will be stratified by DIPSS risk category (Intermediate-1 vs Intermediate-2
vs High), platelet count (>200 × 109/L vs 100–200 × 109/L) and spleen volume (≥1800 cm3
vs <1800 cm3). Double-blind treatment (pelabresib or matching placebo) will be administered
once daily for 14 consecutive days, followed by a 7-day break, which is considered
one cycle of treatment. Ruxolitinib will be administered twice daily for all 21 days
of the cycle. The primary endpoint is SVR35 response (≥35% reduction in spleen volume
from baseline) at Week 24, and the key secondary endpoint is TSS50 response (≥50%
reduction in TSS from baseline) at Week 24. Other secondary endpoints include safety,
pharmacokinetics, changes in bone marrow fibrosis, duration of SVR35 response, duration
of TSS50 response, progression-free survival, overall survival, conversion from transfusion
dependence to independence and rate of red blood cell transfusion for the first 24
weeks.
Results: Study recruitment is ongoing; 400 patients (200 per arm) from North America,
Europe, Asia and Australia will be enrolled. The study opened for enrollment in November
2020.
Image:
Summary/Conclusion: MANIFEST-2 was initiated based on data from the ongoing Phase
2 MANIFEST study with the aim of assessing the efficacy and safety of pelabresib and
ruxolitinib in JAKi treatment-naïve patients with MF. MANIFEST-2 is currently open
for enrollment.
P1031: LOW-DOSE DOACS IN VERY HIGH-RISK MYELOPROLIFERATIVE NEOPLASMS: LESS BLEEDING
BUT MORE ARTERIAL THROMBOTIC EVENTS
L. Herbreteau1,*, L. Papageorgiou2,3, L. Le Clech4, G. Garcia5, C. James5, B. Pan-Petesch1,6,
F. Couturaud7, E. Lippert8,9, G. Gerotziafas2,3, J.-C. Ianotto1,6,9
1Service d’Hématologie Clinique, Centre Hospitalier Universitaire de Brest, Brest;
2Service d’Hématologie Biologique, Hôpital Tenon, Hôpitaux Universitaires de l’Est
Parisien, Assistance Publique Hôpitaux de Paris; 3Research Group “Cancer, Hémostase
et Angiogenèse” INSERM UMR_S 938, Centre de Recherche Saint-Antoine Faculté de Médecine,
Institut Universitaire de Cancérologie, Sorbonne Universités, Paris; 4Service de Médecine
Interne, Maladies Infectieuses et du Sang, Centre Hospitalier de Cornouaille, Quimper;
5Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Bordeaux, Bordeaux;
6GETBO, Groupe d’Etude des Thromboses en Bretagne Occidentale, EA3878; 7Département
de Médecine Interne et de Pneumologie; 8Laboratoire d’Hématologie, Centre Hospitalier
Universitaire de Brest, Brest; 9France Intergroupe des néoplasies Myéloprolifératives,
Paris, France
Background: Direct oral anticoagulants (DOACs) have recently proven their efficacy
and safety, as primary and secondary prevention agents, for thrombosis in cancer patients.
Thrombotic and haemorrhagic events are major causes of morbidity and mortality in
Philadelphia-negative myeloproliferative neoplasm (MPN) patients. However, the best
agent to prevent thrombotic events without increasing the bleeding incidence among
these patients remains uncertain.
Aims: In this study, we aimed to determine if DOACs might be an interesting choice
to reduce the thrombotic risk in high-risk MPN patients.
Methods: We analysed a large multicentric cohort of MPN patients treated with DOACs
for atrial fibrillation (AF) or thrombotic events. We evaluated the overall thrombosis
and haemorrhage rates related to DOACs, and also by the DOAC type (apixaban or rivaroxaban)
as well as the daily dosage prescribed.
Results: We included 135 MPN patients with a median follow-up of 23.8 months since
DOAC initiation. Twenty patients (14.8%) developed 30 thrombotic events (28 arterial
thromboses in 19 patients) for a global incidence of 6.5% patient-years. No difference
was highlighted between apixaban and rivaroxaban in terms of thrombosis risk, but
the incidence of arterial thrombosis was significantly higher on low-dose DOACs (11.9
vs. 4.5% patient-years, p=0.04). Bleeding events were more frequent in the full-dose
group (15.2 vs. 41.2%, p=0.006). However, major and clinically relevant non major
(CRNM) bleeding events occurred in 18 patients (13.3%), with no difference between
the DOAC types or the DOAC doses. In multivariate analysis, age was the only identified
thrombotic risk factor, whereas risk factors for major or CRNM bleeding were a full-dose
treatment regimen and a combination of DOAC and low-dose aspirin.
Image:
Summary/Conclusion: DOACs seem effective in preventing venous thrombosis in MPN patients
with AF or thrombotic events. For these high-risk patients, low-dose DOACs exposed
patients to more arterial thrombosis but fewer bleeding events. Prospective studies
are now needed to evaluate and compare DOACs to the currently recommended antithrombotic
drugs for high-risk MPN patients.
P1032: REAL-LIFE VALIDATION OF MIPSS70: A RETROSPECTIVE MULTICENTER AND NGS ANALYSIS
FROM THE GEMFIN DATABASE
A. Hernández-Sánchez1,2,*, Á. Villaverde-Ramiro2, A. Álvarez-Larrán3, E. Arellano-Rodrigo3,
M. Pérez Encinas4, F. Ferrer Marín5,6,7,8, E. Such9, G. Carreño Gómez-Tarragona10,
N. de las Heras Rodríguez11, M. A. Durán Pastor12, M. I. Mata Vázquez13, L. Fox14,
J. M. Raya Sánchez15, E. Magro Mazo16, M. T. Gómez Casares17, M. J. Ramírez Sánchez18,
B. Xicoy Cirici19, Á. Ramírez Páyer20, M. J. Fernández Llavador21, A. Á. Martín López1,2,
A. Mosquera Orgueira4, J. Martínez Elicegui2, M. Garrote3, B. Bellosillo22, T. González1,2,23,
J. Hernández-Rivas1,2,23, J. C. Hernández Boluda24
1Hospital Universitario de Salamanca; 2Instituto de Investigación Biomédica de Salamanca
(IBSAL), Salamanca; 3Hospital Clínic de Barcelona, Barcelona; 4Complexo Hospitalario
Universitario de Santiago, Santiago de Compostela; 5Hospital Morales Meseguer; 6Universidad
Católica San Antonio de Murcia (UCAM); 7Centro de Investigación Biomédica en Red de
Enfermedades Raras (CIBERER); 8Instituto Murciano de Investigación Biosanitaria, Murcia;
9Hospital Universitari I Politècnic La Fe, Valencia; 10Hospital Universitario 12 de
Octubre, Madrid; 11Hospital Universitario de León, León; 12Hospital Universitari Son
Espases, Palma de Mallorca; 13Complejo Hospital Costa Del Sol, Marbella; 14Hospital
Universitari Vall D’Hebron, Barcelona; 15Hospital Universitario de Canarias, Santa
Cruz de Tenerife; 16Hospital Universitario Príncipe de Asturias, Alcalá de Henares;
17Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas de Gran Canaria; 18Hospital
Universitario de Jerez, Jerez de la Frontera; 19Institut Català d’Oncologia-Hospital
Universitari Germans Trias I Pujol de Badalona, Badalona; 20Hospital Universitario
Central de Asturias, Oviedo; 21Hospital Universitario Dr. Peset, Valencia; 22Hospital
del Mar, Barcelona; 23Centro de Investigación del Cáncer (CIC), Salamanca; 24Hospital
Clínico Universitario de Valencia, Valencia, Spain
Background: Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized
by the presence of driver mutations (JAK2, CALR or MPL) in most of the patients. The
development of next generation sequencing (NGS) has favored the incorporation of mutational
information in novel prognostic scores. However, most of these risk stratification
models also include cytogenetic information, which is only available for a fraction
of patients with this disease, thus reducing their real-life applicability. Mutation-enhanced
international prognostic scoring system for transplant-age patients (MIPSS70) is based
on clinical and mutational risk factors and it is useful in the absence of cytogenetic
information, but it has not been validated yet in a real-life clinical setting.
Aims: To validate MIPSS70 risk stratification model for transplant-age PMF patients
from a multicenter database.
Methods: From the GEMFIN multicenter database, 668 myelofibrosis patients with enough
information to calculate MIPSS70 were selected. As the score is intended for patients
aged ≤70 years with PMF, the final analysis was carried out in 218 patients meeting
these criteria. Patients who received allogeneic stem cell transplantation were censored
on the date of transplantation.
Results: Median age was 61 years (range 18-70), 69% of patients were male and median
follow-up time was 4.65 years. Regarding the presence of MIPSS70 variables at disease
diagnosis, 45% of patients had constitutional symptoms, 43% had hemoglobin <100g/L,
15% leukocytes >25 (x109/L) and 33% platelets <100 (x109/L). Circulating blasts were
≥2% in 30% and most patients presented BM fibrosis grade ≥2 (74%). CALR type 1 mutation
was detected only in 12%, while any high-molecular risk (HMR) mutation was present
in 31% (8% for ≥2 HMR mutations). In brief, in comparison to MIPSS70 training cohort,
the patients in the present series were older and presented a higher proportion of
adverse risk factors (Figure 1A).
More than half of the patients were classified as high risk according to MIPSS70 (52%),
while 35% were intermediate and 13% low risk. The 5-year overall survival (OS) was
44% (median 4.6 years), 77% (median 13 years) and 95% (median not reached) for high,
intermediate and low risk patients respectively (p<0.001, Figure 1B). The 5-year leukemia-free
survival (LFS) was 42%, 77% and 95% for high, intermediate and low risk groups (p<0.001).
Univariate analysis of each MIPSS70 variable on OS was carried out, with statistically
significant differences being confirmed for all variables except fibrosis grade and
HMR mutations. ASXL1 was mutated in 27%, but it did not impact OS in our patients
(HR 0.96, p=0.89). SRSF2 was the only MIPSS70 mutated gene (8%) associated with inferior
OS (HR 2.5, p=0.04), while EZH2 and IDH1/2 mutations were only present in 2.5% of
patients each. However, the analysis of the complete NGS panel allowed the identification
of other genes associated with shorter OS in our series: CBL (HR 3.7, p=0.014), SETBP1
(HR 4.9, p=0.034) and KRAS (HR 4.6, p=0.041), with a tendency for U2AF1 (HR 2.3, p=0.061)
and NRAS (HR 3.6, p=0.083).
Image:
Summary/Conclusion: This study constitutes the first real-life validation of MIPSS70
in a multicenter analysis. The score was able to stratify our cohort in three categories
with significant differences in OS and LFS. Of note, mutation on ASXL1 gene was not
associated with shorter OS in our patients. By contrast, several other genes with
potential prognostic significance were identified. Further cooperative studies are
needed in order to unravel PMF mutational landscape and its prognostic implications.
P1033: A PHASE 2 STUDY OF THE LSD1 INHIBITOR IMG-7289 (BOMEDEMSTAT) FOR THE TREATMENT
OF ESSENTIAL THROMBOCYTHEMIA (ET)
F. Palandri1,*, D. M. Ross2, T. Cochrane3, C. Tate3, S. W. Lane4, S. R. Larsen5, A.
T. Gerds6, A. B. Halpern7, J. Shortt8, J. M. Rossetti9, K. M. Pettit10, J. Liang11,
A. Mead12, M. Marchetti13, A. Vannucchi14, A. Wilson15, J. R. Göthert16, M. Hanna17,
A. Jones18, J. Peppe18, G. Natsoulis18, T. McClure18, W.-J. Hong18, W. S. Stevenson19,
C. N. Harrison20, M. Talpaz10, N. Vianelli21, H. Y. Rienhoff Jr.18
1Institute of Hematology “L. & A. Seràgnoli”, Sant’Orsola-Malpighi University Hospital,
Bologna, Italy; 2Department of Haematology, Royal Adelaide Hospital and SA Pathology,
Adelaide; 3Department of Haematology, Gold Coast University Hospital, Southport; 4QIMR
Berghofer Medical Research Institute, Brisbane; 5Institute of Haematology, Royal Prince
Alfred Hospital, Sydney, Australia; 6Cleveland Clinic Taussig Cancer Institute, Cleveland;
7Department of Medicine, Division of Hematology, University of Washington, Seattle,
United States of America; 8Monash Haematology, Monash Health, Clayton, Australia;
9UPMC Hillman Cancer Center, Pittsburgh; 10Department of Internal Medicine, Division
of Hematology/Oncology, University of Michigan, Ann Arbor, United States of America;
11Middlemore Clinical Trials, Auckland, New Zealand; 12MRC Weatherall Institute of
Molecular Medicine, University of Oxford, Oxford, United Kingdom; 13Azienda Ospedaliera
Nazionale SS. Antonio e Biagio e Cesare Arrigo, Alessandria; 14University of Florence,
AOU Careggi, CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms,
Florence, Italy; 15University College Hospital, NHS Foundation Trust, London, United
Kingdom; 16Department of Hematology, West German Cancer Center (WTZ), University Hospital
Essen, Essen, Germany; 17North Shore Hospital. Waitemata District Health Board, Auckland,
New Zealand; 18Imago BioSciences, Inc., South San Francisco, United States of America;
19Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia;
20Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom; 21Institute of
Hematology, “L. & A. Seragnoli”, Azienda Ospedaliero-Universitaria di Bologna, Bologna,
Italy
Background: Lysine-specific demethylase-1 (LSD1) is critical for the self-renewal
malignant stem cells and maturation of megakaryocytes, the cell central to ET pathogenesis.
Bomedemstat is an oral LSD1 inhibitor active in mouse models of MPNs including improved
survival (Kleppe et al. 2015; Jutzi et al. 2018). IMG-7289-CTP-201 is an ongoing,
global, open-label, Phase 2b study of bomedemstat taken QD for 24+ weeks in patients
with ET who are resistant/intolerant to at least one standard treatment (NCT04254978).
Aims: Key objectives are safety and response, defined as platelet (plt) count ≤400x109/L
without thromboses. Exploratory endpoints include durability of response (defined
as plt ≤400x109/L for ≥12 wks) symptom improvement (TSS) and reduction of WBCs and
mutation burden.
Methods: Key eligibility criteria: cytoreduction required, platelet count >450x109/L,
haemoglobin (Hb) ≥10 g/dL. The starting dose is 0.6 mg/kg/d and titrated, if needed,
to a platelet count of 200-400x109/L.
Results: At data cut-off (3Feb’22), 44 patients had enrolled. Median age was 68 (42-92)
yrs; of the patients most recently treated with hydroxyurea, 88% met ELN criteria
for resistance/ intolerance. The Day 1 mean plt, WBC and Hb values were 825x109/L
(457-2220), 8.9x109/L (3.9-30.6), and 13.1 g/dL (9.4-16.5), respectively. Median TSS
at baseline was 15 (0-74). Sequencing 261 genes (N=42) identified mutations in JAK2
(45%), CALR (43%), and MPL (5%). Other mutations (e.g., EZH2, ASXL1, SF3B1, TP53)
were present in 29%.
Median time on treatment is 29 weeks (0-68). For patients treated ≥12 weeks, 91% (31/34)
achieved a plt count of ≤400x109/L in a median time of 8.1 weeks. 83% of patients
(20/24) treated for >24 weeks achieved a durable response as defined above. Of the
9/30 (30%) patients Day 1 WBC count ≥10x109/L, 89% (8/9) had a WBC count <10x109/L;
all maintained a stable hemoglobin (see figures). In patients with baseline TSS ≥10
(29/43), at Week 12 69% (11/16) showed reductions; 38% (6/16) had improvements >10-points.
The median baseline TSS score for fatigue was 5 (N=41). At Week 24, the median score
for evaluable patients fell to 3 and dropped to a median of 1 at Week 48. At Week
24 (N=14), JAK2 and CALR mutations were equally sensitive: 87% had a decrease in allele
frequencies (mean: -29%; [S.D. 5%]; range -3 to -91). No new mutations were found.
The most common AEs regardless of causality were dysgeusia (52%), fatigue (34%), constipation
(32%), arthralgia (27%), thrombocytopenia (25%), and contusion (21%). Eight patients
reported a total of 12 serious AEs (SAE), with two (thrombocytopenia and mouth haemorrhage
reported in 1 patient) deemed related per the PI. Seven patients discontinued treatment,
6 due to AEs (fatigue, invasive ductal breast carcinoma, headache, respiratory failure,
dysgeusia x 2) and one withdrew consent on Day 1. Similar to an ongoing MF study of
bomedemstat (NCT03136185), there have been no safety signals or deaths related to
drug. At the censor date, 84% (37/44) of patients remain on study.
Image:
Summary/Conclusion: To date, in this patient population, bomedemstat is generally
well-tolerated, reduces platelets, improves symptoms, reduces the mutation burden,
and moderates WBC counts without affecting haemoglobin. For those patients treated
for at least 24 weeks, 79% achieved a durable reduction in platelet count to ≤400x109/L
without thromboembolic events.
A Phase 3 study of bomedemstat for the treatment of ET is being planned.
P1034: HEALTHCARE UTILIZATION OF PATIENTS WITH HYPEREOSINOPHILIC SYNDROME IN EUROPE
J. Hwee1,*, N. Kwon2, R. Alfonso-Cristancho3, L. Baylis4, G. Requena5, S. Du6, A.
Khanal6, L. Huynh6, M. S. Duh6, R. W. Jakes5
1Value Evidence & Outcomes, GSK, Mississauga, ON, Canada; 2Respiratory Research &
Development, GSK, Brentford, Middlesex, United Kingdom; 3Value Evidence & Outcomes,
GSK, Collegeville, PA, United States of America; 4Global Medical Affairs, GSK, Durham,
NC, United States of America; 5Epidemiology, GSK, London, United Kingdom; 6Analysis
Group, Inc., Boston, MA, United States of America
Background: Hypereosinophilic syndrome (HES) is a rare group of blood disorders characterized
by prolonged eosinophilia that can lead to tissue and organ damage. HES was previously
considered to be largely idiopathic, but myeloid and lymphocytic variants are also
now recognized. The disease may manifest in multiple organ systems, most commonly
the heart, nervous system, gastrointestinal tract, skin, and lungs, due to eosinophilic
infiltration. Treatment aims to reduce eosinophil counts, and the current standard
of care consists of high-dose glucocorticosteroids, despite the risks associated with
high doses or long exposure, and antineoplastic agents for those with more severe
disease or who do not respond to steroids. Healthcare resource utilization (HCRU)
in patients with HES is not well characterized, and data from clinical trials may
not be representative of the general disease population encountered in real-world
settings.
Aims: To describe HES-related HCRU among a real-world cohort of patients with a confirmed
diagnosis of HES in Europe.
Methods: This retrospective chart review study included patients ≥6 years old with
a confirmed diagnosis of HES and ≥1 year of follow-up data from index (first physician
encounter: Jan 2015–Dec 2019). Chart review was conducted by physicians in 5 European
countries (France, Germany, Italy, Spain, and the United Kingdom [UK]). Data from
a similar number of patients were reviewed and included across each country. HES-related
hospitalizations and outpatient and emergency room (ER) visits were assessed and reported
descriptively across all patients and separately by country.
Results: Of the 280 patients included, the majority were male (65.0%) and had idiopathic
HES (55.4%); patients had a mean (standard deviation [SD]) age at HES diagnosis of
42.4 (16.2) years. The most common comorbidity was asthma (45.0%). The mean disease
duration was 4.0 (4.5) years, and the mean length of follow-up was 2.8 (1.4) years.
Overall, 85 (30.4%) patients had ≥1 HES-related hospitalization. For patients with
a HES-related hospitalization, the mean (SD) length of stay (LoS) was 11.0 (9.4) days.
HES-related ER visits were reported for 72 (25.7%) patients, and HES-related outpatient
visits were reported for 243 (86.8%) patients. Patients had a mean of 0.4 (1.2) hospitalizations,
0.3 (0.8) ER visits, and 4.3 (4.9) outpatient visits (of which 1.0 [3.3] were unscheduled
outpatient visits) per year (Table). HCRU varied between countries. The proportion
of patients requiring hospitalization was highest in the UK (43.5%) and lowest in
Spain (11.5%). Mean (SD) days of LoS during a HES-related hospitalization was highest
in Spain (15.3 [4.2]) and lowest in the UK (9.9 [11.0]) and France (9.9 [7.3]). The
proportion of patients with outpatient visits was highest in France (93.4%) and lowest
in Italy (78.8%). The proportion of patients with ER visits was highest in the UK
(37.1%) and lowest in Italy (13.5%).
Image:
Summary/Conclusion: These results demonstrate that real-world patients with HES have
substantial HCRU, including lengthy hospitalizations, ER visits, and outpatient visits,
which is higher than that reported in previous clinical trials for control (placebo-treated)
patients with eosinophilic diseases such as EGPA and severe eosinophilic asthma. These
observations highlight the need for novel approaches to treatment to relieve the disease-related
burden of HES for patients and to ameliorate the associated HCRU burden.
P1035: COUNTRY DIFFERENCES IN THE BURDEN OF PATIENTS WITH HYPEREOSINOPHILIC SYNDROME
IN EUROPE
J. Hwee1,*, N. Kwon2, R. Alfonso-Cristancho3, L. Baylis4, G. Requena5, S. Du6, A.
Khanal6, L. Huynh6, M. S. Duh6, R. W. Jakes5
1Value Evidence & Outcomes, GSK, Mississauga, ON, Canada; 2Respiratory Research &
Development, GSK, Brentford, Middlesex, United Kingdom; 3Value Evidence & Outcomes,
GSK, Collegeville, PA, United States of America; 4Global Medical Affairs, GSK, Durham,
NC, United States of America; 5Epidemiology, GSK, London, United Kingdom; 6Analysis
Group, Inc., Boston, MA, United States of America
Background: Hypereosinophilic syndrome (HES) is a group of rare blood disorders characterized
by peripheral eosinophilia that can lead to tissue and organ damage. Diagnosis and
management of HES can be challenging owing to differences in patient characteristics,
disease characteristics and clinical manifestations. At present, there is limited
information available on the burden of HES in Europe.
Aims: To describe real-world patient demographics, disease characteristics, clinical
manifestations, treatment patterns, and clinical outcomes for patients with HES across
Europe.
Methods: This retrospective chart review study included patients ≥6 years old with
a confirmed diagnosis of HES and ≥1 year of follow-up data from index (first physician
encounter: Jan 2015–Dec 2019). Chart review was conducted by physicians in 5 European
countries (France, Germany, Italy, Spain, and the United Kingdom [UK]). Data from
a similar number of patients were reviewed and included across each country. Patients
could be newly diagnosed during the patient identification window or could be diagnosed
prior to their index date, allowing for the inclusion of patients with differing disease
duration and organ involvement. Patient demographics, disease characteristics, clinical
manifestations, treatment patterns, and clinical outcomes were summarized and described
across the overall population and separately by country.
Results: Overall, 280 patients were included in the study. Most patients were male
(65.0%), with the highest proportion of male patients in Spain (73.1%) and France
(75.4%). The majority of patients (55.4%) had idiopathic HES. The most common comorbidity
overall was asthma (45.0%); however, in the UK, anxiety/depression was more common
than asthma (51.6% vs 41.9%), and in France, both anxiety/depression and hypertension
were more common than asthma (37.7% vs 23.0% and 36.1% vs 23.0%, respectively) (Table).
Most patients (88.6%) were newly diagnosed with HES during the study time frame. Italy
had the highest proportion of patients (17.3%) diagnosed prior to the study while
Germany had the lowest (1.9%). The overall mean (standard deviation [SD]) number of
distinct clinical manifestations was 3.7 (3.7), with the highest mean (SD) numbers
of clinical manifestations observed in Italy (4.6 [4.7]) and Spain (4.7 [3.7]). Patients
across all 5 countries were treated with 4 classes of therapy: oral corticosteroids
(OCS; 89.3%), immunosuppressants or cytotoxic agents (63.6%), and biologics (43.9%),
other therapies (10.7%). Spain had the highest mean (SD) maximum daily dose of OCS
(42.4 [19.6] mg). In total, 22.9% of patients experienced a flare, and the mean (SD)
number of flares in patients with ≥1 flare was 0.5 (0.3) flares/year. Patients in
France, Germany, and the UK had the highest mean (SD) numbers of flares (0.7 [0.6],
0.6 [0.3], and 0.6 [0.3] flares/year, respectively). The overall mean (SD) duration
of flares was 2.9 (3.2) months, with the longest flare duration observed in France
(4.8 [6.3] months) and the shortest in the UK (1.6 [1.2]).
Image:
Summary/Conclusion: These results demonstrate that in patients with HES, there are
differences in patient and disease characteristics as well as treatment strategies
across Europe. These differences likely contribute to challenges in the diagnosis
and management of HES and highlight the need for a common strategy to address these
difficulties.
P1036: VOLUMETRIC SPLENOMEGALY IN PATIENTS WITH POLYCYTHEMIA VERA
M.-W. Lee1,*, S.-H. Yeon1, H. Ryu1, I.-C. Song1, H.-J. Lee1, H.-J. Yun1, S. Y. Kim2,
J. E. Lee3, K. S. Shin3, D.-Y. Jo1
1Department of Internal Medicine; 2Department of Laboratory Medicine; 3Department
of Radiology, Chungnam National University College of Medicine, Daejeon, South Korea
Background: At present, palpable splenomegaly is relatively uncommon in patients with
polycythemia vera (PV), possibly because of early diagnosis facilitated by a lowering
of the diagnostic thresholds for hemoglobin and hematocrit levels as well as the wide
application of driver gene mutation tests. Radiological volumetric analysis of spleen
size is now readily available. However, non-palpable splenomegaly in patients with
PV has seldom been addressed.
Aims: In this retrospective study, we evaluated non-palpable, volumetric splenomegaly
defined based on age- and body surface area (BSA)–matched criteria in patients with
PV diagnosed according to the 2016 World Health Organization diagnostic criteria.
Methods: Patients with PV who underwent abdominal computed tomography (CT) and who
had palpable splenomegaly at diagnosis from January 1991 to December 2020 at Chungnam
National University Hospital were enrolled in the study. The spleen volume of each
patient was determined by volumetric analysis of abdominal CT and adjusted for the
patient’s age and BSA. And then the degree of splenomegaly was classified as follows:
no splenomegaly, spleen volume less than the mean plus 2 SD of the reference volume
based on both age and BSA; overt volumetric splenomegaly, spleen volume greater than
the mean plus 3 SD of the reference volume based on both age and BSA; borderline volumetric
splenomegaly, a spleen volume between no splenomegaly and overt splenomegaly; or palpable
splenomegaly, spleen palpable below the left costal margin.
Results: Of the 87 PV patients enrolled in the study, 15 (17.2%) had no splenomegaly,
whereas 17 (19.5%), 45 (51.7%), and 10 (11.5%) had borderline volumetric, overt volumetric,
and palpable splenomegaly, respectively. Spleen volume was significantly positively
correlated with lactate dehydrogenase level (r = 0.227, P = 0.042) and tended to be
positively related to white blood cell count (r = 0.209, P = 0.055) and monocyte count
(r = 0.210, P = 0.059) at diagnosis. Spleen volume was not correlated with hemoglobin
level, platelet count, or JAK2V617F burden at diagnosis. The degree of splenomegaly
did not affect the cumulative incidence of thrombotic vascular events (10-year incidence:
7.7%, 0%, 22.3%, and 50.7%, respectively, P = 0.414). By contrast, splenomegaly tended
to adversely affect myelofibrotic transformation (10-year cumulative incidence: 0%,
0%, 7.1%, and 30.3%, respectively, P = 0.062). Moreover, the cumulative incidence
of myelofibrotic transformation was significantly higher in patients with overt volumetric
or palpable splenomegaly than those with no or borderline volumetric splenomegaly
(10-year incidence: 0% vs. 10.3%, respectively; 15-year incidence: 0% vs. 26.3%, respectively,
P = 0.020). Overall survival (OS) differed among patients with different degrees of
splenomegaly (15-year OS: 100%, 78.6%, 71.7%, and 51.9%, respectively, P = 0.021).
The degree of splenomegaly was independent risk factor for both myelofibrotic transformation
(hazard ratio [HR]: 7.75; 95% confidence interval [CI]: 1.87-21.10; P = 0.005) and
OS (HR: 6.70; 95% CI: 1.89-23.72; P = 0.003).
Summary/Conclusion: The degree of splenomegaly, including volumetric splenomegaly,
based on age- and BSA-matched reference spleen volumes at diagnosis reflects disease
progression in PV patients. Therefore, volumetric splenomegaly should be evaluated
at the time of PV diagnosis and taken into consideration when predicting the prognosis
of patients with this disorder.
P1037: DOES EARLY INTERVENTION IN MYELOFIBROSIS IMPACT OUTCOMES? A POOLED ANALYSIS
OF THE COMFORT I AND II STUDIES
C. Harrison1,*, J.-J. Kiladjian2, A. M. Vannucchi3, R. A. Mesa4, R. M. Scherber5,
J. Hamer-Maansson5, S. Verstovsek6
1Guy’s and St. Thomas’ NHS Foundation Trust, Guy’s Hospital, London, United Kingdom;
2Hôpital Saint-Louis, Assitance Publique – Hôpitaux de Paris, Université de Paris,
Inserm, Paris, France; 3CRIMM, Center for Research and Innovation of Myeloproliferative
Neoplasms, AOU Careggi, University of Florence, Florence, Italy; 4Mays Cancer Institute
at UT Health San Antonio MD Anderson, San Antonio; 5Incyte Corporation, Wilmington;
6The University of Texas MD Anderson Cancer Center, Houston, United States of America
Background: Myelofibrosis (MF) is characterized by cytopenias, splenomegaly, burdensome
symptoms, and poor overall survival (OS). Few studies have investigated if earlier
intervention with targeted MF therapies affects response and OS. In a pooled analysis
of the COMFORT I and II trials of ruxolitinib (RUX), patients (pts) who received RUX
at randomization or after crossover from placebo (PBO) or best available therapy (BAT)
had improved OS.
Aims: To assess the association of MF disease duration before treatment with disease
outcomes using pooled COMFORT data
Methods: COMFORT I (NCT00952289) and COMFORT II (NCT00934544) were phase 3 trials
of RUX vs PBO or BAT, respectively, in pts with intermediate-2 or high-risk MF who
provided written informed consent. In this post hoc analysis, data from RUX-treated
pts in both studies were combined (RUX group), and data from the PBO/BAT arms were
pooled (control group). Pt subgroups were defined based on disease duration before
treatment initiation (≤12 mo or >12 mo from diagnosis). Assessments included frequency
of thrombocytopenia (platelets [PLT] <100 Gi/L or PLT transfusion) and anemia (hemoglobin
<100 g/L or red blood cell transfusion) events, spleen volume response (SVR; spleen
volume reduction ≥35% from baseline [SVR35]), symptom response (MF-Symptom Assessment
Form total symptom score [TSS] reduction ≥50% from baseline [TSS50]; available in
COMFORT I only), and OS. OS was assessed using the Kaplan-Meier method; pts randomized
to PBO/BAT were included in the PBO/BAT group regardless of crossover. A multivariable
analysis (MVA) using a logistic regression model was used to examine factors associated
with SVR.
Results: There were 525 pts in the analysis (RUX: ≤12 mo, n=84; >12 mo, n=216; PBO/BAT:
≤12 mo, n=66; >12 mo, n=159). Median age across groups ranged from 65 to 70 y. Baseline
clinical characteristics were generally similar across subgroups, but pts with shorter
vs longer disease duration were slightly younger with higher blood counts. Among pts
who received RUX, fewer thrombocytopenia events were observed among those treated
earlier (≤12 mo vs >12 mo), with differences seen as early as Wk 4–8 on treatment
(18% vs 33%) and sustained over time; a similar trend was observed for anemia events
(Wk 4–8, 59% vs 72%). The proportion of pts with SVR35 was greater among those who
initiated RUX earlier (≤12 mo vs >12 mo) at Wk 24 (48% vs 33%; P=0.0610) and 48 (44%
vs 27%; P=0.0149; Table). A numerically greater proportion of pts who initiated RUX
at ≤12 mo vs >12 mo achieved TSS50 at Wk 24 (56% vs 40%; P=0.0829; Table). OS at Wk
240 was improved among pts who initiated RUX at ≤12 mo vs >12 mo (63% [95% CI, 51%–73%]
vs 57% [95% CI, 49%–64%]; P=0.0430; Table). Comparatively, OS was longer with RUX
vs PBO/BAT regardless of disease duration. A sensitivity analysis using a 24-mo cutoff
was also conducted but yielded weaker associations between disease duration and SVR,
TSS, and OS. In the MVA, a significantly greater binary SVR was seen among pts with
shorter (≤12 mo) vs longer (>12 mo) MF disease duration (odds ratio, 2.1; P=0.022).
Image:
Summary/Conclusion: These findings suggest that earlier RUX initiation in MF may improve
clinical outcomes, including cytopenias, SVR, symptom burden, and OS. While “watch
and wait” remains a common treatment approach for newly diagnosed patients, these
data suggest that pts with MF may benefit from earlier intervention. Additional studies
to further evaluate the impact of early intervention are warranted.
P1038: CLINICOPATHOLOGIC AND MOLECULAR CORRELATES OF ORGAN DAMAGE ACROSS THE SPECTRUM
OF ADVANCED SYSTEMIC MASTOCYTOSIS
E. Liang1,*, C. Perkins2, R. Lu3, H. Shi4, S. Dimitrijevic5, G. Hoehn4, J. Abuel2,
C. Langford2, P. Abidi2, L. Fechter2, W. Shomali2, J. Gotlib2
1Department of Medicine, Stanford University School of Medicine; 2Division of Hematology,
Stanford Cancer Institute/Stanford University School of Medicine; 3Quantitative Sciences
Unit, Stanford University School of Medicine, Stanford, CA; 4Blueprint Medicines Corporation,
Cambridge, MA, United States of America; 5Blueprint Medicines Corporation, Zug, Switzerland
Background: Advanced SM (AdvSM) comprises 3 subtypes: aggressive SM (ASM), SM with
an associated hematologic neoplasm (SM-AHN), and mast cell leukemia (MCL). The patterns
of organ damage across the spectrum of AdvSM have not been well characterized. The
definition of hematologic and non-hematologic organ damage has evolved from World
Health Organization (WHO) C-findings, to organ damage criteria specified by the International
Working Group-Myeloproliferative Neoplasms Research and Treatment and European Competence
Network on Mastocytosis (IWG). More recently, modified IWG (mIWG) criteria have been
used to define eligible organ damage in trials of AdvSM.
Aims: 1) To describe the distribution of organ damage in AdvSM patients (pts) at initial
presentation or at time of trial enrollment using WHO, IWG, and mIWG criteria; 2)
to evaluate clinicopathologic and molecular correlates of organ damage.
Methods: Our cohort included 244 AdvSM pts: 68 pts from our Stanford IRB-approved
MPN registry and 176 pts from the phase I EXPLORER (NCT02561988) and phase II PATHFINDER
(NCT03580655) studies of avapritinib in AdvSM. Comparisons between continuous and
categorial variables were performed using the Wilcoxon rank sum test and the Fisher’s
exact test, respectively. The Benjamini and Hochberg method was used to control the
false discovery rate in multiple comparisons.
Results: Median age at diagnosis was 68 years (range, 24-88) and 146 pts (60%) were
male. Forty-one pts (17%) had ASM, 165 (68%) had SM-AHN, and 38 (16%) had MCL. The
median number of prior anti-neoplastic therapies was 1 (range, 0-6), with 87 pts (36%)
previously treated with midostaurin.
Comparison of AdvSM Subtypes: Pts with MCL had the highest transfusion requirement,
with 13% and 8% of pts requiring RBC and platelet transfusions, respectively. SM-AHN
pts had the highest median alkaline phosphatase (ALP) (214 U/L), while pts with MCL
had the largest median liver (2860 cm3) and spleen (1120 cm3) volumes by imaging.
Among the two largest SM-AHN subtypes, pts with SM-CMML (n = 72) had a lower ANC (p
= 0.02) and tryptase levels (p = 0.04) than pts with MDS/MPN-U (n = 36). Pts with
SM-AHN fulfilled the WHO thrombocytopenia criterion (<100 x 109/L) more frequently
than MCL and ASM pts, and more WHO hematologic (p = 0.02) and IWG non-hematologic
(p = 0.008) criteria than ASM pts.
Prior Therapy: Pts who received ≥1 prior anti-neoplastic therapy (compared to none)
had greater tryptase levels (p = 0.02) and bone marrow (BM) MC burden (p = 0.03),
lower ALP (p = 0.04), and higher likelihood of meeting WHO malabsorption criteria
(hypoalbuminemia and/or weight loss) (p = 0.03). Compared to midostaurin-naïve pts,
those who had received midostaurin had a greater BM MC burden (p = 0.045), but less
frequently met mIWG splenomegaly criteria (p = 0.04) and met fewer WHO non-hematologic
criteria (p < 0.001).
Mutation Profiles: Compared to pts with KIT D816V only, pts who also had ≥1 SRSF2,
ASXL1, or RUNX1 mutations had a lower median hemoglobin (p = 0.02), platelet count
(p = 0.02), and greater direct bilirubin (p = 0.005), ALP (p = 0.04), and spleen volume
(p = 0.06). They were more likely to fulfill the WHO hypersplenism criterion (p =
0.03) and exhibited more WHO-defined cytopenias (p = 0.006).
Image:
Summary/Conclusion: In AdvSM, patterns of organ damage reflect disease subtype, prior
therapy, and mutation profile beyond KIT D816V (Figure). These data can help inform
the clinical heterogeneity of AdvSM patients who are being evaluated for clinical
trials which currently require 1 or more organ damage findings defined by the WHO
or (m)IWG criteria.
P1039: PALPABLE SPLEEN SIZE IS PROGNOSTIC IN PRIMARY BUT NOT SECONDARY MYELOFIBROSIS
M. Lucijanic1,*, I. Krecak2, E. Soric1, D. Galusic3, H. Holik4, V. Perisa5, M. Moric
Peric6, I. Zekanovic6, R. Kusec1
1Hematology Department, University hospital Dubrava, Zagreb; 2Internal medicine department,
General Hospital Sibenik-Knin county, Sibenik; 3Internal medicine department, University
hospital center Split, Split; 4Internal medicine department, Slavnoski Brod, General
hospital Slavonski Brod; 5Hematology Department, University hospital center Osijek,
Osijek; 6Internal medicine department, General hospital Zadar, Zadar, Croatia
Background: Splenomegaly is a clinical hallmark of primary (PMF) and secondary myelofibrosis
(SMF). Although not a part of disease specific risk scores, spleen size represents
disease burden and correlates with unfavorable disease features. Spleen size reduction
is a function of baseline spleen size and was recently shown to be independent prognostic
factor in composite PMF and SMF cohort of ruxolitinib treated patients.
Aims: To estimate prognostic properties of palpable splenomegaly and potential differences
between PMF and SMF patients.
Methods: We analyzed a multicentric cohort of 191 patients from 6 Croatian centers
with either PMF or post polycythemia vera (PV) and essential thrombocythemia (ET)
SMF diagnosed in period from 2004-2021 who had available data on palpable spleen size.
Diagnoses were established according to 2016 and 2008 criteria. Risk was stratified
according to the DIPSS in PMF and according to the MySEC-PM in SMF patients. Palpable
spleen size was assessed at the time of diagnosis or referral and stratified in three
categories: non-palpable, <10 cm and ≥10 cm.
Results: A total of 191 patients were analyzed, among them 144 PMF (63 early, 81 overt
fibrotic) and 47 SMF (27 post PV; 20 post ET). Median age was 67 years, there were
64% males. Intermediate-2 or high risk disease was present in 45% PMF and 49% SMF
patients. Non-palpable spleen, <10 cm and ≥10 cm splenomegaly were present in 31%,
43% and 26% PMF patients, respectively, and 23%, 49% and 28% SMF patients, respectively.
Palpable spleen size was similar between patients with PMF and SMF (median 3 vs 4 cm,
P=0.325). Patients with palpable spleen ≥10 cm were significantly grouped among patients
with higher risk disease in PMF (P=0.026) but not SMF (P=0.654). Degree of bone marrow
fibrosis was significantly associated with higher spleen size in PMF (P=0.008) but
not SMF (0.809). On the contrary, JAK2 mutational status had no significant association
with spleen size in PMF (P=0.847) but was significantly associated with higher spleen
size in SMF (P=0.035).
Regarding overall survival, larger spleen size was significantly associated with shorter
overall survival in PMF (P<0.001) but not SMF (P=0.825) as shown in Figure. Time to
thrombosis had no significant association with larger spleen size in neither PMF nor
SMF (P>0.05). Time to bleeding was significantly shorter in PMF patients with both
<10 cm and ≥10 cm in comparison to non-palpable spleen (P<0.05) whereas palpable spleen
size had no significant association with in SMF patients (P>0.05).
Image:
Summary/Conclusion: Various clinical features are differently associated with palpable
spleen size in PMF and SMF patients. Larger spleen size is prognostic of worse survival
and higher bleeding risk in PMF but not SMF patients. This might have repercussions
on prognostic scores based on palpable spleen size that might differently perform
in PMF and SMF patients.
P1040: HIGHER ESTIMATED PLASMA VOLUME STATUS IS ASSOCIATED WITH INCREASED THROMBOTIC
RISK AND IMPAIRED SURVIVAL IN PATIENTS WITH PRIMARY MYELOFIBROSIS
M. Lucijanic1,*, I. Krecak2, E. Soric1, D. Galusic3, H. Holik4, V. Perisa5, M. Moric
Peric6, I. Zekanovic6, R. Kusec1
1Hematology Department, University hospital Dubrava, Zagreb; 2Internal medicine Department,
General hospital Sibenik-Knin county, Sibenik; 3Hematology Department, University
hospital center Split, Split; 4Internal medicine Department, General hospital Slavonski
Brod, Slavonski Brod; 5Hematology Department, University hospital center Osijek, Osijek;
6Internal medicine Department, General hospital Zadar, Zadar, Croatia
Background: Blood plasma experiences substantial changes in both volume and composition
in patients with chronic myeloproliferative neoplasms (MPN) and represents large reservoir
of cytokines and other mediators of inflammation. Higher estimated plasma volume status
(ePVS) has been shown to correlate with increased thrombotic risk in polycythemia
vera patients, but its clinical and prognostic associations are unknown in patients
with other MPN subsets.
Aims: To estimate clinical and prognostic associations of ePVS in patients with primary
myelofibrosis (PMF).
Methods: We retrospectively analyzed a cohort of 167 patients with myelofibrosis treated
in 6 Croatian hematology centers in period from 2004-2021. Diagnosis was established
according to the WHO 2016 criteria. ePVS was calculated using the Strauss derived
Duarte formula: 100-hematocrit (%)/hemoglobin (g/dL) and expressed as dl/g. ePVS associations
with baseline clinical parameters and its prognostic associations with overall survival
(OS), time to thrombosis (TTT) and time to bleeding (TTB) were analyzed.
Results: There were 167 patients with PMF analyzed (72 pre-fibrotic and 95 overt fibrotic).
Median age was 68 years, there were 65% males, 67% were JAK2 mutated. Intermediate-2
or high DIPSS risk was present in 48% of patients.
Median ePVS was 5.62 dl/g. Higher ePVS was significantly associated with higher degree
of bone marrow fibrosis, absence of JAK2 mutation, presence of constitutional symptoms,
presence of circulating blasts, transfusion dependency, larger palpable spleen size,
higher Charlson comorbidity index, history of bleeding, lower WBC, lower platelet
count, lower hemoglobin, higher RDW, higher LDH, higher CRP, higher ferritin, lower
albumin, higher fibrinogen, higher risk DIPSS status (P<0.05 for all analyses).
Median follow-up of our cohort was 57 months. During follow-up period a total of 79
patients died, 18 experienced thrombotic event (14 arterial and 5 venous thromboses)
and 14 experienced bleeding event. Higher ePVS stratified at the ROC curve analysis
defined cut-off points was significantly associated with worse OS (ePVS>6.2 dl/g,
HR 1.74, P=0.017) and shorter TTT (ePVS>6.6 dl/g, HR 2.69, P=0.047) as shown in Figure.
There was no significant association with TTB. In a multivariate Cox regression analysis
model association of higher ePVS with increased thrombotic risk (HR 3.22, P=0.019)
was present independently of WBC (HR 1.02, P=0.003), whereas JAK2 mutational status
(HR 2.74, P=0.080) and cardiovascular risk factors (HR 7.12, P=0.060) retained marginal
significance. Association with OS diminished in multivariate analysis after adjustment
for DIPSS.
Image:
Summary/Conclusion: PMF patients with more advanced disease features (more pronounced
cytopenias and splenomegaly) have higher ePVS indicative of expanded plasma volume.
Higher ePVS is associated with higher thrombotic risk and impaired survival.
P1041: IMPACT OF FEDRATINIB ON SPLEEN VOLUME AND MYELOFIBROSIS SYMPTOMS IN PATIENTS
WITH SUBSTANTIAL SPLENOMEGALY: POST HOC ANALYSES FROM THE JAKARTA AND JAKARTA2 TRIALS
J.-J. Kiladjian1,*, A. Tefferi2, F. Passamonti3, A. Vannucchi4, M. Talpaz5, F. Cervantes6,
C. N. Harrison7, R. A. Mesa8, J. Mascarenhas9, N. Schaap10, S. Verstovsek11, T. Devos12,
S. Rose13, J. Zhang13, O. Sy13, A. Pardanani2
1Hôpital Saint-Louis; Université de Paris, INSERM, Paris, France; 2Mayo Clinic of
Rochester, Rochester, United States of America; 3University of Insubria, Varese; 4CRIMM,
University of Florence, AOU Careggi, Florence, Italy; 5University of Michigan Comprehensive
Cancer Center, Ann Arbor, United States of America; 6Hospital Clinic Provincial de
Barcelona, Barcelona, Spain; 7Guy’s and St Thomas’ Hospital, London, United Kingdom;
8Mays Cancer Center at UT Health San Antonio MD Anderson Cancer Center, San Antonio;
9Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, United
States of America; 10Radboud University Medical Centre, Nijmegen, Netherlands; 11The
University of Texas MD Anderson Cancer Center, Houston, United States of America;
12University Hospitals Leuven and Laboratory of Molecular Immunology (Rega Institute),
KU Leuven, Leuven, Belgium; 13Bristol Myers Squibb, Princeton, United States of America
Background: In myelofibrosis (MF), splenomegaly is a marker of disease progression,
and larger spleen size (SS) is correlated with poorer survival and debilitating symptoms.
Fedratinib (FEDR) is an oral, selective Janus kinase 2 (JAK2) inhibitor approved as
first-line (1L) treatment (Tx) of patients (pts) with MF and in pts previously treated
with ruxolitinib (RUX). In the phase 3, placebo-controlled JAKARTA trial (NCT01437787)
and the phase 2, single-arm JAKARTA2 trial (NCT01523171), pts experienced substantial
improvements in spleen volume and MF symptoms after receiving 6 FEDR Tx cycles. Given
the prognostic impact of splenomegaly, these analyses explore whether these clinical
benefits were influenced by pre-Tx SS.
Aims: Investigate the efficacy of FEDR 400 mg/d in JAKARTA and JAKARTA2, in pt subgroups
defined by baseline (BL) SS.
Methods: The JAKARTA trial assessed FEDR 400 mg/d, FEDR 500 mg/d, and placebo in pts
with JAK inhibitor-naïve MF, and the JAKARTA2 trial evaluated FEDR 400 mg/d (starting
dose) in pts resistant/intolerant to prior RUX. Both studies enrolled pts with intermediate-
or high-risk MF, SS >5 cm from the left costal margin by palpation, platelets ≥50×109/L,
and ECOG PS ≤2. These post hoc analyses included pts from both studies who were allocated
to receive FEDR 400 mg/d (the approved dosage) in continuous 28-d Tx cycles.
Pt subgroups were divided according to BL median SS (mSS) in each trial. Changes from
BL in spleen volume and Myelofibrosis Symptom Assessment Form (MFSAF) total symptom
score (TSS) were measured at the end of cycle 6 (EOC6). Spleen volume was assessed
by MRI/CT scan; MFSAF TSS was the sum of 6 MF symptom scores: night sweats, early
satiety, pruritus, pain under ribs-left side, abdominal discomfort, and bone/muscle
pain. Eligible pts must have had spleen volume and/or TSS data at BL and EOC6.
Results: Overall, 96 pts in JAKARTA and 97 pts in JAKARTA2 received FEDR 400 mg/d;
mSS (range) at BL was 16 (5–40) cm and 18 (5–36) cm, respectively. In the JAKARTA
<mSS and ≥mSS cohorts, median BL spleen volume was 1771 (316–3396) mL and 3329 (983–6430)
mL, respectively, and median TSS was 13.0 (0.0–54.9) and 17.2 (0.4–57.0). In JAKARTA2,
median BL spleen volume was 1937 (737–4305) mL and 4002 (1821–7815) mL for pts with
<mSS and ≥mSS, respectively; BL TSS was 17.2 (0.7–48.0) and 23.6 (1.0–44.0).
Overall, 75 (78%) pts in JAKARTA and 51 (53%) pts in JAKARTA2 had spleen volume data
available at BL and EOC6; the <mSS and ≥mSS cohorts, comprised 37 and 38 pts, respectively,
in JAKARTA, and 31 and 20 pts in JAKARTA2. Median spleen volume reduction (SVR) from
BL was similar between the <mSS and ≥mSS cohorts in each trial, and 97% of pts with
≥mSS in both trials achieved some degree of SVR. In JAKARTA, median SVR at EOC6 was
−38% (95% CI, −43 to −31) in the <mSS and −40% (95% CI, −40 to −28) in the ≥mSS cohort;
in JAKARTA2, median SVR was −37 (95% CI, −43 to −30) in the <mSS and −38.5% (95% CI,
−49 to −14) in the ≥mSS cohort.
TSS data were available at BL and EOC6 for 71 (74%) pts in JAKARTA and 51 (53%) pts
in JAKARTA2. Changes from BL in TSS at EOC6 were similar in the JAKARTA <mSS and ≥mSS
cohorts, with median TSS reductions of −49% and −51%, respectively. Median TSS changes
at EOC6 in JAKARTA2 were greater for pts with larger spleens, −36% and −49% in the
<mSS and ≥mSS cohorts, respectively.
Image:
Summary/Conclusion: Pts who completed 6 Tx cycles with FEDR 400 mg/d, whether used
as 1L MF Tx or after RUX Tx, experienced substantial improvements in spleen volume
and MF symptom severity regardless of the extent of splenomegaly at BL.
P1042: SAFETY AND TOLERABILITY RESULTS FROM THE PHASE 3B FREEDOM TRIAL OF FEDRATINIB
(FEDR), AN ORAL, SELECTIVE JAK2 INHIBITOR, IN PATIENTS WITH MYELOFIBROSIS (MF) PREVIOUSLY
TREATED WITH RUXOLITINIB (RUX)
V. Gupta1,*, A. Yacoub2, S. Verstovsek3, R. A. Mesa4, C. N. Harrison5, G. Barosi6,
J.-J. Kiladjian7, H. J. Deeg8, S. Fazal9, L. Foltz10, R. J. Mattison11, C. B. Miller12,
V. Parameswaran13, C. Hernandez14, J. Zhang14, M. Talpaz15
1Princess Margaret Cancer Centre, Toronto, Canada; 2University of Kansas Medical Center,
Kansas City; 3The University of Texas MD Anderson Cancer Center, Houston; 4Mays Cancer
Center at UT Health San Antonio MD Anderson Cancer Center, San Antonio, United States
of America; 5Guy’s and St Thomas’ Hospital, London, United Kingdom; 6Fondazione IRCCS
Policlinico San Matteo, Pavia, Italy; 7Hôpital Saint-Louis, Université de Paris, INSERM,
Paris, France; 8Fred Hutchinson Cancer Center, Seattle; 9Allegheny Health Network
Cancer Institute, Pittsburgh, United States of America; 10St. Paul’s Hospital, University
of British Columbia, Vancouver, Canada; 11University of Wisconsin Carbone Comprehensive
Cancer Center, Madison; 12Ascension Saint Agnes Hospital, Baltimore; 13Avera Cancer
Institute, Sioux Falls; 14Bristol Myers Squibb, Princeton; 15University of Michigan
Comprehensive Cancer Center, Ann Arbor, United States of America
Background: FEDR is an oral, selective Janus kinase 2 (JAK2) inhibitor approved for
treatment (Tx) of patients (pts) with MF, including those previously treated with
RUX. In the single-arm, phase 2 JAKARTA2 trial in pts with MF resistant/intolerant
to prior RUX, the most common adverse events (AEs) during FEDR Tx were gastrointestinal
(GI) events. JAKARTA2 ended in 2013, when a temporary clinical hold was placed on
FEDR due to suspected cases of Wernicke’s encephalopathy (WE). The safety and efficacy
of FEDR in pts with MF previously treated with RUX are being further evaluated in
the ongoing, single-arm, phase 3b FREEDOM trial (NCT03755518), which includes prospective
strategies for preventing or mitigating GI AEs, thiamine decreases, and potential
WE.
Aims: To assess the safety of FEDR and the effectiveness of AE mitigation strategies
in the FREEDOM trial.
Methods: Eligible pts had intermediate- or high-risk MF, platelets ≥50×109/L, and
spleen volume ≥450cm3 by MRI/CT or palpable spleen ≥5 cm below the left costal margin.
Pts must have received prior RUX for ≥3 mo, or for ≥28 d with development of red blood
cell transfusion requirement (≥2 units/mo for 2 mo) or grade ≥3 thrombocytopenia,
anemia, hematoma, or hemorrhage. All pts received FEDR 400 mg/d in continuous 28-d
cycles. AE mitigation strategies include prophylactic and symptomatic use of anti-nausea/vomiting
and anti-diarrheal Tx, thiamine supplementation, FEDR dosing modifications, and administration
of FEDR with food.
Results: In all, 34 pts were enrolled and 16 pts continued to receive FEDR at data
cutoff (April 9, 2021). Reasons for FEDR discontinuation (D/C) in >1 pt were lack
of efficacy (n=5), AEs (n=4), disease progression (n=2), pt decision (n=2), and to
undergo transplant (n=2). Median FEDR Tx duration at cutoff was 28.3 (range, 1.6–101.3)
wk; 14 (41%) pts had completed >12 cycles. At baseline (BL), median (range) age was
68.5 (49–82) y, time from MF diagnosis was 3.4 (0.1–17.4) y, and spleen size was 15
(3–31) cm. Most pts (62%) had primary MF, and all pts had received ≥3 mo of prior
RUX; the most common reason for RUX D/C was loss of response/Tx failure (41%).
During FEDR Tx, 22 (65%) pts received ondansetron and 11 (32%) pts received loperamide.
GI AEs in >10% of pts were constipation (47%), diarrhea (35%), nausea (26%), abdominal
pain (24%), and vomiting (18%). Nausea, vomiting, and diarrhea decreased as Tx continued.
Most GI AEs were grade (G) 1/2 (including nausea, vomiting, and diarrhea) and there
were no Tx-related G3/4 GI AEs. No pt required FEDR reduction, interruption, or D/C
due to a Tx-related GI AE.
Tx-related G3/4 AEs were reported in 11 (32%) pts, including anemia in 7 (21%) pts,
and neutropenia, thrombocytopenia, and hyperkalemia in 2 (6%) pts each.
At BL, 1 pt had thiamine below the 70 nmol/L lower limit of normal (LLN), which normalized
before the pt received FEDR. During FEDR Tx, thiamine levels dropped below the LLN
for 4 pts between cycles 2 and 3 (and for 1 pt at end of Tx); for these pts, levels
returned to normal with thiamine supplementation and no FEDR interruption or reduction
was required (Figure). Five other pts received prophylactic thiamine supplementation.
There were no cases of WE.
Image:
Summary/Conclusion: FEDR was generally well tolerated in pts with MF previously treated
with RUX. The frequency and severity of GI AEs were substantially lower in FREEDOM
than in previous FEDR clinical trials, likely due to early implementation of GI prophylaxis.
Thiamine decreases were uncommon and easily managed with routine monitoring and oral
supplementation as needed.
P1043: OUTCOME OF PATIENTS WITH PREFIBROTIC MYELOFIBROSIS FROM A LARGE ACADEMIC CENTER
L. Masarova1,*, P. Bose1, N. Pemmaraju1, H. Chifotides1, L. Zhou1, Z. Estrov1, H.
Kantarjian1, S. Verstovsek1
1Leukemia, MD Anderson Cancer Center, Houston, United States of America
Background: The 2016 WHO classification includes prefibrotic primary myelofibrosis
(pre-PMF; fibrosis grading 0-1) and overt PMF (fibrosis grading 2–3). Pre-PMF has
favorable survival when compared to overt PMF, however pre-PMF patients with intermediate
-2 or high risks IPSS still have poor outcome (Guglielmelli et al. Blood. 2017;129(24):3227–3236).
Aims: We aimed to characterize pre-PMF patients referred to our institution between
the years of 2000-2020. We specifically investigated the role of known IPSS prognostic
factors separated into i) clinical factors (CF): presence of either hemoglobin [hgb]
< 10 g/dL / white cells [WBC] > 25 x109/L] or peripheral blasts [bl] ≥1%]; ii) age
> 65 years and iii) systemic symptoms (IPSS defined; Cervantes, et al. Blood. 2009;113(13):2895–901).
Methods: This retrospective study included 130 patients with newly diagnosed pre-PMF
(median 1.5 months from diagnosis to presentation) and available bone marrow fibrosis
grading (Thiele, J. et al. Haematologica. 2005. 90, 1128–1132). Demographics were
expressed by descriptive statistics. Overall survival was estimated from the time
of presentation using Kaplan Meier method with log rank test.
Results: Clinical characteristics are detailed in Table 1, PART I. Fibrosis degree
included grade 0 in 9, grade 0/1 in 7 and grade 1 in 114 patients, respectively. IPSS
risks were as follows: low (N 22), intermediate 1 (N 50), intermediate 2 (N 30) and
high (N 28). Thirty-five patients had no i-iii) factors (27%), symptoms only were
in 49 patients (38%), one CF without and with symptoms was noticed in 9 (7%) and 22
(17%) patients, respectively, and 15 (11%) patients had 2 CF +/- symptoms.
Median OS of the entire cohort was 68 months (95% CI 42-94) with 54% of patients being
alive at 5 years. Overall survival (95% CI) per IPSS scores: low-intermediate 1 and
2 - high were not reached, 179 months (65-not expressed), 65 months (32-98) and 27
months (5-49), respectively. Median OS (95% CI) of patients without i-iii) factors
(age < 65 years, no symptoms and no CF) and patients with only symptoms (all ages
included) was not reached; median OS of patients with one CF with and without symptoms
was of 48 months (33-63) and 47 months (37-57), respectively and median OS of patients
with 2 CF +/- symptoms was of 16 months (8-24). Detailed separation of survivals per
age categories and IPSS subgroups is provided in Table 1, PART II.
Over the median follow-up of 43 months (95% CI 28-59), 7 patients progressed to acute
leukemia with time to progression of 40 months (95% CI 9-178). Forty-one percent of
patients (n = 53) required MF therapy during their follow-up, including 35 patients
with JAK inhibitors and 8 with stem cell transplantation, respectively. Patients without
the need for therapy or those who received JAK inhibitor had median OS of 81 (95%
CI 65-not estimated) and 100 months (95% CI 17-183), respectively. This was superior
to treated patients without exposure to JAK inhibitors (median OS of 45 months, 95%
CI 16-47).
Image:
Summary/Conclusion: Presence of more adverse clinical factors is the strongest predictor
of unfavorable outcome of patients with pre-PMF. Use of JAK inhibitors might have
significantly improved the outcome of patients with constitutional symptoms.
P1044: REAL-WORLD RUXOLITINIB TREATMENT PATTERN IN MYELOFIBROSIS PATIENTS WITH THROMBOCYTOPENIA
J. Mei*1,, Y. Wang*2, S. Kabir1, N. Ichikawa3, S. Iino3, C. Lebedinsky1
1Clinical Development, Sumitomo Dainippon Pharma Oncology, Inc, Cambridge; 2Computational
Research, Sumitovant Biopharma, New York, United States of America; 3Oncology Clinical
Development Unit, Sumitomo Dainippon Pharma Co., Ltd, Osaka, Japan
Background: Ruxolitinib (RUX) is the standard treatment for patients (pts) with myelofibrosis
(MF) to reduce splenomegaly and improve MF-associated symptoms. RUX treatment is titrated
based on platelet counts and is associated with high rates of cytopenia effect. The
incidence of moderate thrombocytopenia (platelet count <100 x 109/L) is about 25%
in newly diagnosed MF pts; however, the prevalence is about 67% in all MF pts. Low
platelet count has been identified as an important surrogate marker of survival in
MF pts with associated poor clinical features. Thrombocytopenia is also an independent
predictor in the Dynamic International Prognostic Scoring System plus (DIPPS plus),
with an actual 1.4-fold increased risk of death. Thus, treatment of MF pts with thrombocytopenia
confers a unique challenge and unmet medical need. Real world studies will provide
a perspective on the current management of MF pts with thrombocytopenia.
Aims: To characterize the disease and RUX treatment pattern in MF pts with thrombocytopenia
using de-identified insurance claims data in the US.
Methods: The IBM MarketScan® Medicare Supplemental Database data was retrospectively
analyzed to identify pts aged ≥18 years with ≥1 claim for RUX and ≥2 non-diagnostic
medical claims for MF (ICD-10 D47.4, D7581) from 2017–2019. The first RUX claim on
or after the first MF claim defined the index date. Pts who continuously enrolled
in a health plan for 3 months before the index date and 6 months after the index date
were included. Clinical characteristics and RUX treatments were compared in MF pts
with or without a history of thrombocytopenia prior to RUX initiation.
Results: The database contained 136 MF pts who had received treatment with RUX. Median
age was 61 years, 61% were male, 62% of pts had primary MF, and 38% had post-polycythemia
vera or essential thrombocythemia MF. Prior to initial RUX treatment, 57% of pts had
a diagnosis of splenomegaly, 56% had anemia, 25% had thrombocytopenia, 19% had received
RBC transfusion, and 7% had neutropenia. During RUX treatment, 25% of pts had a new
diagnosis of thrombocytopenia, 22% had neutropenia, and 19% had anemia. Pts with a
history of thrombocytopenia also had more anemia (79% vs 48%, p=0.001) and more RBC
transfusions (44% vs 11%, p<0.001) than those without a history of thrombocytopenia.
History of thrombocytopenia was associated with a lower starting dose of RUX (average
starting daily dose 20 mg vs 27 mg, p=0.005), whereas history of anemia, leukocytosis,
neutropenia, MF type, and splenomegaly were found to have no difference in the RUX
starting dose. In addition, MF pts with history of thrombocytopenia continued RUX
treatment at lower doses compared with those without a history of thrombocytopenia.
During RUX treatment, 57% of pts with history of thrombocytopenia experienced anemia
compared with 42% in pts without history of thrombocytopenia (OR 1.88, p=0.43).
Summary/Conclusion: In this real-world data analysis, history of thrombocytopenia
was a major factor influencing the RUX dose for treatment of MF pts, consistent with
prescribing data that RUX dose is adjusted based on platelet counts. These data show
that pts with thrombocytopenia were treated with lower doses of RUX in clinical practice.
Additionally, history of thrombocytopenia was associated with history of anemia; thus,
these MF pts may be more prone to develop anemia during RUX treatment compared with
pts without a history of thrombocytopenia. Our findings underscore the need for more
treatment options to manage MF pts with thrombocytopenia.
*Equal contribution.
P1045: IS JAK2V617F BUT NOT CALR AS DRIVER MUTATION ENOUGH BY ITSELF IN THE PATHOGENESIS
OF UNUSUAL TYPE VENOUS THROMBOSIS IN MPN PATIENTS?
E. Morsia1,*, E. Torre1, G. Tarantino2, G. Svegliati Baroni2, G. Goteri3, S. Mancini1,
A. Tassoni1, F. Alessandro1, A. Olivieri1, S. Rupoli1
1Hematology; 2Gastroenterology and hepatology; 3Pathological Anatomy and Histopathology,
Ospedali Riuniti di Ancona, Ancona, Italy
Background: Myeloproliferative neoplasms (MPN) are a heterogenous group of hematopoietic
stem cell disorders and clinically they constitute the most frequent underlying cause
of venous thrombosis (VTE) in unusual site, including splanchnic vein thrombosis (SVT)
and cerebral vein thrombosis (CVT). This subgroup of patients has been showed to have
distinct characteristics compared to MPN patients with VTE in usual site, including
deep vein thrombosis and pulmonary embolism.
Aims: The aim of this study is analyzed in a retrospective cohort of patients with
MPN clinical characteristics, molecular features and outcome data in patients who
have experienced an unusual site thrombosis.
Methods: In our study, we retrospectively analyzed a cohort of 577 consecutive patients
with MPN according to WHO 2016 criteria who referred to our institute between 2009
and 2020. In all, 113 patients (19.58%) had a vascular event during a median follow
up of 94.3 months (range, 2.4-416.0). Usual and unusual site venous thrombosis occurred
in 40.7% and 33.6% in MPN patients with thrombosis, respectively.
Results: From the 38 MPN patients with unusual site VTE, 19 (50%) were male. Twenty-seven
patients had an SVT and 11 a CVT. The driven mutation was JAK2V617F in 79% with a
median allelic burden of 20% using NGS. MPN patients with SVT and CVT are younger
(ORR 0.87 (0.82-0.93), pValue < 0.0001), with higher PLT count at diagnosis (ORR 0.06
(0.01-0.38), pValue=0.0085), and higher splenomegaly rate (ORR 0.97 (0.90-0.99), pValue=0.0003)
compared to MPN patients with usual site VTE. Globally survival data in our population
reported an overall survival in MPN with thrombosis of 238 months (95% I.C. 177-336),
poorer in patients with arteriosus events compared to VTE (usual and unusual sites;
pValue=0.017). At all, only few patients experienced recurrence thrombosis during
the follow up and the recurrence thrombosis free survival rate at 5 years in MPN with
thrombosis was 85%. Moreover, older age and level of hemoglobin at time of thrombosis
significantly influence survival in MPN patients with unusual site VTE. Using NGS
on 20 MPN patients with unusual site VTE, molecular analysis identified the mutation
in the hotspot exon 14 region of JAK2 in 13 patients, among them the 92% as isolated
alteration. The most frequent additional mutations were found in CALR mutated patients,
including high risk mutation as ASXL1 and U2AF1 (Figure 1).
Image:
Summary/Conclusion: This finding underlies the central role of JAK2V617F mutation
in the pathogenesis of unusual type VTE in MPN, which occurs as sole molecular alteration
in more than 90% of cases. However, in our cohort most patients who carried CALR as
driver mutation seem to need one or more additional mutations. Our interpretation
on these findings is that JAK2V617F is per se a strong risk factor for unusual venous
thrombosis while CALR mutation must be enriched for additional non-driver alterations,
needing a second hit to drive to thrombosis.
P1046: SEVERE COMPLICATIONS IN JAK2 V617F POSITIVE PEDIATRIC PATIENTS WITH MYELOPROLIFERATIVE
NEOPLASMS
W. Novak1,*, C. Annamária1, R. Crazzolara2, J. Neil3, K. Pirolt4, M. Dworzak1,5, L.
Kager1,5
1St. Anna Children’s Hospital, Department of Pediatrics, Medical University Vienna,
Vienna; 2Department of Pediatrics, Medical University of Innsbruck, Innsbruck; 3Department
of Pediatrics, Paracelsus Medical University, Salzburg; 4Department of Pediatrics,
Klinikum Klagenfurt, Klagenfurt; 5St. Anna Children’s Cancer Research Institute (CCRI),
Vienna, Austria
Background: Myeloproliferative neoplasms (MPNs) are very rare diseases in children
and adolescents and neither detailed risk stratification nor treatment guidelines
are available.
Aims: The aim was to evaluate disease characteristics, complications, and treatment
response of pediatric patients diagnosed with MPNs.
Methods: We retrospectively analyzed clinical data from a cohort of 14 pediatric patients
diagnosed and treated in Austria over the past 12 years.
Results: Of these 14 patients (female, N=9), 10 patients were diagnosed with essential
thrombocythemia (ET), three patients with polycythemia vera (PV) and one patient with
primary myelofibrosis (PMF). One of the three main driver mutations known to cause
MPN (JAK2, MPL or CALR mutation) was found in nine patients, eight of whom carrying
a JAK2 V617F mutation (ET, N=5; PV, N=3; female, N=5; male, N= 3) and the patient
with PMF harbored a CALR type 1 mutation.
Thrombotic complications occurred in 3/5 female (0/3 male) JAK2 V617F positive patients;
one patient with ET developed recurrent sinus vein thrombosis (SVT) and two patients
with PV developed a Budd-Chiari syndrome (BCS). One patient with BCS died. A transformation
into a pre-PMF was observed in the other patient with BCS. In addition, another JAK2
V617F positive patient showed transformation from ET to PV seven years after diagnosis.
Although 7/11 tested patients developed an acquired von Willebrand syndrome, there
were no major bleeding complications. Two symptomatic JAK2 V617F positive female ET
patients were treated off-label with ropeginterferon alfa-2b. The indications for
treatment were microvascular symptoms (headache) and recurrent SVT. To our knowledge,
this new long-acting interferon has not previously been used in children and adolescents.
The treatment was well tolerated, no complications occurred and both patients quickly
resolved their clinical symptoms.
Summary/Conclusion: Our data support results from studies in adult MPN patient populations,
which showed that JAK2 V617F positivity is a risk factor for thrombotic complications
and female gender is an additional risk factor for atypical thrombosis (BCS and SVT).
P1047: REAL-WORLD SAFETY OF RUXOLITINIB IN PATIENTS WITH INTERMEDIATE OR HIGH RISK
OF PRIMARY MYELOFIBROSIS, POST-POLYCYTHEMIA VERA MYELOFIBROSIS OR POST-ESSENTIAL THROMBOCYTHEMIA
MYELOFIBROSIS IN CHINA
Z. Xu1, M. Duan2, Q. Jiang3, Q. Leng4, N. Xu5, Y. Zhang6, C. Zhao7, W. Wu8, Q. Zhang9,
J. Fu10, J. Zhang11, R. Fu12, Z. Yan13, J. Zhang14, C. Lin15, G. Ouyang16, Z. Wang17,
L. Ma18, H. Hao19, X. Li20, S. Ran21, Y. Chen22, T. Li23, Z. Xiao1,*
1State Key Laboratory of Experimental Hematology, National Clinical Research Center
for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy
of Medical Sciences & Peking Union Medical College, Tianjin; 2Peking Union Medical
College Hospital; 3Peking University People’s Hospital, Beijing; 4Anshan Central Hospital,
Liaoning; 5Nanfang Hospital, Southern Medical University, Guangdong; 6Henan Provincial
People’s Hospital, Henan; 7Affiliated Hospital of Qingdao University, Shandong; 8Ruijin
Hospital, Shanghai Jiao Tong University, Shanghai; 9Gansu Provincial People’s Hospital,
Gansu; 10Shaoxing People’s Hospital, Zhejiang; 11The Second Hospital of Hebei Medical
University, Hebei; 12Tianjin Medical University General Hospital, Tianjin; 13North
China University of Technology Affiliated Hospital, Hebei; 14Nanchang University Affiliated
Ganzhou Hospital, Jiangxi; 15Zhangzhou Affiliated Hospital of Fujian Medical University,
Fujian; 16Ningbo First Hospital, Zhejiang; 17Wuxi Second People’s Hospital, Jiangsu;
18Sun Yat Sen Memorial Hospital of Sun Yat Sen University, Guangdong; 19Hebei General
Hospital, Hebei; 20Affiliated Hospital of Southwest Medical University, Sichuan; 21Beijing
Novartis Pharma Co., Ltd, Beijing; 22China Novartis Institutes For Biomedical Research
Co., Ltd; 23China Novartis Institutes For Biomedical Research Co., Ltd., Shanghai,
China
Background: Ruxolitinib, a potent and selective inhibitor of Janus kinase 1 (JAK1)/JAK2
tyrosine kinases, was approved for treatment of intermediate- and high-risk patients
with myelofibrosis (MF) based on the findings from the pivotal studies, COMFORT I
and II. Although an Asia regional study was conducted, the enrolled Chinese population
was limited (N=63).
Aims: We aimed to evaluate the safety profile and the treatment of ruxolitinib in
Chinese patients with MF under routine clinical practice.
Methods: This was a retrospective, non-interventional, multicenter, post-marketing
surveillance study collecting data up to 48 weeks after ruxolitinib initiation. Patients
aged ≥18 years with a confirmed diagnosis of intermediate or high-risk primary MF
(PMF), post-polycythemia vera MF (PPV-MF) or post-essential thrombocythemia MF (PET-MF)
who had received or were currently receiving ruxolitinib treatment per clinical judgment
according to China approved label were included.
Results: In total, 480 patients from 20 sites were enrolled and 428 patients completed
the study treatment. The most reported reasons for treatment discontinuation were
death (17.3%), adverse events (AE, 7.7%) and abnormal laboratory value (5.8%). Gender
distribution were balance (male 51.6% vs female 48.4%) with median age of 61.0 years
and median BMI of 22.5 kg/m2. Majority of the patients were diagnosed with PMF (69.8%),
followed by PPV-MF (15.7%), and PET-MF (14.5%).
The information on dosing and safety is detailed in Table 1. Ruxolitinib dosing was
according to the local approved label (median initial daily dose 30.0 mg). The median
time from first diagnosis to medication was 1.5 months. Overall, patients received
ruxolitinib for a median of 11.1 months with median dose intensity of 28.7 mg/day.
The most prescribed dose was 15 mg BID across varying durations of exposure to ruxolitinib
(up to 48 weeks). A total of 196 (40.8%) patients had dose modification.
On-treatment AE and serious AE (SAE) were reported in 361 (75.2%) and 57 (11.9%) patients,
respectively. Of those, 60.2% and 6.0%, were related to study drug, respectively.
The severity of majority of the AE were grade 2 and 3 (26.7 & 25.2%, respectively)
while SAE were grade 3 & 4 (3.5% & 4.4%, respectively). A total of 4.0% and 2.3% of
patients reported AEs and SAEs leading to discontinuation. The most commonly reported
AEs were anemia (47.3%), decreased platelet count (22.1%), and abnormal liver function
(12.7%). The most commonly reported SAEs were anemia (3.8%), platelet count decreased
(1.0%), infectious pneumonia and pulmonary inflammation (both 0.8%). On-treatment
AESI was reported by 18 (3.8%) patients, consisted of bleeding events (2.3%), serious
or opportunistic infections (0.8%), and second primary malignancies (SPM, 0.6%). The
SPM were pulmonary malignant tumors (n=2) and liver cancer (n=1). A total of 18 on-treatment
deaths (MF progression=9, AE=4, and others=5) were reported.
During the study period, we found that the number of patients with efficacy data were
decreasing with time (baseline=183&116, Week 16=126 & 96, Week 32=90 & 62, Week 48=38
& 19 for spleen size measurement and total symptom score, respectively). This reflects
the need for improve compliance in disease management.
Image:
Summary/Conclusion: This was the largest ruxolitinib real-world study in China. The
safety profile of ruxolitinib in patients with intermediate- or high-risk PMF, PPV-MF,
or PET-MF was generally consistent with previous global and Chinese studies of MF.
The incidence of SPM was lower than that reported previously4. No new unexpected safety
signals have been identified.
P1048: MYF3001: A RANDOMIZED OPEN LABEL, PHASE 3 STUDY TO EVALUATE IMETELSTAT VERSUS
BEST AVAILABLE THERAPY IN PATIENTS WITH INTERMEDIATE-2 OR HIGH-RISK MYELOFIBROSIS
REFRACTORY TO JANUS KINASE INHIBITOR
J. Mascarenhas1,*, C. N. Harrison2, J.-J. Kiladjian3, R. S. Komrokji4, S. Koschmieder5,
A. M. Vannucchi6, T. Berry7, D. Redding7, L. Sherman7, S. Dougherty7, L. Peng7, L.
Sun7, F. Huang7, Y. Wan7, F. M. Feller7, A. Rizo7, S. Verstovsek8
1Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United
States of America; 2Guy’s and St Thomas’ Hospital, London, United Kingdom; 3Hôpital
Saint-Louis, Université Paris, Paris, France; 4H Lee Moffitt Cancer Center, Tampa,
FL, United States of America; 5RWTH Aachen University, Aachen, Germany; 6AOU Careggi,
University of Florence, Florence, Italy; 7Geron Corporation, Parsippany, NJ; 8The
University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
Background: Myelofibrosis (MF) is a life-threatening myeloproliferative neoplasm.
The Janus Kinase inhibitors (JAKi) ruxolitinib and fedratinib are the only FDA approved
treatment options for MF. Despite benefits reported with ruxolitinib in the front-line
setting, a high proportion of patients discontinue treatment (Abdelrahman 2015), and
the median overall survival (OS) is 11-16 months (Kuykendall 2018; Newberry 2017;
Schain 2019; Palandri 2019; Mascarenhas 2020), highlighting a great unmet need for
pts non-responsive to a JAKi treatment. Imetelstat, a first-in-class telomerase inhibitor,
has shown meaningful clinical improvement in IMbark, a Phase 2 study in patients with
intermediate-2 or high-risk MF who have relapsed after or are refractory to JAKi (Mascarenhas
JCO 2021). Treatment with 9.4 mg/kg imetelstat resulted in 32.2% symptom response
(total symptom score [TSS] reduction ≥50%) at Week 24 and median overall survival
(OS) of 29.9 months with overall study follow up of 27.4 months. Dose-dependent inhibition
of telomerase with imetelstat resulted in on-target activity that correlated with
clinical benefits; dose-dependent reduction in variant allele frequency of MF driver
mutations indicated targeting of the underlying malignant clone.
Aims: The Phase 2 results support continued study of imetelstat 9.4 mg/kg in a Phase
3 randomized controlled study, for which the study design is described here.
Methods: Study MYF3001 (IMpactMF; NCT04576156) is an open label, randomized (2:1),
multicenter, Phase 3 study of imetelstat compared with best available therapy (BAT)
in ~320 patients with intermediate 2 or high-risk MF refractory to JAKi treatment.
Patients will be randomized to receive imetelstat 9.4 mg/kg IV every 21 days or investigator
selected BAT including hydroxyurea, thalidomide, interferon, danazol, hypomethylating
agents, chemotherapy, or other non-JAKi containing therapy as appropriate). Eligible
patients will be stratified based on a) Intermediate 2 or high-risk per Dynamic International
Prognostic Scoring System; b) platelet count at entry (platelets ≥ 75 and < 150 x
109/L vs ≥ 150 x 109/L). Patients who meet progressive disease criteria and discontinue
BAT may be eligible to crossover to imetelstat.
Results: The primary endpoint is OS and one interim analysis is planned when approximately
>71% of death events have occurred. Secondary endpoints include symptom and spleen
response rates at week 24, progression-free survival, clinical response assessment
per modified 2013 International Working Group - Myeloproliferative Neoplasms Research
and Treatment criteria, time to and duration of response, reduction in degree of bone
marrow fibrosis, safety, pharmacokinetics and patient-reported outcomes. Biomarkers
and mutation analyses will be performed to evaluate the impact of imetelstat on reduction/depletion
of malignant clones. (Figure 1)
Image:
Summary/Conclusion: Approximately 180 sites are planned in North and South America,
Europe, Middle East, Australia and Asia. The study is open for enrollment.
P1049: A PHASE 2 STUDY OF BEZUCLASTINIB (CGT9486), A NOVEL, HIGHLY SELECTIVE, POTENT
KIT D816V INHIBITOR, IN ADULTS WITH ADVANCED SYSTEMIC MASTOCYTOSIS (APEX): METHODS,
BASELINE DATA, AND EARLY INSIGHTS
D. J. DeAngelo1,*, V. Pullarkat2, M. Piris-Villaespesa3, T. I. George4,5, J. L. Patel4,5,
C. Ustun6, P. Bose7, M. L. Heaney8, A. Pilla9, M. Massaro9, B. Exter9, H. A. Jolin9,
Z. Mikhak9, T. Tashi5
1Dana-Farber Cancer Institute, Boston; 2City of Hope, Duarte, United States of America;
3Hospital Universitario Ramón y Cajal, Madrid, Spain; 4ARUP Laboratories; 5Huntsman
Cancer Institute, University of Utah, Salt Lake City; 6Rush University Cancer Center,
Chicago; 7The University of Texas MD Anderson Cancer Center, Houston; 8Columbia University
Medical Center, New York; 9Cogent Biosciences, Cambridge, United States of America
Background: Systemic mastocytosis (SM) is a rare disease characterized by accumulation
of pathogenic mast cells in bone marrow and extracutaneous tissues with or without
skin involvement. In approximately 95% of adult patients, SM pathogenesis is driven
by a gain-of-function mutation (D816V) in exon 17 of the KIT gene. Advanced systemic
mastocytosis (AdvSM) is an aggressive, life-threatening form of SM with three variants:
aggressive systemic mastocytosis (ASM), SM with an associated hematologic neoplasm
(SM-AHN), and mast cell leukemia (MCL). Overall survival ranges from <6 months to
3-4 years. Bezuclastinib (CGT9486) is an orally administered, highly selective and
potent tyrosine kinase inhibitor (TKI) that targets KIT D816V, avoiding other closely
related kinases with clinical liabilities such as PDGFRα, PDGFRβ, and CSF1R. Bezuclastinib
has demonstrated low brain penetration in preclinical tissue distribution studies,
potentially addressing challenges associated with other targeted agents such as CNS
effects and intracranial hemorrhage. Bezuclastinib has been administered to >100 participants
in clinical trials, including 51 patients with advanced solid tumors (e.g., GIST)
in a Phase 1b/2a study. In that study, bezuclastinib led to a reduction in KIT exon
17 mutational burden, temporally correlated with a reduction in tumor burden, and
an acceptable safety profile.
Aims: To describe the Apex study methods and present baseline data and preliminary
findings from Part 1 of this Phase 2 clinical trial evaluating bezuclastinib in patients
with AdvSM (NCT04996875).
Methods: This open-label, 2-part, multicenter study aims to enroll approximately 140
adult patients with AdvSM per WHO criteria assessed with SM-related organ damage and
baseline serum tryptase of ≥20 ng/mL. Part 1 patients are randomized 1:1:1:1 to 50,
100, or 200 mg of bezuclastinib twice daily, or 400 mg once daily. Data from Part
1 will help determine the optimal dose of bezuclastinib, which will be utilized to
further assess safety, efficacy, PK, and PD in Part 2. The primary efficacy endpoint
is overall response rate (% patients classified as confirmed responders; CR, CRh,
PR and CI) according to mIWG-MRT-ECNM response criteria, assessed by a Central Response
Review Committee.
Results: Patients diagnosed with ASM, SM-AHN, and MCL have been enrolled in Part 1
(dose optimization) at centers in Europe and the United States. Baseline values include
median (min, max) serum tryptase of 170 ng/mL (95.7-303), KIT D816V variant allele
fraction 8.7% (7.04-13.48) by ddPCR, and bone marrow mast cells of 70% (7-80) by IHC.
Dosing continues in these patients and the study is actively enrolling. Preliminary
safety, PK, and PD data from Part 1 will be presented. Safety data will include AEs/SAEs/AECIs
and dose modification frequencies while PD biomarker data, as early evidence of target
engagement and clinical activity, will include changes in mast cell and KIT D816V
mutational burden.
Summary/Conclusion: There is a significant unmet need for novel, safe, and effective
treatment options for patients with AdvSM. Bezuclastinib, an orally administered and
highly selective TKI with potent activity against KIT D816V, may be a treatment option
in AdvSM. Part 1 of the Apex study aims to determine the optimal dose to further evaluate
in Part 2. Early insights into the safety and tolerability profile and biomarker data
will further inform clinical development of bezuclastinib in AdvSM.
P1050: THROMBOCYTOPENIC MYELOFIBROSIS (MF) PATIENTS PREVIOUSLY TREATED WITH A JAK
INHIBITOR IN A PHASE 3 RANDOMIZED STUDY OF MOMELOTINIB (MMB) VERSUS DANAZOL (DAN)
[MOMENTUM]
A. Vannucchi1,*, R. Mesa2, A. Gerds3, H. K. Al-Ali4, D. Lavie5, A. Kuykendall6, S.
Grosicki7, A. Iurlo8, Y. T. Goh9, M. Lazaroiu10, M. Egyed11, M. L. Fox12, D. McLornan13,
A. Perkins14, S.-S. Yoon15, V. Gupta16, J.-J. Kiladjian17, R. Donahue18, J. Kawashima18,
S. Verstovsek19
1Center Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, University
of Florence, Florence, Italy; 2UT Health San Antonio Cancer Center, San Antonio, TX;
3Cleveland Clinic Department of Hematology and Medical Oncology, Avon, OH, United
States of America; 4University Hospital of Halle, Halle, Hungary; 5Hadassah Hebrew
University Medical Center, Jerusalem, Israel; 6Moffitt Cancer Center, Tampa, FL, United
States of America; 7Medical University of Silesia, Katowice, Poland; 8Foundation IRCCS
Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy; 9Singapore General Hospital,
Singapore, Singapore; 10Policlinica de Diagnostic Rapid Brasov, Brasov, Romania; 11Somogy
County Mór Kaposi General Hospital, Kaposvár, Hungary; 12Department of Hematology,
Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Hospital Universitari, Vall
d’Hebron Barcelona Hospital Campus, Barcelona, Spain; 13Guy’s and Saint Thomas’ NHS
Foundation Trust, London, United Kingdom; 14Monash University, Melbourne, Australia;
15Seoul National University Hospital, Seoul, South Korea; 16Princess Margaret Cancer
Centre, Toronto, ON, Canada; 17Saint-Louis Hospital (AP-HP), Paris, France; 18Sierra
Oncology, Inc., San Mateo, CA; 19The University of Texas MD Anderson Cancer Center,
Houston, TX, United States of America
Background: MMB, a novel oral ACVR1/ALK2 and JAK1/2 inhibitor, showed clinical activity
on MF symptoms, red blood cell (RBC) transfusion requirements (anemia), and spleen
volume in the SIMPLIFY trials, including in MF patients (pts) with thrombocytopenia.
Aims: MOMENTUM is a pivotal phase 3 study of symptomatic and anemic MF pts previously
treated with a JAK inhibitor (JAKi) testing MMB vs DAN. This analysis evaluated MOMENTUM
pts with baseline (BL) platelet counts (PLT) ≤150 x 109/L on key symptom, anemia,
and spleen volume endpoints at 24 weeks (wks).
Methods: Eligibility: Primary or post-ET/PV MF; DIPSS high risk, Int-2, or Int-1;
MF Symptom Assessment Form Total Symptom Score (MFSAF TSS) ≥10; hemoglobin (Hgb) <10 g/dL;
prior JAKi for ≥90 days, or ≥28 days if RBC transfusions ≥4 units in 8 wks or Gr 3/4
thrombocytopenia, anemia, or hematoma; palpable spleen ≥5 cm; PLT ≥25 x 109/L. JAKi
taper and washout was ≥21 days. Randomization: 2:1 to MMB 200 mg QD plus DAN placebo
or DAN 600 mg QD plus MMB placebo for 24 wks. Primary endpoint: TSS response (≥50%
reduction from BL) rate at wk 24. Key secondary endpoints, assessed sequentially at
wk 24: RBC transfusion independence (TI) rate, splenic response rate (SRR; ≥25% reduction
in volume from BL), change from BL in TSS, SRR (≥35% reduction from BL) and rate of
zero transfusions since BL. Informed consent was obtained from all participants.
Results: 64% of the MOMENTUM pts had BL PLT of ≤150 x 109/L. Of this subset, 60 (74%)
of 81 MMB pts and 25 (58%) of 43 DAN pts completed the 24-week randomized treatment
(RT) phase. In this subset, median BL TSS were 29 (MMB) and 24 (DAN), Hgb were 7.9
(MMB) and 8.0 (DAN) g/dL, and PLT were 67 x 109/L (MMB) and 64 x 109/L (DAN). BL mean
spleen volume was 2504 (MMB) and 2282 (DAN) cm3. Prior JAKi was ruxolitinib in 124
pts (100%) and fedratinib in 6 pts (5%); mean duration of prior JAKi was 136 weeks.
Efficacy results are in Table. These results are consistent with the overall intent-to-treat
(ITT) analysis set (N=195). Most common Gr ≥3 treatment-emergent adverse events (TEAEs)
in the RT phase were thrombocytopenia (MMB, 31%; DAN, 16%) and anemia (MMB, 7%; DAN,
14%); Gr ≥3 bleeding events occurred in 9% of MMB and 5% of DAN pts. TEAEs led to
study drug discontinuation in 15% of MMB and 19% of DAN pts, and serious TEAEs were
reported in 36% of MMB and 40% of DAN pts, in RT phase. A trend toward improved OS
up to wk 24 was seen with MMB vs DAN [HR (95% CI)=0.490 (0.195, 1.235)]. Additional
analyses of pts with BL PLT <100 x 109/L (N=100) and BL PLT <50 x 109/L (N=31) show
similar treatment effects of MMB vs DAN.
Image:
Summary/Conclusion: In thrombocytopenic MF pts who were symptomatic and anemic, MMB
was superior to DAN for symptom responses, transfusion requirements, and spleen responses
and showed comparable safety and favorable survival. MMB may address a critical unmet
need in thrombocytopenic MF pts. NCT04173494.
P1051: A PHASE 2 STUDY OF IMG-7289 (BOMEDEMSTAT) IN PATIENTS WITH ADVANCED MYELOFIBROSIS
H. Gill1,*, A. Yacoub2, K. Pettit3, T. Bradley4, A. Gerds5, M. Tatarczuch6, J. Shortt7,
N. Curtin8, J. Rossetti9, K. Burbury10, A. Mead11, J. Göthert12, S. Koschmieder13,
A. Jones14, J. Peppe14, J. Dias15, G. Natsoulis16, T. McClure17, M. Kleppe15, W.-J.
Hong14, W. Stevenson18, J. Ewing19, J. Chacko20, E. Rumi21, A. Halpern22, F. Palandri23,
N. Vianelli23, F. Passamonti24, R. Mesa25, M. Marchetti26, C. Harrison27, A. Vannucchi28,
J. Watts29, D. Ross30, M. Talpaz31, H. Rienhoff32
1Medicine, University of Hong Kong, Hong Kong, China; 2Internal Medicine, The University
of Kansas Cancer Center, Leawood; 3Internal Medicine, University of Michigan, Ann
Arbor; 4Hematology and Medical Oncology, University of Miami, Sylvester Comprehensive
Cancer Center, Miami; 5Hematology and Medical Oncology, Cleveland Clinic, Cleveland,
United States of America; 6Haematology and Oncology, Monash Health; 7Clinical Sciences,
Peter MacCallum; 8Monash Health, Monash University, Melbourne, Australia; 9Medicine,
UPMC Hillman Cancer Center, Pittsburgh, United States of America; 10Oncology, Peter
MacCallum Cancer Centre, Melbourne, Australia; 11MRC Weatherall Institute of Molecular
Medicine, University of Oxford, Oxford, United Kingdom; 12Hematology, University Hospital
Essen, Essen; 13Hematology, Oncology, Hemostaseology and Stem Cell Transplantation,
Aachen University, Aachen, Germany; 14Hematology; 15Basic Sciences; 16Genetics; 17Biometrics,
Imago BioSciences, South San Francisco, United States of America; 18Kolling Institute
of Medical Research, Royal North Shore Hospital, Sydney, Australia; 19Haematology,
University Hospitals Birmingham, Birmingham; 20Haematology, The Royal Bournemouth
& Christchurch Hospitals, Bournemouth, United Kingdom; 21Haematology, Fondazione IRCCS
Policlinico San Matteo, Pavia, Italy; 22Medicine, University of Washington, Seattle,
United States of America; 23Azienda Ospedaliero, Universitaria di Bologna Policlinico
S.Orsola-Malpighi, Bologna; 24Haematology, University of Insubria, Varese, Italy;
25UT Health San Antonio Cancer Center, University of Texas Health Science Center,
San Antonio, United States of America; 26Haematology, Azienda Ospedaliera SS Antonio
e Biagio e Cesare Arrigo, Alessandria, Italy; 27Haematology, Guy’s and St Thomas’,
London, United Kingdom; 28Center for Research and Innovation of Myeloproliferative
Neoplasms, University of Florence, Florence, Italy; 29Medicine, University of Miami,
Sylvester Comprehensive Cancer Center, Miami, United States of America; 30Haematology,
Flinders Medical Centre and University, Adelaide, Australia; 31Medicine, University
of Michigan, Ann Arbor; 32Hematology, Imago BioSciences, San Carlos, United States
of America
Background: Lysine-specific demethylase-1 (LSD1) is an activity critical for the self-renewal
of malignant myeloid cells and maturation of megakaryocytes, cells central to the
pathogenesis of MF. Bomedemstat is an orally active LSD1 inhibitor that in mouse models
ameliorated the hallmarks of MPNs and improved survival (Kleppe et al. 2015; Jutzi
et al. 2018).
Aims: IMG-7289-CTP-102 is an ongoing, global, open-label Phase 2 study evaluating
bomedemstat dosed QD in MF patients (NCT03136185). Key eligibility criteria include
patients intolerant, refractory, resistant, or inadequately controlled by approved
therapy, and platelet count ≥100 x 109/L. Key objectives are safety and reduction
of spleen volume (SVR) by MRI/CT and total symptoms scores (TSS) using the MPN-SAF
instrument.
Methods: Serial bone marrow (BM) biopsies and imaging studies are read centrally.
261 genes are serially sequenced to quantify changes in the allelic frequencies of
mutations (MAF) and identify new mutations. The starting dose is 0.6 mg/kg/d titrating,
as needed, to a platelet count of 50-75x109/L.
Results: At 89 patients, the study is now fully enrolled: 46% primary MF, 33% post-essential
thrombocythaemia-MF, 21% post-polycythaemia vera-MF. Median age is 68 (35-88) with
52% males. Prior treatment with ruxolitinib was reported in 83% (74/89); 46% had also
received at least 1 additional treatment. 30% of patients (27/89) had received ≥1
RBC transfusion prior to dosing. By IPSS, 53% were high-risk, 40% int.-2, and 7% int.-1.
At screening (N=103), sequencing to a mean depth of >1000 bp, JAK2 was mutated in
69%, CALR in 21%, MPL in 6%; 59% had ≥2 mutations of which 71% were high-molecular
risk mutations in ASXL1, IDH1/2, EZH2, U2AF1, TP53 and/or SRSF2.
At data cutoff (3 Feb 2022), the median duration of treatment is 28 weeks (2-131).
Of patients for whom TSS data is available at 24 weeks in those with baseline values
≥20, 72% (18/25) had a reduction in TSS; 24% (6/25) reported a ≥50% reduction. Of
patients evaluable for SVR (N=50), 64% had a reduction in spleen volume from baseline;
in 28%, SVR was ≥20%. Of evaluable patients (N=41), 90% had stable (∆ <±1.0 g/dL)
or improved (≥1.0 g/dL) hemoglobin. Of patients with BM fibrosis scoring post-baseline
(N=52), 31% improved by 1 grade and 50% were stable. CCL5 and S100A8/A9 cytokines
were elevated at baseline in 50% (16/32) and 78%, respectively; at Day 84, 81% and
68%, respectively, showed reductions of at least 10% and of those, 100% (CCL5) and
47% (S100A8/A9) normalized. In follow-up sequencing at around Week 24, of 60 mutant
alleles in 32 patients, the mean MAF fell by 39% in 48%. JAK2 MAFs fell by 31% [SD
5%] in 46% (N=24); ASXL1 MAFs fell 40% in 71%. Clones with JAK2 and/or ASXL1 mutations
were most affected. No new mutations have been identified and no patient has transformed
to AML.
The most common non-hematologic AEs reported by patients was dysgeusia in 36% (32/90)
and diarrhoea in 34% (31/90). All dysgeusia events were grade 1/2 and 1 led to treatment
discontinuation. Of 14 related SAEs, 4 were Grade 2, 9 Grade 3 and 1 Grade 4 (thrombocytopenia).
Twenty-nine patients remain on bomedemstat. Early terminations due to AEs occurred
in 18 (20%) patients (9 related to bomedemstat), and 13 discontinued for other reasons.
There have been no safety signals, DLTs, or deaths related to drug.
Summary/Conclusion: In patients with advanced MF, bomedemstat alone had an acceptable
tolerability profile, relieved symptoms, reduced spleen volume and mutation burden
while improving fibrosis and anemia without safety signals.
P1052: IMPACT OF SF3B1 MUTATION IN MYELOFIBROSIS
J. Senapati1,*, S. Verstovsek1, L. Masarova1, N. Pemmaraju1, G. Montalban Bravo1,
S. Pierce1, L. Zhou1, G. Garcia-Manero1, H. Kantarjian1, P. Bose1
1Leukemia, MD Anderson Cancer Center, Houston, United States of America
Background: Splicing factor 3B subunit 1 (SF3B1) mutations have been shown to confer
a unique phenotype in MDS and MDS/MPN overlap syndromes, with ring sideroblasts, thrombocytosis
and a relatively favorable prognosis. In myelofibrosis (MF), the frequency of SF3B1
mutation is <10% and it may play a less important role in disease outcomes (Lasho
et. al, Leukemia, 2011).
Aims: We aimed to characterize the phenotypic and genotypic associations of SF3B1
mutations in pts with MF and compare them to SF3B1 wild type MF.
Methods: We retrospectively analyzed all pts with WHO-defined MF (primary, including
pre-fibrotic MF, or progressed from PV or ET) seen at our center from 1/2017 through
7/2021. All pts had a BM biopsy with karyotyping and most of the pts had an 81-gene
myeloid mutation panel performed by next-generation sequencing (including JAK2, CALR,
MPL and SF3B1) at presentation or at progression to MF from PV or ET. We compared
the disease phenotypes, MPN driver and co-occurring mutations, cytogenetics, DIPSS,
transfusion requirements, treatment characteristics and outcomes between pts with
SF3B1 -mutated (SF3B1
+) and -wild type (SF3B1
-) MF.
Results: 381 pts with MF were identified, 29 (8%) of whom were SF3B1
+. There were similar frequencies of JAK2, CALR and MPL driver mutations and “triple
negative” MF in the SF3B1
+ and SF3B1
- groups (Table 1). The median number of SF3B1 mutations was 1 (range, 1-2); K666N
being the most common (52%), followed by K700E (14%). 10% SF3B1
+ pts were DIPSS high risk, compared to 19% SF3B1
- pts (p=0.3). There were no significant differences between the 2 groups in terms
of baseline hemoglobin level, white blood cell count, platelets, co-occurring mutations,
and symptom burden but more SF3B1
+ pts were red blood cell (RBC)-transfusion dependent at presentation than SF3B1
- pts (38% vs. 15%, p=0.003). A total of 152 (43%) SF3B1
- pts and 14 (48%) SF3B1
+ pts received ruxolitinib-based therapy (p=0.7). At a median follow up (mFU) of 17.2
months (mos) for the entire cohort (range, 0.1- 52.2 mos; mFU 22.6 mos for SF3B1
+ and 16.8 mos for SF3B1
-), 41 pts had died [4 (14%) SF3B1
+ and 37(10%) SF3B1
-] and 7 pts (1 SF3B1
+ and 6 SF3B1
-) had experienced leukemic transformation. The median estimated OS was 230 mos for
SF3B1
- vs. not reached for SF3B1
+ pts (p = 0.57, Fig. 1).
Table 1:
Patient chracteristics, treatment and outcomes
Characteristics
SF3B1+ (N= 29)
SF3B1- (N= 352)
p value
N/Median (%) [range]
Age, years
72 [54-91]
69 [21-89]
Gender
Males
24 (83)
202 (57)
Diagnosis
PMF
21 (72)
226 (64)
Post ET/PV MF
8 (28)
126 (36)
Baseline CBC
Hb (g/dl)
9.7 [6.9-18.4]
10.5 [5.5-17.6]
0.4
WBC (109/L)
8.3 [1.3-73.3]
8.5 [1.1-212.1]
0.9
Platelet (109/L)
230 [19-1188]
216 [1-1271]
0.5
Driver mutation
JAK2
18 (62)
214 (60)
0.9
CALR
5 (17)
82 (23)
0.6
MPL
3 (10)
29 (8)
0.7
Triple negative
3 (10)
32 (9)
0.7
JAK2 allele burden (n=28 SF3B1+,343 SF3B1-)
45 [1-81]%
47 [1-99]%
0.5
Co-mutations (n=20 SF3B1+, 205 SF3B1-)
Splicing
3 (15)
59 (29)
0.3
Epigenetic
15 (75)
157 (76)
0.9
TP53
2 (10)
10 (5)
0.3
CTG (n=25 SF3B1+, 264 SF3B1-)
Non diploid karyotype
12 (48)
102 (39)
0.4
BM fib (n=29 SF3B1+, 274 SF3B1-)
Grade 3
3 (24)
113 (41)
0.001
DIPSS
High
3 (10)
67 (19%)
0.3
Transfusion dependency
RBC
11 (38)
52 (15)
0.003
Platelet
0 (0)
4 (1)
0.99
Events
Leukemic transformation
1 (3)
6 (2)
0.4
Death
4 (14)
37 (10)
Image:
Summary/Conclusion:
SF3B1 mutation is an uncommon event in MF and does not substantially affect the disease
phenotype and outcomes. SF3B1 mutated MF pts appear to be more be RBC transfusion
dependent given the underlying biology of SF3B1 mutation.
P1053: ALBUMIN AND C-REACTIVE PROTEIN PROVIDE PROGNOSTIC INFORMATION INDEPENDENTLY
FROM MIPSS70 IN OVERT MYELOFIBROSIS. A RETROSPECTIVE STUDY.
N.-M. Messerich1, T. Volken2, S. Cogliatti3, T. Lehmann4, A. Holbro5, I. Demmer3,
R. Benz6, W. Jochum3, T. Silzle4,*
1Department of Intensive Care, Cantonal Hospital St. Gallen, St. Gallen; 2Institute
of Public Health, ZHAW School of Health Sciences, Winterthur; 3Institute of Pathology;
4Clinic for Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen;
5Division of Hematology, University Hospital Basel, Basel; 6Clinic for Hematology
and Oncology, Spital Thurgau AG, Münsterlingen, Switzerland
Background: Accurate prognostication is essential for patients with myelofibrosis
(MF). The Dynamic Prognostic Scoring System (DIPSS) relies on age and several clinical
and laboratory parameters and can be refined by information about chromosomal and/or
molecular aberrations. C-reactive protein (CRP) and albumin have been reported to
add prognostic information to both DIPSS and DIPSS-plus, which incorporates certain
chromosomal aberrations and to scoring systems including both molecular and cytogenetic
data (e.g. Molecular International Prognostic Scoring System 70 (MIPSS70)-plus V2).
The impact of CRP and albumin in the context of the MIPSS70, which is useful, if metaphase
cytogenetics are not obtainable, but next generation sequencing (NGS)-data are on
hand, has not yet been reported.
Aims: To define the prognostic impact of albumin and CRP in the context of the MIPSS70.
Methods: We performed a retrospective chart review of all patients diagnosed at our
institution with MF between 2000 and 2020 and collected information from hospital
records as documented at diagnosis. For cases lacking NGS during diagnostic-workup,
NGS was performed retrospectively, if DNA was available from diagnostic samples (Oncomine
Myeloid Research Assay, Thermo Fisher Scientific, Waltham, MA, USA).
Results: 79 patients (median age 72 years, range 28-87; primary MF n=54, MF following
ET or PV: n=22) were evaluable. Thirty-eight (48.1%) died during follow-up (median
34 months; range 0-184). CRP-values were available for 71/79 (89.9%), albumin-values
in 62/79 (78.5%) and both parameters in 57/79 (72.2%) patients.
A CRP-elevation (>8mg/l, n=24) was associated with a shorter overall survival (OS)
compared to a CRP <8mg/l (n=47, median 44 vs. 89 months, p<0.001, see fig A), as was
an albumin below the median of the population (</≥ 40 g/l, n=31 each, median 50 vs.
101 months, p=0.018; see fig B). In univariate analyses, both CRP >8mg/l and albumin
<40 g/l were associated with a higher risk of death (HR 3.85, 95% CI 1.85-8.0, p<0.001
and HR 2.49, 95% CI 1.13-5.49, p=0.024).
A logistic regression model showed a continuously rising probability of death with
lower albumin levels even in the range of normal (at our institution: 34-45 g/l; OR=0.85,
95% CI 0.73-0.99; p=0.043)
The MIPSS70 was available for 60/79 (76%) patients. Due to the low number of low-risk-patients
(n=2) we split the cohort into a “MIPSS70dichlow/intermediate” risk group (MIPSS70-low
and MIPSS70-intermediate, n=45) and a “MIPSSdichhigh” risk group, comprising the 15
MIPSS70 high-risk patients, for further analyses.
CRP >8 mg/l and albumin <40 g/l retained their prognostic value, if included each
into a bivariate model together with the MIPSS70dich (HR 2.498, 95% CI 1.13-5.52,
p=0.0236 and HR 5.486, 95% CI 1.89-15.96, p=0.0018).
After inclusion of CRP >8 mg/l and albumin< 40 g/l together with MIPSS70dich into
a multivariate model, only albumin <40 g/l added prognostic information (HR 3.8, 95%
CI 1.27-11.48, p=0.0173).
Image:
Summary/Conclusion: Albumin and CRP deserve further evaluation as prognostic factors
in myelofibrosis, since they add prognostic information irrespective of the molecular
risk profile. In a common view, CRP and hypalbuminaemia reflect the extent of MF-associated
inflammation. However, already albumin-levels in the lower range of normal are associated
with an increased mortality. Given its role as main antioxidant in the extracellular
space and its potential anti-inflammatory/anti-thrombotic properties, higher-albumin
levels could be associated with an increased capability to counteract MF-associated
inflammation.
P1054: MYLOX-1: AN OPEN-LABEL, PHASE IIA STUDY OF THE SAFETY, TOLERABILITY, PHARMACOKINETICS
AND PHARMACODYNAMICS OF ORAL LOXL2 INHIBITOR, GB2064, IN MYELOFIBROSIS
C. Harrison1, J. Mascarenhas2, R. Rampal3, D. Cilloni4, B. Lindmark5,*, B. Singh5,
B. Jacoby5, S. Verstovsek6
1Guy’s and St Thomas Hospital, London, United Kingdom; 2Icahn School of Medicine at
Mount Sinai; 3Memorial Sloan Kettering, New York, United States of America; 4University
Hospital San Luigi Gonzaga, Orbassano, Italy; 5Galecto Inc, Copenhagen, Denmark; 6MD
Anderson Cancer Center, Houston, United States of America
Background: GB2064 is a high-affinity, selective, pseudo-irreversible, small-molecule
inhibitor of LOXL2, a secreted glycoprotein that crosslinks extracellular matrix collagens
and elastin which contributes to stiffness and loss of function of fibrotic organs.
GB2064 is being developed as an oral treatment for myelofibrosis (MF), a rare myeloproliferative
disease with high morbidity and mortality. Janus kinase (JAK) inhibitor therapy has
brought significant advancements in the treatment of MF, but a significant proportion
of patients would eventually discontinue treatment, predominantly due to the development
of cytopenia (Kyukendall et al Ann, Hematol 2018). Thus, there remains a substantial
unmet need for developing well-tolerated disease-modifying treatments that reduce
bone marrow fibrosis to improve haematologic parameters, splenomegaly, symptom burden
and quality of life.
Aims: To assess the safety, tolerability, pharmacokinetics (PK), pharmacodynamics
(PD) and clinical effects of oral GB2064 (1000 mg twice daily [BID]) dosed for 9 months
to participants with primary or secondary myelofibrosis (PMF/SMF).
Methods: Open-label study in 16 adult participants diagnosed with PMF or SMF in accordance
with World Health Organization diagnostic criteria (Barbui et al, Blood Cancer 2018;
Cruz et al, Expert Rev Hematol 2020), who are not taking a JAK inhibitor and therefore
likely to be refractory, intolerant or ineligible for such inhibitors, with Eastern
Cooperative Oncology Group performance status 0-2 and clinical laboratory parameters
within appropriate limits per protocol. Primary endpoint is safety and tolerability.
Safety and tolerability, PK, PD and appropriate MF-specific assessments will take
place at all visits, except Day 7, Day 15 and Month 4 when only safety and tolerability
will be assessed (Fig A). Bone marrow biopsies, magnetic resonance imaging (MRI) of
spleen and quality of life measures, MPN-10 and EQ-5D-5L, are performed at prespecified
timepoints within the protocol. Exploratory endpoints include LOXL2 binding assay
in the circulation, relationships between PK plasma exposures, PD markers, and markers
of clinical activity, fibrosis and inflammation biomarkers (YKL-40, PAI-1, PDGF, CCN2,
collagen formation and degradation neoepitopes). The study is not formally powered.
Participants who derive benefit may continue therapy for an additional 3 years (Fig
B).
Results: More than half of the intended participants have been enrolled and are on
treatment with GB2064 as of February 2022.There have been no significant safety concerns
observed at a dose of 1000 mg BID to date.
Image:
Summary/Conclusion: MYLOX-1 is designed to explore the safety and clinical effects
of GB2064, a novel small-molecule LOXL-2 inhibitor, addressing bone marrow fibrosis
as a main element of myelofibrosis, with the aim of decreasing extramedullary haematopoesis
and improving haematological parameters, symptom burden and the quality of life for
patients with MF.
P1055: CLINICAL AND GENETIC RESULTS OF THE PHASE IB/II TRIAL MPNSG-0212: RUXOLITINIB
PLUS POMALIDOMIDE IN MYELOFIBROSIS WITH ANEMIA
F. Stegelmann1,*, E. Jahn1, S. Koschmieder2, F. Heidel3, A. Hochhaus4, H. Hebart5,
S. Isfort2, A. Reiter6, M. Bangerter7, C. F. Waller8, D. Wolleschak9, C. Scheid10,
J. Göthert11, P. Schafhausen12, T. Kindler13, M. P. Radsak13, N. Gattermann14, R.
Möhle15, N. von Bubnoff16, A. Schrade1, T. Brümmendorf2, H. Döhner1, M. Griesshammer17,
K. Döhner1
1Internal Medicine III, University Hospital of Ulm, Ulm; 2Hematology, Oncology, Hemostaseology
and Stem Cell Transplantation, RWTH Aachen University, Aachen; 3Klinik und Poliklinik
für Innere Medizin C, Universitätsmedizin Greifswald, Greifswald; 4Klinik für Innere
Medizin 2, Universitätsklinikum Jena, Jena; 5Internal Medicine, Stauferklinikum, Mutlangen;
6III. Medical Department, University Medical Centre Mannheim, Mannheim; 7Hematology
and Oncology, Practice, Augsburg; 8Department of Internal Medicine I, University Hospital
of Freiburg, Freiburg; 9Medical Center, Otto-von-Guericke University, Magdeburg; 10Department
I of Internal Medicine, University of Cologne, Cologne; 11Department of Hematology,
University Hospital of Essen, Essen; 122nd Medical Clinic, University Hospital of
Hamburg-Eppendorf, Hamburg; 13Internal Medicine III, Johannes Gutenberg University,
Mainz; 14Hematology, Oncology and Clinical Immunology, Heinrich Heine University,
Düsseldorf; 15Hematology and Oncology, University Tübingen, Tübingen; 16Hematology
and Oncology, University Hospital of Schleswig-Holstein, Lübeck; 17Hematology/Oncology,
Universitätsklinik Minden, Minden, Germany
Background: Ruxolitinib (RUX) alleviates disease-associated symptoms including splenomegaly
in patients (pts) with myelofibrosis (MF). However, management of cytopenia remains
challenging.
Aims: As single-agent pomalidomide (POM) improved cytopenia in 14-29% of MF pts in
our previous MPNSG-0109 trial, we sought to investigate the combination of RUX plus
POM in MF pts with anemia (Hb <10 g/dL and/or RBC transfusion dependency [RBC-TD]).
Methods: MPNSG-0212 is a multicenter, open-label, phase-Ib/II trial (NCT01644110)
comprising 39 pts in cohort 1 (co1, recruited 2013-2017) and 52 pts in co2 (2017-2021).
Co1 pts received RUX 10 mg BID plus POM 0.5 mg QD, while POM was intended to be increased
in co2 to 1 and 2 mg QD after 3 and 6 28-day-cycles, respectively. Primary endpoint
was response according to IWG-MRT and RBC-TI criteria at end of cycle 12 (EOC12).
In addition, genomic landscape was characterized in all pts using targeted NGS of
269 candidate genes (Illumina NextSeq550).
Results: Co1 and co2 pts had similar characteristics: median age was 71 years (range
49-86), median Hb level 8.6 g/dL (5.4-11.7), and median spleen size 17.5 cm (11.4-36);
30% were RBC-TD, 66% intermediate-2, and 25% high-risk according to DIPSS; mutations
(muts) in JAK2, CALR, or MPL were identified in 57%, 23%, and 20%, respectively; 55%
had ≥1 high-molecular risk (HMR) mut, with ASXL1 being the most common (41%), followed
by SRSF2 (24%), EZH2 (10%) and IDH2 (9%). Of note, pts in co2 were more frequently
pre-treated with RUX compared to co1 (44% vs 15%; p=.006).
Median treatment time at data cut-off was 12 cycles in co1 (2-98) and co2 (3-46);
3 pts of co2 have not yet reached EOC12.
In co1, 8/39 pts (21%) achieved response at EOC12: partial remission (PR, n=1), clinical
improvement (CI, n=6), or RBC-TI (n=1); 18/39 pts (46%) were treated for >12 cycles
due to response (n=8), clinical benefit (CB, n=5) defined as Hb increase ≥1 g/dL or
>50% improvement of ≥1 quality of life (QoL) symptom according to MPN-SAF, or stable
disease (SD, n=5). Median Hb level at EOC12 was 9.3 g/dL; 15/39 pts (38%) were on
treatment for >30 cycles, and 3 pts remained on long-term treatment (cycle 76, 97,
and 98).
In 73% and 40% of pts in co2, POM dose was increased to 1 mg and 2 mg QD after cycle
3 and 6, respectively. 5/49 pts (10%) showed response at EOC12 (CI, n=3 and RBC-TI,
n=2), while 14 additional pts (29%) had CB; median Hb level at EOC12 was 8.6 g/dL;
22/49 pts (45%) were treated for >12 cycles; 3 pts were still on treatment (cycle
21, 36, and 46).
Targeted NGS did not reveal a distinct mutational pattern associated with treatment
response. Median overall survival (OS) of co1 and co2 was 3.6 and 2.6 years, respectively
(p=.66). Among all, CALR, but not JAK2 or MPL muts were associated with better OS
(p=.04), whereas HMR muts were in trend prognostically adverse (p=.057). Pts with
more than 3 muts had a worse median OS (1.2 vs 4 years, p=.002).
Combination therapy was well tolerated in both cohorts. Most common grade 1/2 adverse
events (AE) were dyspnea (28%) and fatigue (23%), while the most frequent grade 3
AE was worsening of anemia (32%) in the first weeks of combination treatment not limiting
therapy.
Summary/Conclusion: Treatment with RUX and POM was safe and feasible in our study
of advanced MF with an adverse genetic profile. Almost half of co1 and co2 pts were
treated >12 cycles due to clinical benefit of the combination therapy with a subset
of 38% of pts in co1 receiving treatment for >30 cycles. Dose increase of POM in co2
did not result into better anemia response or improvement of OS.
Authors FS and EJ contributed equally.
P1056: SCREENING OF JAK2 EXON 12 SOMATIC MUTATIONS BY HIGH-RESOLUTION MELTING CURVE
ANALYSIS
D. Kurochkin1,*, I. Maslyukova1, T. Subbotina1,2, A. Khazieva3, E. Dunaeva4, K. Mironov4
1Siberian Federal University; 2The Federal State-Financed Institution «Federal Siberian
Research Clinical Centre under the Federal Medical Biological Agency»; 3Regional Clinical
Hospital, Krasnoyarsk; 4Central Research Institute of Epidemiology of The Federal
Service on Customers’ Rights Protection and Human Well-being Surveillance, Moscow,
Russia
Background:
JAK2 exon 12 mutations are seen in about 2-5% of JAK2V617F-negative cases of polycythemia
vera (PV). Nowadays about 40 different JAK2 exon 12 mutations associated with PV have
been identified and classified. To identify all possible variants, it is necessary
to use sequencing. However, due to the high cost of sequencing, developing a two-stage
algorithm for detect mutations in JAK2 exon 12 using inexpensive screening is of immediate
practically necessity. We have previously proposed a two-stage algorithm for detect
mutations in JAK2 exon 12 using inexpensive screening test by heteroduplex analysis
(Subbotina T et al, Haematologica 2017).
Aims: The aim of this study was to demonstrate the feasibility of HRM analysis using
the CFX96 thermocycler and the Precision Melt Analysis software (Bio-Rad, USA) as
the preliminary screening test for detection of JAK2 exon 12 mutations.
Methods: DNA samples of 5 JAK2 exon 12 mutation positive PV patients were included
in this study. The identification of the JAK2 exon 12 mutation types and allele burden
measurement was carried out by pyrosequencing (Subbotina T et al, Haematologica 2014).
All 5 patients have different mutation variant in the 12 exon of the JAK2 and different
levels of allelic burden: c.1624_1629delAATGAA – 67%; с.1619_1627TCAgAAATg>AAA – 14%;
c.1623_1628delAAATGA – 15%; c.1622_1627delGAAATG – 33%; c.1612_1616CACAA>TT – 21%.
HRM analysis was performed using a Precision Melt Supermix reagent kit in the presence
of Eva Green dye (Bio-Rad, USA). PCR with an additional high-resolution melting step
was carried out on a CFX96 thermocycler (Bio-Rad, USA) according to the following
program: denaturation at 95°C for 2 minutes, then 40 cycles at 95°C for 10 seconds,
58°C in for 30 seconds. The high resolution melting program consisted of denaturation
at 95°C for 30 seconds, renaturation at 60°C for 1 minute, and melting at 65°C to
95°C with a 0.2°C gradient in 10 seconds. Each DNA sample was analyzed in duplicate.
For the two out five JAK2 exon 12 mutations a threshold determination of the mutant
allele presence was analyzed. To analyze the threshold for determining the proportion
of the mutant allele, dilution of cloned wild-type and mutated samples was performed
to obtain samples with different levels of allelic burden: 50%, 25%, 12.5%, 6.25%,
3.13%, 1.56%, 0.78%.
Results: Figure 1 shows the differential melting plots of the DNA fragments. The analyzed
samples were divided into five clusters: first cluster – melting curves of wild type
DNA; second cluster – melting curves of DNA from samples with deletion type mutations:
c.1624_1629delAATGAA and c.1622_1627delGAAATG; the third cluster – the DNA melting
curves from the sample with the combined mutation: c.1612_161616CACAA>TT; the fourth
cluster – DNA melting curves from the sample with deletion type mutation: c.1623 _1628delAAATGA;
fifth cluster – DNA melting curves from a sample with the combined mutation: c.1619_1627TCAGAAATG>AAA.
The detection thresholds in case of the c.1624_1629delAATGAA and с.1619_1627TCAgAAATg>AAA
mutation analysis are 6.25% of the presence of the mutant allele in the samples (data
not shown).
Image:
Summary/Conclusion: Therefore, the HRM analysis that was conducted on the CFX96 allows
to screen highly specific for the PV diagnosis mutations in exon 12 of the JAK2 gene.
The inclusion of this screening research in the laboratory testing algorithm improves
the efficiency and accessibility of molecular genetic technologies in the diagnosis
of PV.
P1057: REAL-WORLD UTILIZATION OF FEDRATINIB FOR MYELOFIBROSIS FOLLOWING RUXOLITINIB
FAILURE
C. Harrison1,*, J. Mascarenhas2, P. Abraham3, J. A. Nadal3, A. Balanean4, A. McBride3,
J. K. Kish4, D. Liassou4, B. A. Feinberg4, A. T. Gerds5
1Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom; 2The Tisch Cancer
Intitute, Icahn School of Medicine at Mount Sinai, New York; 3Bristol Myers Squibb,
Princeton; 4Cardinal Health, Dublin; 5Cleveland Clinic Taussig Cancer Institute, Cleveland,
United States of America
Background: In August 2019, fedratinib (FEDR) became the second treatment, after ruxolitinib
(RUX), to be approved for intermediate (Int)- or high-risk (HR) primary or secondary
myelofibrosis (MF).
Aims: To characterize real-world patient characteristics and treatment patterns for
patients with MF receiving FEDR after RUX therapy in US clinical practices.
Methods: Adults with Int- or HR MF initiating FEDR on or after August 16, 2019 (FEDR
approval date) after discontinuing RUX were identified from community oncology practices.
Eligible patients had ≥90 days of follow-up from FEDR initiation, completed ≥1 FEDR
cycle, and had a spleen size assessment at FEDR initiation. Treating physicians completed
electronic case report forms for the selected eligible patients, capturing patient
characteristics at time of MF diagnosis and during RUX and FEDR therapy, including
bone marrow fibrosis grade, biomarkers, risk score, performance status score, spleen
size (cm above the costal margin), symptoms (per Modified Myelofibrosis Symptom Assessment
Form version 4.0), and complete blood count data. Providers reported treatment dosing
and modifications and dates/rationale for RUX treatment failure (defined a priori,
including refractory or suboptimal response, disease progression [PD], or intolerance).
Results: Among 150 eligible patients, the median age at diagnosis was 68 years; 55%
were male, 68% were non-Hispanic White, 85% had primary MF, 55% were JAK2 V617F-positive,
and 9% were JAK2/MPL/CALR-negative. RUX initiation dosage was ≥20 mg twice/daily (bid)
for approximately 50% of patients. At RUX initiation, 65% had ECOG scores of 0/1,
and 90% had palpable spleen (Table). A total of 19 patients (13%) had RUX dosage reductions
due to neutropenia (7), thrombocytopenia (6), other adverse event (AE)/toxicity (3),
patient request (2), and anemia (1); 4 patients (3%) had treatment interruptions due
to thrombocytopenia (2), neutropenia (1), other AE/toxicity (1), and patient request
(1); and 18 patients (12%) had dosage increases due to titration (13) and persistent
MF symptoms (5). Primary reasons for RUX discontinuation were PD (70%), refractory
or suboptimal response (25%), or intolerance (5%). RUX discontinuation due to PD was
most often attributed to MF symptom return (65%), increased spleen size to >50% vs
best achieved response (23%), and other reasons (12%). Median RUX duration was 4.0
months (IQR, 3.0–6.1) for refractory or intolerant patients and 11.7 months (IQR,
6.5–17.1) for those with PD. FEDR was initiated at 400 mg daily for 74% of patients.
At FEDR initiation, 43% had International Prognostic Scoring System (IPSS)/Dynamic
IPSS HR, 37% Int-2 risk, and 88% had palpable spleen; mean spleen size was 16.0 cm,
and median platelet count was 98.0×109/L (Table). Symptoms reported at FEDR initiation
included fatigue (72%), abdominal discomfort (51%), night sweats (44%), early satiety
(25%), bone pain (18%), itching (13%), and pain under the left rib (13%). At the end
of follow-up, 44 patients (29.3%) were deceased, and mean time to death among deceased
patients was 6.5 (standard deviation, 2.7) months from initiation of FEDR.
Image:
Summary/Conclusion: This study showed a short duration of RUX therapy for real-world
patients with MF, and poorer clinical status at FEDR initiation than at RUX initiation.
Patients at risk of early failure of RUX should be considered for earlier FEDR interventional
treatment.
P1058: REAL-WORLD OUTCOMES WITH FEDRATINIB THERAPY IN PATIENTS WITH PRIMARY MYELOFIBROSIS
POST-RUXOLITINIB DISCONTINUATION
F. Passamonti1,*, Y. Lou2, M. Chevli3, P. Abraham4
1University of Insubria and ASST Sette Laghi, Ospedale di Circolo, Varese, Italy;
2Bristol Myers Squibb, Lawrenceville, United States of America; 3Bristol Myers Squibb,
Uxbridge, United Kingdom; 4Bristol Myers Squibb, Princeton, United States of America
Background: Dysregulation of the Janus kinase 2 (JAK2) hematopoiesis-signaling pathway
in myelofibrosis (MF) disrupts bone marrow production of blood cells, causing anemia,
fatigue, and splenomegaly. Dual JAK1/JAK2 inhibitor ruxolitinib (RUX) was the first
drug approved in the USA for intermediate/high-risk MF, but many patients only have
a partial response or develop cytopenia, ultimately discontinuing treatment. Fedratinib
(FEDR), approved in August 2019, is active against wild-type and mutationally activated
JAK2 and receptor tyrosine kinase FLT3, but its real-world effectiveness in patients
discontinuing RUX has not been evaluated.
Aims: To compare baseline characteristics and survival outcomes for patients with
MF receiving FEDR vs those not receiving FEDR after discontinuing RUX in routine US
clinical practice.
Methods: Patients receiving RUX for primary MF were identified using Flatiron Health’s
nationwide electronic health record-derived database. Eligible patients were those
with ≥2 recorded visits in the database between Jan 1, 2011 and Oct 31, 2020, ≥18
years of age at the index date (start of FEDR therapy for patients receiving FEDR
and last date of RUX therapy for those not receiving FEDR), with data ≥1 month before
and after index, and no record of receiving unclassified clinical study drugs prior
to index. Patients were stratified by treatment with FEDR post-RUX (FEDR and non-FEDR
group); the non-FEDR group was further stratified by time of RUX discontinuation (before
[subgroup A] and after [subgroup B] US approval of FEDR) to enable a contemporaneous
comparison with the FEDR group. Demographics and clinical characteristics were assessed
at index. Overall survival (OS) was defined as time from index until death or censoring
and assessed by Kaplan–Meier analysis; landmark survival was defined as the proportion
of patients who survived at a given point. Associations of baseline variables and
survival were assessed by Cox proportional hazards model.
Results: A total of 229 patients were evaluated (FEDR group: n=70; non-FEDR group:
n=159). Median age at index was 71.0 for the FEDR group and 70.0 years for the non-FEDR
group. Baseline demographic characteristics were broadly similar for the 2 groups.
Median follow-up from index in the FEDR and non-FEDR group was 7.0 and 6.0 months,
respectively. At index, 90.2% (46/51) and 74.3% (84/113) of patients had an ECOG performance
status (PS) score of 0–1, respectively. Median duration of FEDR therapy in the FEDR
group was 3.7 (range, 0–12.2) months, and 47.1% (33/70) received 400 mg FEDR once/day.
Median OS was not reached in the FEDR group (vs 17 months in the non-FEDR group).
Landmark survival in the FEDR and non-FEDR groups was 71.6% and 53.5% at 12 months,
respectively. In the non-FEDR subgroup B, the corresponding survival rate at 12 months
was 47.9%. The Cox proportional hazards model suggested that being male, of ‘other’
race (non-White, non-Black/African–American, non-Asian, non-missing), and having Charlson
comorbidity index ≥1, ECOG PS 2–4, and body mass index <18.5 kg/m2 were significantly
associated with poorer survival. Based on the risk-adjusted analysis, we witnessed
an apparent trend toward increased OS with FEDR, but the observed differences did
not achieve statistical significance (HR [95% CI]: 0.6 [0.3–1.2]; P=0.1466).
Image:
Summary/Conclusion: FEDR treatment of real-world patients with primary MF post-RUX
may offer improved likelihood of survival rate up to 1 year after index compared with
non-FEDR therapy. These differences in survival rate were sustained when the FEDR
group and the non-FEDR B subgroup were compared.
P1059: COMPARATIVE EFFICACY OF FEDRATINIB AND PACRITINIB FOR THE TREATMENT OF MYELOFIBROSIS
IN PATIENTS WITH LOW PLATELET COUNTS: A SIMULATED TREATMENT COMPARISON STUDY
G. Tremblay1,*, P. Daniele1, P. Abraham2, S. Rose2, A. McBride2
1Purple Squirrel Economics, Montreal, Canada; 2Bristol Myers Squibbs, Princeton, United
States of America
Background: Myelofibrosis (MF) is a Philadelphia chromosome-negative myeloproliferative
neoplasm with an estimated prevalence of 4–6/100,000 persons in the United States
(US). Among intermediate- to high-risk patients, Janus kinase 2 (JAK2) inhibitors
are the primary treatment for MF. Fedratinib was approved by the US Food and Drug
Administration (FDA) based on the results of the JAKARTA trials (NCT01437787; NCT01523171),
and a new drug application for pacritinib was submitted to the FDA for the treatment
of MF in patients with severe thrombocytopenia.
Aims: To assess the comparative efficacy of fedratinib and pacritinib in patients
with MF and thrombocytopenia (platelets < 100 × 109/L), in the absence of head-to-head
clinical trials, an indirect treatment comparison (ITC) was conducted.
Methods: According to the results of a systematic literature review, JAKARTA, JAKARTA2,
and PERSIST-2 (NCT01773187) trials formed the basis of the ITC. A pooled analysis
data set was developed using individual patient-level data from the fedratinib 400-mg
arms of JAKARTA and JAKARTA2 including patients with platelets < 100 × 109/L. Published
summary data from the pacritinib 200-mg arm of PERSIST-2 served as the comparator.
Simulated treatment comparisons (STCs) were used to compare spleen volume reduction
(SVR) ≥ 35% while adjusting for mutually reported baseline patient characteristics
such as age, sex, Dynamic International Prognostic Scoring System status, Eastern
Cooperative Oncology Group (ECOG) performance status (PS), JAK2 V617F mutation status,
prior ruxolitinib exposure, and laboratory tests. Final adjustment models were selected
based on fit statistics and the literature review. Indirect relative risk (RR) was
estimated with 95% confidence intervals (CIs) using unanchored naive ITC (unadjusted)
and STC (adjusted) methodologies. A naive ITC was also conducted for the subgroup
of patients who received ruxolitinib in prior lines of therapy. In addition, a sensitivity
analysis was conducted to evaluate the impact of differential median baseline platelet
counts, which PERSIST-2 did not report. Using the full pooled population of the JAKARTA
trials, outcomes were simulated at 3 median baseline platelet counts (25, 50, and
75 × 109/L) and compared using similar methodology to the main analysis.
Results: The main analysis suggests that fedratinib is associated with a greater proportion
of SVR ≥ 35% than pacritinib. According to the naive ITC, fedratinib was numerically
favored over pacritinib in terms of SVR ≥ 35% (RR, 1.67 [95% CI, 0.94–2.97]). After
the STC adjustment, fedratinib was statistically favored relative to pacritinib for
SVR ≥ 35% (RR, 1.76 [95% CI, 1.00–3.10]). The naive ITC results were similar for patients
who had received ruxolitinib in prior lines of therapy (RR, 2.82 [95% CI, 1.02–7.82]).
Results of the platelet-count-adjusted sensitivity analysis showed a significant difference
in favor of fedratinib with respect to SVR ≥ 35%. Differential baseline platelet count
had minimal impact on the ITC results, with similar RRs regardless of simulated median
baseline platelet count.
Image:
Summary/Conclusion: This analysis used a population-adjusted ITC to assess the comparative
efficacy of pacritinib and fedratinib in patients with MF and thrombocytopenia. Following
population adjustment, fedratinib was associated with a greater proportion of patients
achieving SVR ≥ 35% than pacritinib. Further real-world evidence studies should be
conducted to assess the effectiveness of these treatments in clinical settings.
P1060: REAL-WORLD CLINICAL OUTCOMES AFTER 3 AND 6 MONTHS OF TREATMENT WITH FEDRATINIB
FOLLOWING RUXOLITINIB FAILURE
J. Mascarenhas1,*, A. T. Gerds2, J. K. Kish3, P. Abraham4, A. Balanean3, J. A. Nadal4,
D. Liassou3, B. A. Feinberg3, A. McBride4, C. Harrison5
1The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York; 2Cleveland
Clinic Taussig Cancer Institute, Cleveland; 3Cardinal Health, Dublin; 4Bristol Myers
Squibb, Princeton, United States of America; 5Guy’s and St Thomas’ NHS Foundation
Trust, London, United Kingdom
Background: Relief of symptoms and splenomegaly is an important treatment (Tx) goal
for patients with myelofibrosis (MF). Fedratinib (FEDR) and ruxolitinib (RUX) are
approved in the USA for Intermediate (Int)- or High-risk (HR) primary or secondary
MF.
Aims: To describe spleen size, and hematologic and MF-specific symptoms, during the
first 6 months of FEDR therapy after prior Tx with RUX in US clinical practices.
Methods: Adults with Int- or HR MF who initiated FEDR on or after August 16, 2019
(FEDR approval date) and with ≥90 days of follow-up were identified from community
oncology practices. Eligible patients were those treated with RUX and then FEDR, who
had spleen palpation at FEDR initiation, and who had completed ≥1 cycle of FEDR. Treating
physicians completed electronic case report forms for the selected eligible patients,
extracting patient characteristics, Tx pattern data, and clinical outcomes such as
spleen size, MF-related symptoms per the Myelofibrosis Symptom Assessment Form version
4.0 (MFSAF v4.0), hemoglobin (Hb), and platelet and white blood cell (WBC) counts,
at each visit in the first 6 months of FEDR Tx. Data were collected from February
to March 2021. Mean spleen size, symptom count, Hb, and platelet and WBC counts at
FEDR initiation vs at 3 and 6 months post-FEDR were compared via paired 2-sided t-test.
Percentage reduction in spleen size and proportion of transfusion-dependent patients
between FEDR initiation and 3 and 6 months post-FEDR were compared via 1-sided chi-square
test. Only patients assessed at FEDR initiation (with palpable spleen or complete
blood count available) and with a corresponding assessment at 3 or 6 months were included
in the comparative analysis.
Results: Among 150 eligible patients, the mean age at FEDR initiation was 68 years
(range, 36–85). Median duration of RUX Tx prior to FEDR was 7.6 months (range, 0.7–65.5).
At FEDR initiation, MF risk was categorized as International Prognostic Scoring System
(IPSS)/Dynamic IPSS HR in 43% of patients, Int-2 in 37%, Int-1 in 9%, and unknown
in 10%. A total of 74% of patients started FEDR at the recommended dosage of 400 mg
daily. Median duration of post-FEDR follow-up was 5.1 months (IQR, 3.0–17.3) for patients
initiated at FEDR 400 mg daily and 4.4 months (IQR, 3.0–14.3) for those initiating
at <400 mg daily. At data cutoff, 55% of patients were still on FEDR. The primary
reason for FEDR discontinuation was disease progression (43%). Mean duration of FEDR
Tx was 4.7 months. Of 67 patients who discontinued FEDR, 44 were deceased at data
cutoff. At FEDR initiation, 88% of patients had palpable spleen; mean spleen size
was 16.0 cm (standard deviation [SD] 5.8) above the costal margin, which decreased
to 13.2 cm (SD 7.9) at 3 months (P=0.0001) and 7.2 cm (SD 7.4) at 6 months (P=0.012).
Mean number of MF-related symptoms declined significantly at 3 and 6 months; mean
platelet count increased significantly at 6 months, from 160×109/L to 186×109/L (Table).
Overall, 65.2% of patients experienced spleen size reduction, 18.8% had no change,
and 16.0% had an increase. Mean best percentage change in spleen size was −29.2% (median
−9.8%); complete resolution of palpable spleen occurred in 21% of patients.
Image:
Summary/Conclusion: This study illustrates the real-world effectiveness of FEDR after
RUX failure in patients with Int- or HR MF. FEDR provided significant reductions in
spleen size and reported number of MF-related symptoms after 3 months of Tx, including
complete spleen responses. Greater benefits were observed with longer Tx duration.
P1061: INDIRECT TREATMENT COMPARISONS OF FEDRATINIB VERSUS NAVITOCLAX PLUS RUXOLITINIB:
EFFECT ON SPLEEN VOLUME AND SYMPTOMS IN RUXOLITINIB-EXPOSED MYELOFIBROSIS PATIENTS
P. Abraham1,*, X. Liao2, M. Chevli3, S. Smith2
1Bristol Myers Squibb, Princeton, United States of America; 2BresMed Health Solutions
Ltd, Sheffield; 3Celgene, a Bristol-Myers Squibb Company, Uxbridge, United Kingdom
Background: Myelofibrosis (MF) is a life-threatening myeloproliferative neoplasm characterized
by stem cell-derived clonal myeloproliferation, bone marrow fibrosis, anemia, and
splenomegaly. The Janus kinase (JAK) pathway is the critical pathway in its pathogenesis.
Ruxolitinib (RUX), a JAK1/2 inhibitor, was the first therapy for intermediate- and
high-risk MF approved in the United States; however, a high unmet need remains for
alternative treatment options for patients who discontinue or are no longer responding
to RUX. The efficacy and safety of fedratinib (FEDR), a JAK2 inhibitor approved by
the US Food and Drug Administration in 2019, was investigated post RUX in the single-arm
JAKARTA2 trial (NCT01523171). Clinical data from a similar population treated with
navitoclax plus RUX (NAV+RUX) have been reported. The efficacy of FEDR relative to
NAV+RUX in patients with MF previously treated with RUX has not been evaluated.
Aims: To explore the comparative efficacy of FEDR versus NAV+RUX in patients with
MF previously treated with RUX for the binary endpoints of ≥35% spleen volume reduction
(SVR) from baseline to the end of cycle 6 (EOC6; 24 weeks) and ≥50% reduction in total
symptom score (TSS) from baseline to the EOC6.
Methods: Evidence for FEDR was informed by JAKARTA2 patient-level data, and evidence
for NAV+RUX was informed by known reported evidence from the REFINE study (NCT03222609).
The suitability of these studies for indirect treatment comparison (ITC) was assessed
by considering the comparability of study design, population, intervention, and outcomes.
Given the lack of a common comparator in the identified studies, unanchored ITCs were
performed for SVR using matching-adjusted indirect comparison (MAIC) and simulated
treatment comparison (STC) methods. MAICs used effective sample size (ESS) of the
FEDR cohort as an indicator of the amount of overlap. Univariable and multivariable
regression models were used to identify potential prognostic factors to adjust for
in the ITCs. Additionally, all Dynamic International Prognostic Scoring System (DIPSS)-Plus
criteria reported were considered.
Results: A subgroup of 58 JAKARTA2 patients with an Eastern Cooperative Oncology Group
performance status (ECOG PS) score of 0 or 1 and intermediate-2 or high-risk disease
most closely aligned with the REFINE population (N=34) was used in the analyses. Baseline
mean platelet count was similar between subgroups. Across all analyses, results suggested
FEDR consistently increased the odds/risk of a spleen response compared with NAV+RUX.
The MAIC, matching on ECOG PS score, suggested that the odds of having an SVR for
patients in the FEDR group was 2.19 times that of the NAV+RUX group (95% confidence
interval [CI], 1.26 to 3.66), and the risk of having an SVR for patients in the FEDR
group was 17.6% higher (95% CI, −2.1 to 37.0) (Figure). The results from the MAIC
that additionally matched on all possible DIPSS-Plus criteria (age, hemoglobin, and
platelet count) were consistent. Results from the 2 methods (MAIC and STC) were also
consistent. For TSS reduction, the sample size (N=20) in REFINE was considered too
small to perform a meaningful ITC; however, the absolute response rates for TSS reduction
were similar across the 2 groups (29% [16/56] in the FEDR group and 30% [6/20] in
the NAV+RUX group).
Image:
Summary/Conclusion: In patients with MF previously treated with RUX, these analyses
suggest treatment with FEDR was associated with a greater proportion of patients achieving
a spleen response compared with NAV+RUX. Limited data were available for comparison
of TSS.
P1062: A REAL-WORLD EVALUATION OF THE ASSOCIATION BETWEEN ELEVATED BLOOD COUNTS AND
THROMBOTIC EVENTS IN POLYCYTHEMIA VERA: AN ANALYSIS OF DATA FROM THE REVEAL STUDY
A. T. Gerds1,*, R. Mesa2, J. M. Burke3, M. R. Grunwald4, R. Scherber5, J. Yu5, J.
Hamer-Maansson5, S. T. Oh6
1Cleveland Clinic Taussig Cancer Institute, Cleveland; 2UT Health San Antonio MD Anderson
Cancer Center, San Antonio; 3Rocky Mountain Cancer Centers, Aurora; 4Levine Cancer
Institute, Atrium Health, Charlotte; 5Incyte Corporation, Wilmington; 6Washington
University School of Medicine, St. Louis, United States of America
Background: Polycythemia vera (PV) is characterized by clonal hematopoiesis leading
to elevated peripheral blood counts and an increased risk of thrombotic events (TEs).
Advanced age and TE history form the conventional risk model used to determine TE
risk/treatment strategy. Associations between TEs and elevated hematocrit (HCT) levels
exist, but associations with white blood cell (WBC) or platelet (PLT) counts have
not been assessed consistently. The large, real-world, p
r
ospective Obs
e
rvational Study of Pts with Polycythemia
Ve
ra in US Clinic
al
Practices (REVEAL; NCT02252159) followed pts with PV treated in community or academic
centers.
Aims: This analysis evaluated associations between elevated blood counts and TEs in
pts with PV using data from REVEAL.
Methods: Eligible pts had ≥3 lab values (blood counts) post-enrollment; pts with a
post-enrollment TE but no lab value <6 mo before that TE were excluded. The association
between blood counts and TEs was assessed using a time-dependent covariate Cox proportional
hazards model. Time to first post-enrollment TE was modeled with time censored at
last known visit for pts with no TE. Each lab parameter was modeled with sex, age,
disease duration, and TE history at enrollment as baseline covariates and treatment
as a time-dependent covariate. Blood counts were included as binary time–dependent
covariates using the following thresholds: HCT >45%, WBC >11×109/L, PLT >400×109/L.
Linear interpolation was used to determine lab values between observed lab values.
Alternative thresholds for WBC (<7, ≥7 to <8.5; ≥8.5 to <11, and ≥11×109/L, and >12×109/L
with HCT controlled at ≤45%) and PLT counts (>600×109/L) were evaluated. Statistical
significance was considered at P<0.05.
Results: 2271/2510 pts were eligible (median age, 66 y [range, 22–95]; male, 54.1%).
Median disease duration was 4.1 y (range, 0–56.3), 456 (20.1%) had TE history; 52.6%
of pts received hydroxyurea. Of 106 pts who had TEs, 30 had arterial TEs (most commonly,
transient ischemic attack [n=15]) and 76 had venous TEs (most commonly, deep vein
thrombosis [n=37]).
Elevated HCT levels (>45%, hazard ratio [HR]=1.84 [95% CI, 1.234–2.749], P=0.0028),
WBC (>11×109/L, HR=2.35 [1.598–3.465], P<0.0001), and PLT counts (>400×109/L, HR=1.60
[1.088–2.359], P=0.0170) were each associated with increased TE risk (Table 1). WBC
count ≥11×109/L is associated with the highest TE risk compared with WBC count <7×109/L
(HR=2.61 [95% CI, 1.594–4.262], P<0.0001). Elevated WBC >12×109/L was significantly
associated with increased risk of TE with HCT controlled at ≤45%. PLT count (>600×109/L)
increased TE risk (HR=1.37 [95% CI, 0.763–2.468]) compared with PLT count ≤600×109/L,
but this was not statistically significant (P>0.05). In all models, advanced age,
female sex, and TE history were associated with increased TE risk.
Image:
Summary/Conclusion: This analysis of REVEAL, the largest real-world cohort of PV pts
to date, demonstrated that elevated HCT levels (>45%), WBC (>11×109/L), and PLT counts
(>400×109/L) were associated with increased TE risk. An association of elevated WBC
>12×109/L with increased risk of TE was also observed when HCT was controlled, indicating
that TE risk may be reduced by controlling WBC as well as HCT. These data support
the need to incorporate blood count into risk stratification and treatment strategies
for pts with PV in clinical practice and to move beyond the conventional risk model.
Further studies to understand the causal relationship between elevated blood counts
and TEs are warranted.
P1063: EFFICACY AND SAFETY OF PARSACLISIB-RUXOLITINIB COMBINATION THERAPY IN MYELOFIBROSIS
PATIENTS WITH LOW VS HIGHER BASELINE PLATELET COUNT: A SUBGROUP ANALYSIS OF DATA FROM
A PHASE 2 STUDY
A. Yacoub1,*, U. Borate2, R. Rampal3, H. Ali4, E. Wang5, A. Gerds6, G. Hobbs7, M.
Kremyanskaya8, E. Winton9, C. O’Connell10, S. Goel11, S. Oh12, G. Schiller13, A. Assad14,
S. Erickson-Viitanen14, F. Zhou14, N. Daver15
1University of Kansas Cancer Center, Westwood; 2Oregon Health & Science University,
Portland; 3Memorial Sloan Kettering Cancer Center, New York; 4City of Hope National
Medical Center, Duarte; 5Roswell Park Comprehensive Cancer Center, Buffalo; 6Cleveland
Clinic, Cleveland; 7Massachusetts General Hospital, Boston; 8Icahn School of Medicine
at Mount Sinai, New York; 9Emory University, Atlanta; 10University of Southern California,
Los Angeles; 11Montefiore Medical Center, Bronx; 12Washington University School of
Medicine, St. Louis; 13David Geffen School of Medicine, University of California Los
Angeles, Los Angeles; 14Incyte Corporation, Wilmington; 15University of Texas MD Anderson
Cancer Center, Houston, United States of America
Background: Despite the demonstrated efficacy of ruxolitinib (a potent and selective
JAK1 and JAK2 inhibitor), patients with myelofibrosis (MF) often exhibit inadequate
or loss of response to chronic ruxolitinib therapy, possibly due to persistent PI3K
pathway activation. Parsaclisib (INCB50465) is a potent and highly selective next
generation PI3Kδ inhibitor. In the ongoing phase 2 INCB 50465-201 trial (NCT02718300),
add on parsaclisib has shown preliminary efficacy in patients with MF who experienced
a suboptimal response to ruxolitinib. JAK inhibitors, including ruxolitinib, are associated
with thrombocytopenia; therefore, patients with low platelet count (PC) are commonly
more difficult to treat.
Aims: Here we present a subgroup analysis of efficacy and safety data from the ongoing
INCB 50465-201 study by baseline PC.
Methods: Eligible adults had primary or secondary MF with suboptimal response (palpable
spleen >10 cm below left subcostal margin [LSM]; or palpable spleen 5–10 cm below
LSM and active symptoms) after ≥6 months of ruxolitinib monotherapy (5–25 mg BID;
stable dose ≥8 weeks). Patients remained on their last stable ruxolitinib dose and
received add on parsaclisib (10 or 20 mg QD for 8 weeks; same dose QW thereafter)
or parsaclisib (5 or 20 mg QD for 8 weeks; 5 mg QD thereafter). For this analysis,
spleen volume (SV), total symptom score (TSS) assessed by Myelofibrosis-Symptoms Assessment
Form (MFSAF) v3.0 daily diary, and safety, were evaluated by baseline PC (low PC,
50 to <100×109/L; higher PC, ≥100×109/L).
Results: At data cutoff (August 27, 2020), 67 patients were enrolled (low PC, n=21;
higher PC, n=46; median age 68 years). Median prior duration of ruxolitinib use was
34.7 months for low PC compared with 14.9 months for higher PC; baseline symptoms
were worse for patients with low PC than higher PC (median [range] MFSAF-TSS, 21.4
[0.6–47] vs 10 [0–43]). Table 1 summarizes the responder analysis for SV reduction
(SVR) based on baseline PC. At week 12, slightly more patients with low PC achieved
≥10% SVR compared with patients with higher PC (50.0% vs 39.4%), whereas, at week
24, responses were similar between the 2 groups (35.2% vs 37.1%). Of patients with
≥10% SVR at week 24, 4/6 with low PC and 9/13 with higher PC were on all daily dosing
regimens. Median (range) percentage change in MFSAF-TSS was −20.5 (−56.6 to +17.1)
for patients with lower PC compared with −22.2 (−100 to +500) for patients with higher
PC at week 12; and −26.1 (−54.7 to +2.4) compared to −23.1 (−91.3 to +222.5) at week
24, respectively. In both subgroups, nonhematologic treatment-emergent adverse events
(TEAEs) were mostly grade 1 or 2. Most common (≥20%) TEAEs were dyspnea (33%), falls
(33%), peripheral edema (29%), and nasal congestion (24%) for low PC; diarrhea (28%),
nausea (24%), abdominal pain (24%), cough (20%), and fatigue (20%) for higher PC.
For patients with low PC compared to higher PC, 9/21 (43%) compared to 3/46 (7%) patients
had parsaclisib dose interruption due to thrombocytopenia. One patient with low PC
had ruxolitinib interruption due to thrombocytopenia.
Image:
Summary/Conclusion: Add-on parsaclisib showed efficacy in patients from both low and
higher baseline PC groups. Given the acceptable safety profile and efficacy of add-on
parsaclisib, MF patients with both low and higher PC may be able to benefit from parsaclisib-ruxolitinib
combination therapy. Phase 3 trials in ruxolitinib-treated and ruxolitinib-naive patients
are underway to further assess the combination of JAK and PI3K inhibitors.
P1064: CIRCULATING CYTOKINE LEVELS IN PATIENTS WITH PHILADELPHIA-NEGATIVE MYELOPROLIFERATIVE
NEOPLASMS – PROFILE AND CLINICAL CORRELATIONS – A SINGLE CENTER STUDY
G. Tsvetkova1,*, S. Mihailova2, N. Ivanov2, M. Ivanova2, E. Bekirova1, G. Balatzenko3,
A. Vlahova4, E. Hadjiev1
1Medical University - Sofia, Faculty of Medicine, Department of Internal diseases,
Clinic of clinical hematology; 2Medical University - Sofia, Faculty of Medicine, Department
of Clinical Immunology, University Hospital “Alexandrovska”-Sofia; 3Laboratory of
Cytogenetics and Molecular Biology, National Specialized Hospital for Active Treatment
of Hematological Diseases; 4Medical University - Sofia, Faculty of Medicine, Department
of General and Clinical Pathology - Sofia, University Hospital “Alexandrovska”-Sofia,
Sofia, Bulgaria
Background: Myeloproliferative neoplasms(MPNs) are a group of disorders with heterogeneous
features and often progressive clinical course. Over the past few years a remarkable
advance has been made in the knowledge on the pathogenesis and therapeutic management
of the Philadelphia(Ph)-negative MPNs. Despite the wide variety of studies in this
field, there are still unsolved issues regarding their biology, disease course, prognosis
and treatment. The improved recognition of molecular markers is not sufficient for
a comprehensive explanation of the variable clinical symptoms and complications among
these patients. The role of the chronic inflammation has been widely considered as
a co-factor, involved in the pathogenesis and progression of MPNs.
Aims: To analyze the serum levels of a panel of cytokines, associated with the mechanisms
of chronic inflammation, among patients with Ph-negative MPNs and healthy controls
and the possible correlations of these cytokines with the disease’s features.
Methods: One hundred and fifty adults were included in the study, distributed in comparable
groups, as follows: 36 patients with Essential thrombocytemia (ET), 49 patients with
Polycythemia vera (PV), 30 patients with Myelofibrosis (MF) and 35 age-matched healthy
controls. The patients with MPNs were diagnosed according to the latest WHO criteria.
The cytokines’ levels were tested in serum samples with a specific kit Cytokine Human
Panel for Luminex™. Assessments of the symptoms burden were performed with MPN-SAF-
questionnaire.
Results: Among the 11 tested cytokines we detected significantly higher serum levels
of Interleukin-2, Interleukin-5, Interleukin-6, Interleukin-8, Interleukin-18 and
GM-CSF among the MF patients (p<0.05), in comparison with the healthy controls, as
well as significantly higher levels of Interleukin-6 in the MF-group, compared to
the PV-patients. Elevated serum Interleukin-6, Interleukin-8, Interleukin-18 and GM-CSF
were observed in the ET-group versus the control group levels (p<0.05). The cytokine
profile analysis also revealed higher serum levels of Interleukin-2, Interleukin-5,
Interleukin-6 and Interleukin-18 in the patients with PV, than in the healthy subjects.
In terms of estimation of the clinical impact of the circulating inflammatory cytokines
among the analyzed MPN cohort - for the ET-patients a positive correlation was present
between GM-CSF and the fatigue severity and between Interleukin-6 levels and the weight
loss severity index, both assessed according to the MPN-SAF score. A positive correlation
was detected between Interleukin-10 levels and the bone pain burden and the degree
of weight loss for the patients with MF. Interleukin-18 values demonstrated a positive
correlation with the weight loss among the patients with PV.
Summary/Conclusion: Our single center study results can give a ground to assume the
potential role of the chronic inflammation and the pathogenetic impact of the circulating
inflammatory cytokines on the MPNs clinical features and course. The cytokine profile
of the different subgroups of MPNs is a topic of interest and can be monitored as
a potential predictor of disease progression. It can motivate future research attempts,
aiming to alter the clinical evolution of MPNs by affecting the pathogenetic mechanisms
of the inflammatory process. It can create prerequisites for future studies regarding
more effective therapeutic targets, related to both – the molecular pathways of the
mutations in MPNs and the molecular mechanisms of the chronic inflammation.
P1065: CONGENITAL ERYTHROCYTOSIS DUE TO HETEROZYGOUS VARIANTS IN THE BISPHOSPHOGLYCERATE
MUTASE GENE
M. J. van Dijk1,*, B. A. van Oirschot1, M. C. Stam-Slob1, B. van der Zwaag1, E. J.
van Beers1, J. J. Jans1, P. van der Linden2, J. M. Torregrosa Diaz3, B. Gardie4,5,6,
F. Girodon6,7,8, R. Schots9, N. Thielen10, R. van Wijk1
1University Medical Center Utrecht, Utrecht; 2Spaarne Gasthuis, Haarlem, Netherlands;
3University Hospital of Poitiers, Poitiers; 4L’institut du Thorax, Inserm, CNRS, University
of Nantes, Nantes; 5Ecole Pratique des Hautes Etudes (EPHE), Université Paris Sciences
et Lettres; 6Laboratory of Excellence GR-Ex, Paris; 7Service d’Hématologie Biologique,
Pôle Biologie, Centre Hospitalier Universitaire (CHU) de Dijon; 8INSERM U1231, Université
de Bourgogne, Dijon, France; 9Universitair Ziekenhuis Brussel, Brussels, Belgium;
10Diakonessenhuis, Utrecht, Netherlands
Background: Congenital erythrocytosis is an inherited condition characterized by increased
red blood cell mass, reflected by persistently elevated hemoglobin (Hb) and hematocrit
levels. Erythrocytosis can be primary due to an intrinsic defect of erythropoiesis,
or secondary, including germline variants in genes involved in the oxygen-sensing
pathway and Hb variants with altered oxygen affinity. A very rare cause involves variants
in bisphosphoglyerate mutase (BPGM), which regulates the red blood cell concentration
of 2,3-bisphosphoglycerate (2,3-BPG), an important modifier of Hb-oxygen affinity.
Aims: We report three European families with erythrocytosis and partial BPGM deficiency
due to heterozygosity for variants in the BPGM gene, including one novel variant.
Methods: The study was performed on adults of three families with JAK2-negative erythrocytosis.
DNA analysis concerned sequencing of relevant coding exons, including flanking splice-site
consensus sequences, of the eight major genes involved in secondary congenital erythrocytosis
(EPOR, HBB, HBA1, HBA2, VHL, EPAS1, ELGN1, and BPGM). Identified variants were evaluated
using Ensembl, gnomAD, PolyPhen-2, and SIFT. BPGM enzyme activity was determined as
described by Beutler. Quantitative analysis of 2,3-BPG was performed using liquid
chromatography tandem mass spectrometry. Hb-oxygen affinity, reflected by p50 levels
(the oxygen pressure when Hb is 50% saturated with oxygen), was analyzed using the
Hemox Analyzer (TCS Scientific).
Results: In the first family (Table 1), a novel missense variant (c.535C>T p.(Arg179Cys)
was identified in BPGM. Both the 65-year-old mother with complaints of headaches,
and her 39-year-old son with a medical history of retinal vein occlusion were heterozygous
for this variant. Allele frequency in gnomAD was 3.98-5. The predicted effect ranged
from either deleterious (SIFT) to benign (HumVar model of Polyphen-2). Subsequent
functional analysis showed low-normal BPGM activity, whereas 2,3-BPG and p50 levels
were decreased (Table 1). In the second and third families, three male individuals
were heterozygous for the c.269G>A p.(Arg90His) missense variant in BPGM. This variant
was only once previously reported, and has an allele frequency of 1.06-5. All affected
subjects had mild erythrocytosis without any additional clinical features, and showed
decreased BPGM activity, decreased 2,3-BPG levels and decreased p50 (Table 1).
Image:
Summary/Conclusion: We describe five individuals from three families in which heterozygosity
for missense variants in BPGM is associated with congenital erythrocytosis. Our findings
further strengthen the hypothesis of an autosomal dominant inheritance pattern of
BPGM variants. Our data also demonstrate the added value of functional analyses to
validate gene panel and in silico analysis results for their clinical relevance. Future
cases may help to interpret the impact of BPGM variants on hematological and clinical
phenotype.
P1066: OUTCOMES OF COVID-19 IN PATIENTS WITH MYELOPROLIFERATIVE NEOPLASMS - AN OBSERVATIONAL
COHORT STUDY FROM A LARGE ACADEMIC CANCER CENTER IN THE UNITED STATES
S. Venugopal1,*, H. Song2, C. Young2, M. A. Cutherell2, J. A. Baganz2, S. Zatorsky2,
S. E. Woodman3, N. Pemmaraju1, P. Bose1, L. Masarova1, L. Zhou1, S. Pierce1, H. Kantarjian1,
S. Verstovsek1
1Leukemia; 2Data Engineering-Analytics; 3Genomic Medicine, The University of Texas,
MD Anderson Cancer Center, Houston, United States of America
Background: Infections are one of the main causes of morbidity and mortality in patients
with Myeloproliferative Neoplasms (MPN). Several studies from all over the world has
reported on outcomes of the SARS-CoV-2 infection (COVID-19) pandemic in MPN pts but
there is no robust data on characterizing the clinical outcomes of patients with MPN
and COVID-19 in the United States (US).
Aims: Therefore, we aimed to investigate the effect of COVID-19 on MPN patients’ clinical
outcomes including the effect of MPN-subtypes, and SARS-CoV-2 vaccination on COVID-19
infection.
Methods: We conducted a single center, retrospective, observational cohort study.
Between April 2020 – December 2021, data was obtained from 388 patients flagged in
the electronic health records system with MPN and COVID-19, of which 53 MPN patients
with positive SARS-CoV-2 test were identified. COVID-19 infection was ascertained
by a positive real-time reverse transcriptase polymerase chain reaction from nasal
swab. Presenting clinical information was abstracted from review of unstructured notes
as well as structured data from the foundry. Demographic and baseline parameters were
summarized. The Kaplan-Meier method was utilized to compare the median time from date
of diagnosis to date of last follow up or death.
Results: Among 53 patients with MPN and COVID-19 infection, the median age was 58
yrs (range,21-83) with equal distribution between men and women of which 46% had a
body mass index ≥30. Most common comorbidities included hypertension (60%), and diabetes
(19%). 17 (33%) patients required hospitalization of which all (33%) of them required
oxygen supplementation. 12 pts (23%) had radiological evidence of pneumonia of which
4 pts (8%) required high flow oxygen and 3 pts (6%) required mechanical ventilation.
2 pts had new thrombosis of portal vein and coronary artery each. 10 pts (19%) received
remdesivir and 4 (8%) received tocilizumab, and 9 pts (17%) received steroids. At
a median follow up of 11.8 months, 7 pts died at a median of 24 days after COVID-19
diagnosis and the case fatality rate was 13%. Except for 1 pt,all the other 6 pts
who died had BMI≥30.
In the US, COVID-19 predominantly occurred in pts with myelofibrosis (56%), polycythemia
vera (29%), and essential thrombocythemia (15%). 54% received MPN directed therapy
of which 17 (33%) pts received ruxolitinib, and 11 pts (21%) received hydroxyurea.
At the time of infection, median WBC was 6.2 x 109/µL (range,0.3-16.3),Hb was 11.6
gm/dL (range,6.8-15.8),platelet count was 178 x 109/µL (range,2-766), and the median
neutrophil to lymphocyte ratio was 4.1. In this cohort, 22 pts (42%) were infected
prior to the availability of vaccination. 42% of the entire cohort has received vaccination
of which 45% pts were infected after first dose of vaccination. Among the 7 deaths,
6 patients had underlying myelofibrosis, and one pt had polycythemia vera, and all
deaths occurred in unvaccinated population except for one death that occurred post
vaccination. Hospitalization rate was higher in the unvaccinated (33%) pts than those
who were vaccinated (22%).
Summary/Conclusion: To date this is the largest cohort of MPN patients with COVID-19
infection in the US accounting for 14% of the MPN patients with COVID-19 in our center.
BMI≥30 appear to be an important risk factor for COVID-19 infection and mortality
in MPN pts. SARSCOV2-vaccination appear to decrease the risk of mortality and hospitalization.
Patterns of inflammatory markers, and long term follow up of recovered MPN pts with
COVID-19 will be presented in the meeting.
P1067: PHASE 2 STUDY OF ORAL THALIDOMIDE-CYCLOPHOSPHAMIDE-DEXAMETHASONE FOR RECURRENT/REFRACTORY
ADULT LANGERHANS CELL HISTIOCYTOSIS
J.-N. Wang1,*, T. Liu1, A.-L. Zhao1, B.-J. Pan2, J. Sun2, J. Li1, D.-B. Zhou1, M.-H.
Duan1, X.-X. Cao1
1Department of Hematology; 2Department of Pathology, Peking Union Medical College
Hospital, Beijing, China
Background: Langerhans cell histiocytosis (LCH) is a clonal histiocytic neoplasm with
various clinical manifestations and heterogeneous prognoses. No standard therapy is
currently available for adults with recurrent/refractory LCH.
Aims: To determine the efficacy and safety of oral thalidomide combined with cyclophosphamide
and dexamethasone (TCD) regimens in the treatment of recurrent/refractory LCH among
adult patients.
Methods: This single-center, single-arm, phase 2 study enrolled 32 recurrent/refractory
LCH patients between October 2019 and August 2021. The final follow-up date was October
31st, 2021.The TCD regimen (thalidomide 100 mg daily, cyclophosphamide 300 mg/m2 Day
1, 8, 15, and dexamethasone 40 mg Day 1, 8, 15, 22 every 4 weeks) was administered
for 12 cycles and then thalidomide alone as maintenance for 12 months. The primary
endpoint was event-free survival (EFS). Events were defined as progression during
or after TCD therapy or death from any cause.
Results: The median number of prior lines of systemic treatment was 1 (range 1-4),
and all patients received a cytarabine-based regimen as first-line or second-line
therapy. After a median follow-up of 22 months (range 5-24 months), no patient died
of all causes. The overall response rate was 87.5%, including 18 patients (56.3%)
achieving complete remission and 10 patients (31.3%) as partial remission. The estimated
24-month EFS was 64.0%. Fourteen patients (43.7%) had risk organ involvement. Patients
with risk organ involvement had a similar EFS compared to patients without risk organ
involvement (P=0.38). The common toxicities of the TCD regimen include grade 1-2 constipation
(12.5%), grade 1-2 tiredness (9.4%) and grade 2 peripheral neuropathy (12.5%).
Summary/Conclusion: Oral thalidomide, cyclophosphamide and dexamethasone are effective
and safe regimen for recurrent/refractory LCH patients, particularly for patients
with risk organ involvement.
P1068: RISK-ADJUSTED SAFETY ANALYSIS OF PACRITINIB IN PATIENTS WITH MYELOFIBROSIS
A. Vannucchi1, N. Pemmaraju2,*, B. Scott3, M. Savona4, S. Oh5, F. Palandri6, H. K.
Al-Ali7, M. Sobas8, M. F. McMullin9, V. Gupta10, A. Yacoub11, R. Mesa12, S. Buckley13,
K. Roman-Torres13, S. Verstovsek2, C. Harrison14
1Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy;
2The University of Texas MD Anderson Cancer Center, Houston, TX; 3Fred Hutchinson
Cancer Research Center, Seattle, WA; 4Vanderbilt-Ingram Cancer Center, Vanderbilt
University School of Medicine, Nashville, TN; 5Washington University School of Medicine,
St. Louis, MO, United States of America; 6IRCCS Azienda Ospedaliero-Universitaria
di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy; 7Krukenberg Cancer
Center, University Hospital Halle, Halle, Germany; 8Wroclaw Medical University, Wroclaw,
Poland; 9Queen’s University Belfast, Belfast, United Kingdom; 10Princess Margaret
Cancer Centre, Toronto, ON, Canada; 11The University of Kansas Cancer Center, Kansas
City, KS; 12UT Health San Antonio Cancer Center, San Antonio, TX; 13CTI BioPharma,
Seattle, WA, United States of America; 14Guy’s and St Thomas’ NHS Trust, London, United
Kingdom
Background: Pacritinib is a novel JAK2/IRAK1 inhibitor that has shown clinically significant
activity in patients with myelofibrosis, including those with platelet counts <50
x 109/L. Recently, JAK inhibitors have come under increased scrutiny due to specific,
emerging toxicities with drugs in this class.
Aims: This safety analysis focuses on these toxicities of interest for patients treated
with pacritinib 200 mg twice daily (BID) and best available therapy (BAT), including
ruxolitinib, on the phase 3 PERSIST-2 and phase 2 PAC203 studies. Data are presented
as risk-adjusted incidences to account for differential time at risk for adverse events
(AEs) between arms due to cross-over.
Methods: Patients treated with pacritinib 200 mg BID on PERSIST-2 and PAC203, and
those treated with BAT, including ruxolitinib on PERSIST-2, were included. Risk-adjusted
AEs, representing event rate per 100 patient-years, were calculated for the following:
overall and fatal AEs, bleeding AEs (determined by Standardized Medical Dictionary
for Regulatory Activities Query [SMQ]), cardiac AEs (by SMQ), major cardiac events
(determined by major adverse cardiovascular events [MACE] classification), infections,
thromboses, and secondary malignancies.
Results: A total of 160 patients were analyzed as the pooled pacritinib 200 mg BID
group (n=106 in PERSIST-2; n=54 in PAC203) and 98 patients in the BAT group (44 on
ruxolitinib). At baseline, the median platelet count was 57 x 109/L for the pooled
pacritinib group and BAT group; hemoglobin was 9.2 vs 9.7 g/dL and 66% vs 53% had
prior JAK2 inhibitor therapy respectively.
The rate of AEs was higher on pacritinib versus BAT, while the rate of fatal AEs was
lower (Table 1). Both bleeding and cardiac events occurred at slightly lower rates
on pacritinib compared to BAT. There were no MACE for pacritinib, whereas there were
for BAT, including in patients treated with ruxolitinib. Malignant neoplasms (excluding
worsening myelofibrosis and leukemia) occurred at similar rates on pacritinib and
BAT, though rate of non-melanoma skin cancers was lower in the pooled pacritinib group
(3/100 patient-years) versus BAT (7/100 patient-years), including ruxolitinib (11/100
patient-years). Infection occurred more frequently on pacritinib versus BAT, though
fungal and viral infections occurred less frequently, as did Herpes Zoster reactivation
(pooled pacritinib: 0/100 patient-years vs BAT: 2.4/100 patient-years, including ruxolitinib:
5.5/100 patient-years). Thrombosis occurred at similar rates on pacritinib and BAT.
Image:
Summary/Conclusion: Risk-adjusted analysis demonstrates that the safety profile of
pacritinib 200 mg BID is comparable or superior to BAT, including ruxolitinib. Pacritinib
200 mg BID may represent a full-dose therapeutic option for patients with myelofibrosis,
including those with thrombocytopenia.
P1069: RETROSPECTIVE COMPARISON OF PATIENT OUTCOMES ON PACRITINIB VERSUS RUXOLITINIB
IN PATIENTS WITH MYELOFIBROSIS AND THROMBOCYTOPENIA
C. Harrison1,*, P. Bose2, R. Mesa3, A. Gerds4, S. Oh5, J.-J. Kiladjian6, V. García-Gutierrez7,
A. Vannucchi8, C. Scheid9, M. Sobas10, S. Verstovsek2, S. Buckley11, K. Roman-Torres11,
J. Mascarenhas12
1Guy’s and St Thomas’ NHS Trust, London, United Kingdom; 2The University of Texas
MD Anderson Cancer Center, Houston, TX; 3UT Health San Antonio Cancer Center, San
Antonio, TX; 4Cleveland Clinic Taussig Cancer Institute, Cleveland, OH; 5Washington
University School of Medicine, St. Louis, MO, United States of America; 6Hôpital Saint-Louis,
Université de Paris, Paris, France; 7Hospital Universitario Ramón y Cajal, Madrid,
Spain; 8Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence,
Italy; 9University of Cologne, Cologne, Germany; 10Wroclaw Medical University, Wroclaw,
Poland; 11CTI BioPharma, Seattle, WA; 12Tisch Cancer Institute, Icahn School of Medicine
at Mount Sinai, New York, NY, United States of America
Background: Pacritinib is a JAK2/IRAK1 inhibitor for patients with myelofibrosis and
thrombocytopenia. Unlike the JAK1/2 inhibitor ruxolitinib, which is used at lower
doses in patients with thrombocytopenia, pacritinib has been studied at full dose
regardless of platelet count.
Aims: We retrospectively analyzed the phase 3 PERSIST-2 study, comparing pacritinib
with ruxolitinib, with a focus on disease symptomatology.
Methods: PERSIST-2 enrolled patients with platelet counts ≤100 x 109/L and randomized
them 1:1:1 to pacritinib 200 mg twice daily (BID), pacritinib 400 mg once daily (QD),
or best available therapy (BAT). This analysis focused on the 200-mg BID dose and
patients who received ruxolitinib prior to week 24. Endpoints included the percentage
of patients with ≥35% spleen volume reduction (SVR), ≥50% modified total symptom score
(mTSS) reduction, and improvement in disease symptoms measured by Patient Global Impression
of Change (PGIC). Safety analyses were based on all treated patients; efficacy analyses
were based on the intention-to-treat population randomized at least 22 weeks prior
to study end. The Fisher Exact test was used to describe differences in response.
Logistic regression was used to adjust for differences in baseline characteristics.
Results: Safety analysis included 106 patients on pacritinib and 44 on ruxolitinib;
efficacy analysis included 74 on pacritinib and 32 on ruxolitinib (median daily dose
of 10 mg). Baseline characteristics were similar between the groups, including median
platelet count (55 vs 61 x 109/L) and percentage receiving RBC transfusion (46% vs
43%). The following differences in baseline characteristics were accounted for in
multivariable modeling: percentage of patients with grade 3 fibrosis (58% vs 36%),
primary myelofibrosis (77% vs 50%), ≥1% peripheral blasts (45% vs 61%), and prior
JAK2 inhibitor (48% vs 73%).
Patients treated with pacritinib vs ruxolitinib achieved higher rates of SVR (22%
vs 3%, p=0.02) and numerically higher rates of mTSS response (35% vs 19%; p=0.11)
at week 24 (Figure 1A). A greater percentage of patients on pacritinib reported “much”
or “very much” improved symptoms (35% vs 16%, p=0.06; Figure 1A). Among ruxolitinib-treated
patients with an available PGIC measure at week 24, 50% reported either no improvement
or worsening symptoms, while 76% of pacritinib-treated patients reported improvement
(Figure 1B). After adjusting for imbalances in baseline characteristics, there was
no diminution of treatment effect on SVR or mTSS, and the hazard ratio for survival
on pacritinib vs ruxolitinib was 0.46 [95% CI: 0.15-1.43].
Fatal adverse events occurred at slightly lower rates on pacritinib vs ruxolitinib
(8% vs 11%). Bleeding occurred at similar rates on pacritinib and ruxolitinib (43%
vs 41%). There were low rates of herpes zoster reactivation (n=0 vs 1), fungal skin
infection (n=0 vs 1), pulmonary aspergillosis (n=1 vs 0), deep venous thrombosis (n=0
vs 1), and pulmonary embolism (n=1 vs 0) on pacritinib and ruxolitinib, respectively.
Image:
Summary/Conclusion: Pacritinib yielded higher response rates and a similar safety
profile to lower-dose ruxolitinib in patients with myelofibrosis who have moderate
or severe thrombocytopenia.
P1070: NAVITOCLAX MONOTHERAPY IN PATIENTS WITH MYELOFIBROSIS PREVIOUSLY TREATED WITH
JAK-2 INHIBITORS: SAFETY AND TOLERABILITY
V. Pullarkat1, A. Cruz-Chacon2, S. Gangatharan3, A. Melnyk4, G. A. Palumbo5, M. Bellini6,
S. K. Tantravahi7, Q. Qin8, J. Potluri8, P. Vachhani9,*
1Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National
Medical Center, Duarte, CA, United States of America; 2Hospital Centro Comprensivo
de Cancer Universidad de Puerto Rico, San Juan, Puerto Rico; 3Department of Haematology,
Fiona Stanley Hospital, University of Western Australia, Perth, Western Australia,
Australia; 4Texas Oncology-Abilene, Abilene, TX, United States of America; 5Dipartimento
di Science Mediche Chirurgiche e Tecnologie Avanzate “GF Ingrassia”, Università degli
Studi di Catania, Catania; 6ASST Papa Giovanni XXIII, Bergamo, Italy; 7University
of Utah and Huntsman Cancer Institute, Salt Lake City, UT; 8AbbVie Inc., North Chicago,
IL; 9O’Neal Comprehensive Cancer Center at UAB, Birmingham, AL, United States of America
Background: Navitoclax is an oral, small-molecule inhibitor of anti-apoptotic B-cell
lymphoma 2 family proteins (BCL-XL, BCL-2, BCL-W) that is being evaluated in the ongoing,
multicenter, multi-cohort, phase 2 trial (REFINE; NCT03222609). Previous results from
REFINE Cohort 1a indicated that addition of navitoclax to ruxolitinib resulted in
clinically meaningful outcomes with acceptable safety profile in patients with myelofibrosis
(MF) with progression or suboptimal response to ruxolitinib monotherapy (Pemmaraju
et al. J Clin Oncol. 2022).
Aims: To report preliminary safety results of navitoclax monotherapy (Cohort 2) in
patients with suboptimal response to prior Janus kinase inhibitors (JAKis).
Methods: This phase 2, open-label trial in patients with MF enrolled patients into
4 cohorts according to JAKi experience; all patients provided informed consent. Patients
in Cohort 2 had discontinued prior JAKi therapy and received navitoclax monotherapy
orally at the starting dose of 100 mg/day (QD) or 200 mg QD if baseline platelet count
was 75–150 × 109/L or >150 × 109/L, respectively. Eligible patients had previously
received JAKi for ≥12 weeks, or ≥28 days with red blood cell transfusion dependence
(≥2 units/month for 2 months) or with ≥ grade 3 adverse event (AE) of thrombocytopenia
or anemia, while on JAKi; all patients had splenomegaly. The primary endpoint was
spleen volume reduction of ≥35% (SVR35) from baseline (BL) assessed centrally at Week
24. Secondary endpoints included change in grade of bone marrow fibrosis according
to the European consensus grading system and anemia response as per International
Working Group criteria. Exploratory endpoints included duration of response of SVR35
and overall survival. Safety and AEs were monitored throughout the study.
Results: As of Oct 4, 2021, 30 patients were enrolled and received navitoclax. A majority
of patients were male (63%), median age was 68 years (range 55–84), with a median
prior ruxolitinib exposure of 100 weeks (range 11–496). Fifteen patients (50%) had
secondary MF, of which 8 patients (27%) had post–polycythemia vera MF and 7 patients
(23%) had post-essential thrombocytopenia MF (Table). Fourteen patients (47%) started
navitoclax at a dose of 100 mg QD and 16 patients (53%) started at 200 mg QD. The
median follow-up time was 4.2 months (range 0.2–15.5). Median duration of navitoclax
exposure was 9.4 weeks (range 0.1–67.1). Twenty-seven patients (90%) experienced ≥1
AE, the most common being: thrombocytopenia (n=16; 53%), diarrhoea (n=9; 30%), and
nausea (n=8; 27%). Grade ≥3 AEs were experienced by 63% of patients, with thrombocytopenia
(n=11; 37%) and anemia (n=7; 23%) being the most common. Three patients had serious
AEs of dyspnea, hypoxia, and pulmonary hypertension. Navitoclax dose reductions and
interruptions were experienced by 15 patients (50%) and 16 patients (53%), respectively.
Three patients experienced AEs leading to navitoclax discontinuation (pulmonary hypertension,
increased bilirubin, and low platelets; n=1 each). Eight patients discontinued navitoclax
due to: AE (n=3), consent withdrawal (n=1), physician decision (n=1), disease relapse
(n=1), and progressive disease (n=2). Two patients died >30 days after the last dose
of navitoclax. Details will be included in the presentation.
Image:
Summary/Conclusion: Navitoclax monotherapy in patients with MF after prior JAKi had
a similar safety profile as previously reported in Cohort 1a. Efficacy analyses are
underway to evaluate the activity of navitoclax monotherapy in patients with MF and
will be available for the presentation.
P1071: IMPACT OF ACETYLSALICYLIC ACID DOSAGE ON INCIDENCE OF THROMBOSIS AND MORTALITY
IN PATIENTS WITH MYELOPROLIFERATIVE NEOPLASMS.
A. Wild1,2,*, S. Wimmerová3
1Haematology department, Teaching F.D.Roosevelt Hospital, Banská Bystrica; 2Medical
faculty; 3Public Health Faculty, Slovak Medical University, Bratislava, Slovakia
Background: Bcr/abl – negative myeloproliferative neoplasms (MPN) are associated with
a higher risk of thrombosis and higher vascular mortality. Acetylsalicylic acid (ASA)
used in prevention decreases risk of vascular events in patients with MPN. There is
some evidence that ASA in dose 100 mg once daily results in incomplete inhibition
of a platelet function over the whole 24-hour interval in most patients with essential
thrombocythemia (ET). High on-treatment reactivity (HOTR) was corrected with dosing
ASA twice daily. Evidence of clinical consequences of these findings has been missing.
Aims: Authors compared thrombosis incidence, mortality and safety among cohorts of
MPN patients with adequate on-treatment reactivity (AOTR) on ASA dosed once daily,
twice daily and patients with HOTR.
Methods: Response to ASA in patients with MPN was evaluated by a platelet light transmission
aggregometry induced by arachidonic acid. AOTR was defined as maximal amplitude up
to 20% just before the next dose of ASA. In patients with HOTR, ASA dosing was changed
to twice daily with a dose from 50 mg step by step up to 150 mg till AOTR was reached.
A few patients didn´t change their doses at their own decision despite HOTR. The patients
were regularly followed-up. Incidence of thrombosis, adverse events and mortality
were compared among the cohorts.
Results: The platelet aggregation was tested in 69 patients with MPN on ASA. Patients
with ET, polycythemia vera (PV), primary myelofibrosis, post-PV plus post-ET myelofibrosis
and MPN not otherwise specified presented 16, 42, 36, 5 and 1%, respectively. The
adequate responses (AOTR) were found in 57 (83%), cohort 1. The insufficient responses
were found in 12 persons (17%), in 30% of patients on an enterosolvent form of ASA.
In these 12 patients the dose of ASA was changed to 2x50mg in 4 and to 2x100mg in
1 patient (cohort 2) and remained unchanged in seven (cohort 3). There were no differences
in frequency of risk factors besides higher frequency of diabetes mellitus in group
3 compared to group 1 (p=0,021). Non-adherence to regular administration of ASA was
found in 10% of patients.
The follow-up was 36,8 months. All-cause mortality was higher in the cohort 3 compared
to the cohort 1 (p=0,009), see fig. 1, but thrombotic episodes incidence not. The
cohort 2 had not different incidence of both thrombotic episodes and deaths compared
to the cohort 1. No proximal gastrointestinal bleeding and other gastropathies occurred.
There was no bleeding episode on ASA in dose of 2x50mg daily. A non-severe bleeding
dependant on any ASA dose were found.
Repeated measurement of platelet aggregation was done in 29 patients in median 49,5
months after previous measurements. There was no statistically significant difference
of adequate/high platelet reactivity on ASA between the first and the last measurement.
Image:
Summary/Conclusion: The frequency of MPN patients with higher platelet reactivity
on ASA was lower than in the most data in literature. Correction of an ASA dosage
to twice daily reaching AOTR resulted in lower mortality. Other studies are required
to confirm these data. Initial dose 2x50mg seems to be effective and safer than higher
dosage. The long-term reproducibility of platelet aggregation testing results was
preserved.
P1072: NATURAL HISTORY OF JUVENILE MYELOMONOCYTIC LEUKEMIA
W. Yang1,*, X. Zhu1
1Pediatric department, State Key Laboratory of Experimental Hematology, National Clinical
Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute
of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking
Union Medical Colleg, Tianjin, China
Background: Juvenile Myelomonocytic leukemia (JMML) is a rare clonal disease that
occurs in infants and young children, and most patients respond poorly to chemotherapy.
Hematopoietic stem cell transplantation (HSCT) is the only way to cure this disease.
The clinical heterogeneity of JMML is large, and the disease progresses rapidly in
most patients, whereas a few patients progress slowly and survive for long periods
without treatment.
Aims: This study retrospectively analyzed clinical data of untreated JMML patients
from our department. Explore their natural course of disease and prognosis factors
and provide clinical research data for screening the prognostic factors for natural
history of JMML.
Methods: Patients who were diagnosed JMML from January 2008 to December 2021 in our
center were included in this study. The clinical characteristics, bone marrow morphology,
cytogenetics, mutated genes, hypersensitivity of GM-CSF, abdominal ultrasound and
other laboratory test results were retrospectively analyzed.
Results: In this study,88 patients were diagnosed JMML from 2008 till now in our single
institute, 44 cases (50%) without treatment (including chemotherapy and HSCT) in the
whole course. The median age of onset was 13 months (range 2-48), and the incidence
was mostly male, with the male to female ratio =32: 12, median time between onset
and diagnosis was 2 (0.3-36.0) months. Among them, 15 cases (34.1%) had simple abnormal
blood image as chief complaint, and 17 cases (38.6%) had non-specific rash on physical
examination. Lymphadenopathy was found in 23 cases (52.3%) with a median subcostal
value of 3.9 (0-11) cm in liver and 5.4 (1-16) cm in spleen. The median WBC count
at initial diagnosis was 28.2 (range 9.1-128.6) ×109/L, and the hemoglobin value was
88.8 (range 52.0-153.0) g/L. Platelet count(PLT) was 44.0 (range 1.0-389.0) ×109/L,
absolute value of monocyte was 4.5 (1.3-30.5) ×109/L, absolute value of median lymphocyte
was 8.2 (2.4-48.7) ×109/L, and median fetal hemoglobin was 10 (0-88) %. Thirty-four
patients underwent cytogenetic examination, of which 31 (91.2%) had normal karyotype
and 3 patients had chromatid 7. Thirty patients underwent gM-CSF hypersensitive examination,
with a median CFU-GM-3 (0-73) BMMNC. JMML mutations were detected in 36 children,
including 6 patients with second-generation sequencing, and 30 patients with first-generation
sequencing JMML hotspot mutations, including 13 (36.1%) PTPN11 mutations, 4 (11.1%)
KRAS mutations, 9 (25.0%) NRAS mutations, and 1 (2.8%) NF1 mutations. One (2.8%) had
mutations of NRAS and PTPN11, and 8 (22.2%) had no known JMML mutants. The study was
followed up to 2022.1, with a median follow-up time of 20.6 (range 1-132) months.
Among the 44 patients, 8 cases (18.2%) were lost to follow-up, 28 cases (63.6%) died,
and 8 cases (18.2%) survived. To explore the influence of clinical characteristics
and laboratory test results on patients’ OS, the study found that PLT < 40×109/L,
spleen subcostal > 5 cm and PTPN11 were important adverse factors affecting OS. The
5-year overall survival rate of 44 children was 40%, and the 5-year overall survival
rate of those with PTPN11 mutation was significantly lower than that of other mutation
gene types.
Summary/Conclusion: The natural history of JMML was significantly different among
different mutated gene types, among which PLT, spleen size and PTPN11 mutated gene
type were independent prognostic factors affecting the natural course of JMML.
P1073: MUTATIONAL LANDSCAPE AND CLINICAL OUTCOMES OF PATIENTS WITH MYELOID AND LYMPHOID
NEOPLASMS WITH EOSINOPHILIA (MLN-EOS) AND ABNORMALITIES OF PDGFRA, PDGFRB, FGFR1,
FLT3 AND JAK REARRANGEMENT
Y. Zhang1,*, L. Nguyen2, C. Lu3, E. Wang4, M. Lauw4, S. Ball1, N. Dong1, L. Moscinski5,
O. Chan6, S. Yun6, D. Sallman6, L. Sokol6, B. Shah6, J. Lancet6, R. Komrokji6, A.
Kuykendall6, E. Padron6, L. Zhang6
1Moffitt Cancer Center; 2Department of Pathology, James A Haley Veterans’ Hospital,
Tampa; 3Department of Pathology, Duke University School of Medicine, Durham; 4Department
of Pathology, University of California San Francisco, San Francisco; 5Department of
Pathology; 6Department of Hematologic Malignancies, Moffitt Cancer Center, Tampa,
United States of America
Background: To recognize the growing list of recurrent genetically defined eosinophilia
driven by constitutively active tyrosine kinase fusion genes, WHO created a provisional
entity of myeloid/lymphoid neoplasms with eosinophilia (MLN-Eo) with rearrangement
of PDGFRA, PDGFRB, FGFR1, or PCM1- JAK2.
Aims: We aim to describe the mutational landscape and outcome with upfront targeted
tyrosine kinase inhibitor (TKI) therapy in patients (pts) with MLN-Eo.
Methods: We retrospectively reviewed clinical and molecular data on ptss with MLN-Eos
with PDGFRA, PDGFRB, FGFR1, JAK2 or FLT3 rearrangement at Moffitt Cancer Center, Duke
Cancer Center, and UCSF Comprehensive Cancer Center. The next generation sequencing
of the bone marrow or peripheral blood was either done using our in-house targeted
54 gene myeloid panel or the commercially available panel, FoundationOneTM Heme Genomic
profile.
Pts were divided into two categories based on morphology at diagnosis: chronic and
blastic phase disease. Median time to blastic phase and overall survival (mOS) were
calculated using Kaplan Meier method and compared with log-rank test. Hazard ratio
was calculated using the Cox proportional hazards (PH) model.
Results: A total of 41 pts were included in the analysis. Thirteen patients had available
comprehensive NGS results. 23 pts had PDGFRA fusion (21) or activating mutations (2);
4 had PDGFRB; 8 had FGFR1; 2 had JAK2, PCM-JAK2, and BCR-JAK2; 4 had FLT3, ETV6-FLT3
(3) and FLT3-TRIPP (1). Twelve (92%) patients had at least one other coexisting mutation.
The mutations in at least two patients included TET2, RUNX1, TP53, and PTPN11 (Fig
1). Pathogenic PTPN11 mutation was present in all activating PDGFRA mutation but absent
in other cases.
Among 23 patients in chronic-phase disease 14 patients (61%) received the corresponding
TKI to their fusion in the frontline setting. None transformed into blastic-phase
disease. Six patients in the de novo blastic phase received the upfront TKI alone
or combined with chemotherapy. All achieved complete remission (CR) and remained alive
at a median follow-up of 44 months.
In the OS analysis, upfront TKI use was associated with longer survival (HR 0.065;
95%CI, 0.008-0.50, p=0.008) in univariate analysis. The improved OS emained significant
(HR 0.043, 95% CI 0.003-0.63, p=0.021) in multivariate analysis when adjusted for
sex, age, hypereosinophilia in the blood, PDGFRA/B fusion status, complex cytogenetics
at diagnosis, the blastic-phase at diagnosis, and alloHSCT.
Image:
Summary/Conclusion: Our study showed the high frequency of secondary somatic mutations
in MLN-eos and described the mutational landscape similar to other myeloid malignancies.
Targeted TKI therapies in the upfront setting are associated with excellent outcomes
in patients with MLN-Eos. The elucidation of potential mechanisms needs further exploration.
P1074: PRIMARY REFRACTORY HODGKIN’S LYMPHOMA: A MOROCCAN MONOCENTRIC RETROSPECTIVE
STUDY
M. Ababou1,*, A. Hammani1, A. Siham1, W. Elfaria1, E. M. Mahtat1, S. Jennane1, H.
EL Maaroufi1, K. Doghmi1
1Clinical Hematology, Military hospital Mohammed v, Rabat, Morocco, Rabat, Morocco
Background: Primary refractory Hodgkin’s lymphoma (PRHL)is defined as disease progression
during initial first-line therapy or early relapse of less than 3 months [1], this
entity is a therapeutic challenge for hematologists, as the response rate to second-line
therapy remains lower (51% vs. 83%, P < 0.0001)[2]
Aims: Compare different parameters between patients with PRHL and patients with non-PRHL
to reveal factors that could predict the primary refractory character of our patients.
Evaluate the evolution in terms of therapeutic response and survival of our patients
with PRHL compared to non-PRHL
Methods: This is a monocentric retrospective descriptive study that collected all
the files of patients followed for Hodgkin Lymphoma at the clinical hematology department
of the Military Hospital of Instruction Mohammed V in Rabat between January 2010 and
December 2020. Their epidemiological, clinical and prognostic characteristics were
recorded, and the efficacy and tolerance of the therapies used were evaluated. We
used the t-student, Mann-Whitney, χ2 tests (or Fisher’s exact test) to compare the
differences between PRHL and non-PRHL patients. To assess the difference in survival
between the two groups, we used the Log-rank test on Kaplan-Meier survival curves.Treatment
outcomes were measured in terms of response rate, overall survival (OS) and progression-free
survival (PFS). Statistical analyses were performed using JAMOVI software (version
1.6)
Results: Classical Hodgkin lymphoma was diagnosed in 122 patients. 27 patients were
primary refractory (22.1%). Our comparison between the group of PRHL and non-PRHL
patients in terms of age, sex, time to consultation, performance status (PS) score,
clinical signs at admission, histological subtype, presence of Bulky mass on chest
X-ray, the Ann Arbor stage and the prognostic risk group did not show any significant
difference, which makes the identification of primary refractory patients in pre-therapy
a difficult challenge. In our serie 83(68%) patients were re-evaluated at 2 courses
by PET-Scan (Positron Emission Tomography, 45(54.3%) patients were in Complete Remission
(CR), 34(41%) in Partial Remission (PR). Comparing PRHL and non-PRHL patients according
to the response at 2 courses, a highly significant difference was observed (p=0.001).
Using a univariate logistic regression model, we found that a PR at 2 courses was
more likely to be PRHL than a complete response (OR= 4.95; CI95% [1.55-15.78]; p=0.007).
At the end of first-line treatment, 77.1% of all patients achieved complete remission,
11.5% achieved partial response, 1 patient had a stable response and 10.6% were in
progression. The overall response at the end of treatment for PRHL patients was 44.4%
vs 93.6% for the non-PRHL group (p<0.001). 10.7% of patients relapsed within a median
of 23 months. With a median follow-up of 65.5 months, the OS and PFS of all patients
at 5 years were 91.2% and 90.6% respectively. Comparing primary refractory and non-refractory
patients, the OS at 5 years was 73% [56.2-94.8] vs 96.5% [92.7-100] (p<0.0001) (Figure
1).
Image:
Summary/Conclusion: Primary refractory Hodgkin’s lymphoma remains a form with a poor
prognosis, the overall 5-year survival of this form in our series is 73% compared
to 96.5% for the non-PRHL patients. PET-2 may be a predictive step for this form,
studies are needed to identify the place of immunotherapy in the management of these
patients
P1075: EVALUATION OF CYTOKIN GENE POLYMOPHISMS AND GENE EXPRESSIONS IN PATIENTS WITH
HODGKIN LYMPHOMA
H. Haydaroglu Sahin1,*, D. D. Demirbas1, D. M. Akkurd1, V. Okan1
1Haematology Department, Gaziantep University Sahinbey Research and Practice Hospital,
Gaziantep, Turkey
Background: In Hodgkin Lymphoma (HL), Reed-Sternberg (RS) cells play a role in the
etiopathogenesis and clinical course of the disease and that is thought to be the
result of expression of cytokines.
Aims: In this study; it was aimed to evaluate the role of gene polymorphisms and gene
expression of five cytokines (tumour necrosis factor alfa (TNF-α), transforming growth
factor beta-1 (TGF-β1), interferon gamma (IFN-γ), interleukin-6 (IL-6), and interleukin-10
(IL-10)) in the etiopathogenesis of the disease, clinical parameters and prognosis
in HL patients.
Methods: A total of 70 patients and 70 control group who were diagnosed, treated and
followed up for HL in the Hematology clinic of Gaziantep University Faculty of Medicine
were included in the study. Cytokine gene polymorphisms and expression levels were
analyzed by DNA isolation from peripheral blood of the patient and control groups
and by using of Sequence Specific Primary polymerase chain reaction (PCR-SSP) method.
Results: TNF-α (-308) gene variant GA genotype (high expression), IFN-γ (+874) gene
variant TT genotype (high expression), IL6 (-174) gene variant GG genotype (high expression),
TGF-β1 (T10/C10) gene variant TT genotype, TGF-β1 (C25/G25) gene variant GC genotype,
IL10 (-1082) gene variantGG genotype, IL10 (-819) gene variantCC genotype and IL10
(-592) gene variantCC genotype was significantly higher in HL patients compared to
healthy controls (respectively; OR= 0.3636, p= 0.0259; OR= 0.3164, p= 0.0349, OR=
0.4189, p = 0.0299, OR= 0.4329, p= 0.0257, OR= 0.4889, p<0.001, OR= 0.4189, p= 0.0299,
OR= 0.3951, p= 0.0193, OR= 0.3951, p= 0.0193). While IL6 (-174) G allele (p = 0.0302),
TGF-β1 (T10 / C10) T allele (p = 0.0224), IL10 (-1082) G allele (p = 0.0232), IL10
(-819) C allele (p = 0.0156), IL10 (-592) C allele (p = 00156) was higher in HL patients
compared to the control group, expression of IL6 (-174) C allele (p = 0.0302), TGF-β1
(T10 / C10) C allele (p = 0.0224), IL10 (-1082) A allele (p = 0.0232), IL10 (-819)
T allele, IL10 (-592) A allele (p = 0.0156) was found to be low. In haplotype analysis;
TGF-β1 (T / C10, C / G25) gene “TCGC, CCGG, TTGC” (moderately increased plasma TGF-β1
level) and IL10 (-1082, -819, -592) gene “GCC GCC” (high IL10 plasma level) haplotype
variants was significantly higher in HL patients than in the control group (respectively;
OR= 0.3406, p= 0.0161; OR= 0.4149, p= 0.0299).In multivariate analysis; Bone marrow
involvement (p <0.001) and carrier of IL6 (-174) gene polymorphism CC variant genotype
(p= 0.02) were detected as an independent risk factor associated with poor prognosis
on 5-year PFS.
Summary/Conclusion: The results of the study suggests that TNF-α (-308) GA genotype,
IFN-γ (+874) TT genotype, IL6 (-174) GG genotype, TGF-β1 (T10 / C10) TT genotype,
TGF-β1 (C25 / G25) GC genotype, IL10 (-1082) GG genotype, IL10 (-819) CC genotype,
IL10 (-592) CC genotype, TGF-β1 (T / C10, C / G25) gene “TCGC, CCGG, TTGC” and IL10
(-1082, -819, -592) gene “GCC GCC” haplotype may be a risk factor for HL development.
P1076: A RETROSPECTIVE ANALYSIS ON OLD AND EMERGING PROGNOSTIC FACTORS IN CLASSICAL
HODGKIN’S LYMPHOMA IN THE PET-GUIDED ERA
A. Cellini1,*, C. A. Cavarretta1, S. Pravato1, M. Pizzi2, M. Gregianin3, F. Crimì4,
F. Piazza1, L. Trentin1, A. Visentin1
1Hematology and Clinical Immunology Unit, Department of Medicine; 2Surgical Pathology
and Cytopathology Unit, Department of Medicine, University of Padua, Padua; 3Nuclear
Medicine Unit, Veneto Institute of Oncology IOV-IRCCS, Castelfranco; 4Institute of
Radiology, Department of Medicine, University of Padua, Padua, Italy
Background:: Hodgkin’s Lymphoma (HL) has been commonly recognized as a chemo-sensitive
and potentially curable neoplastic disease. However, some patients are refractory
or relapse after frontline therapy, and their prognosis is severely impacted by such
events. Baseline prognostic factors that can reliably identify patients with high-risk
disease are currently lacking, as classical prognostic factors have lost their meaning
in the modern era, where a PET-guided approach and novel therapies are being incorporated
in the treatment of HL. Such therapeutic options, however, need to be directed to
the right subset of patients to avoid overtreatment, unwanted toxic effects, or financial
burden.
In recent years lymphocyte to monocyte ratio (LMR) and neutrophil to lymphocyte ratio
(NLR) have been evaluated as potential outcome predictors in patients affected by
HL. These factors, however, need to be further evaluated to be included in prognostic
scores that reliably predict outcomes in HL patients treated in the current era.
Aims: This study’s aim was to identify potential predictors of worse overall survival
(OS) and progression free survival (PFS) in patients affected by HL treated with a
PET-adapted chemotherapeutic approach.
Methods: We included HL patients diagnosed at the University Hospital of Padova between
2004 and 2020. All patients were treated with ABVD, were staged with PET-TC at baseline,
and evaluated after 2 cycles (interim, iPET) and at the end of therapy. iPET positive
patients (Deauville score 4-5) were intensified with escalated BEACOPP. The number
of cycles and the addition of radiation therapy as mandated by stage and local policy.
Low LMR was considered as ≤2.1 and high NLR as ≥6. Survival curves were compared with
Log-rank test, Cox proportional hazards test was used for multivariate analysis. 95%
Confidence intervals (CI) were also reported.
Results: Of the 261 patients analyzed, 101 (39%) had early-stage and 160 (61%) had
advanced-stage (IIB or higher) HL. Median age was 32 years, 127 (49%) of patients
were male, 134 (51%) presented with B-symptoms and 91 (35%) had bulky disease.
After a median follow up of 59 months the 5-year OS and PFS for the whole population
were 93.4% (CI 88.7–96.1) and 73.6% (CI 67.3–78.9), respectively. In univariate analysis
stage ≥III, age ≥65 years, B symptoms, hemoglobin <105 g/L, low LMR, high NLR were
associated with a shorter PFS (p<0.05), while stage ≥III, advanced age, low LMR and
high NLR were associated with worse OS (p<0.05). In multivariate analysis, only stage
≥III significantly predicted worse PFS (HR 2.04; 1.13–3.69), while age ≥65 years (HR
5.38; 1.49–19.40) and high NLR (HR 4.14; 1.15–14.87) were associated with decreased
OS.
Focusing on the 160 advanced stage patients’ subgroup, stage ≥III and low LMR predicted
a decreased PFS both in univariate and multivariate analysis with a HR of 2.14 (CI
1.30–4.08) and 2.14 (CI 1.10-4.31), respectively. Age ≥65 years and high NLR were
associated with a worse OS both in univariate and multivariate analysis, with a HR
of 7.48 (CI 2.14–26.17) and of 6.57 (CI 1.70–25.48), respectively.
Summary/Conclusion: In this study, we provide evidence that current practice, based
on a PET-guided therapy intensification, is unsatisfactory, since the survival of
stage III-IV is worse than the one reached by earlier stage patients. We confirmed
the reliability prognostic efficacy of NLR and LMR. Whether advanced stage patients
with low LNR and high NLR might benefit from a frontline brentuximab or nivolumab-based
therapies is unknown, and deserve further investigation.
P1077: HODGKIN LYMPHOMA IN PATIENTS WITH HIV: NATIONAL RETROSPECTIVE MULTICENTER STUDY
A. Chekalov1,*, M. Popova1, I. Tsygankov1, Y. Rogacheva1, N. Volkov1, A. Beynarovich1,
K. Lepik1, M. Demchenkova2, T. Schneider3, Y. Kopeikina3, V. Potapenko4, I. Zyuzgin5,
M. Kolesnikova6, T. Pospelova6, E. Zinina7, A. Myasnikov8, K. Kaplanov9, T. Ksenzova10,
E. Pavlyuchenko11, N. Mikhaylova12, V. Baykov13, A. Kulagin13
1Department of Oncology, Hematology and Transplantation for teenagers and adults,
Raisa Gorbacheva Memorial Research Institute of Children Oncology, Hematology and
Transplantation, Saint Petersburg; 2Irkutsk Regional Cancer Center, Irkutsk; 3Leningrad
Regional Clinical Hospital, Saint Petersburg; 4Municipal educational hospital №31,
Saint-Petersburg; 5National Medicine Research Center of oncology named after N.N.
Petrov, Saint Petersburg; 6Municipal Clinical Hospital No. 2 of Novosibirsk Region,
Center of Hematology, Novosibirsk; 7Surgut’s Clinical Hospital, Surgut; 8Republican
hospital named under V.A. Baranov, Petrozavodsk; 9Volgograd Regional Clinical Oncology
Dispensary, Volgograd; 10Regional Clinical Hospital, Tumen; 11II Mechnikov North-Western
State Medical University; 12Department of Oncology, Hematology and Transplantation
for teenagers and adults, Raisa Gorbacheva Memorial Research Institute of Children
Oncology, Hematology and Transplantation; 13Raisa Gorbacheva Memorial Research Institute
of Children Oncology, Hematology and Transplantation, Saint Petersburg, Russia
Background: Patients with HIV infection have a significantly higher risk of developing
cancer than the general population. In the era of antiretroviral therapy (ART) the
cancer risk and mortality have been decreased, however, the risk of developing Hodgkin
lymphoma (HL) increased. The ART allows treating HIV-infected patients with lymphomas
with protocols for patients in the general population. Withal the data on the results
of the treatment HL in patients with HIV controversy.
Aims: To study epidemiology and evaluate the results of the treatment of HL in patients
with HIV in national multicenter study.
Methods: The study included 45 patients with HL in patients with HIV who received
treatment in 9 Russian centers from 2007 to 2021. The median follow-up was 9 months
(1-129). Patients and treatment characteristics were analyzed. Overall survival (OS)
and progression-free survival (PFS) were calculated within two years from the diagnosis
using the Kaplan-Meier method.
Results: The median age was 39 years (25-66), men - 25 (55.6%), women - 20 (44.4%).
Histological variants of HL in most cases were represented by nodular sclerosis (56%)
and mixed-cell variant (41%). The advanced stage of the disease (3-4 Ann Arbor) was
observed in 72.7% of patients, B-symptoms at the onset of the disease - 68.2%. The
majority of patients (97.7%) received ART at the diagnosis of HL. The median number
of CD4+ cells/µl at the onset of HL was 352.8 (50-692) cells/μl. General somatic status
at the start of chemotherapy ECOG 0-1 - 34 (82.9%), ECOG≥2 - 7 (17.1%). As the first
line of therapy, patients with localized stages of HL received ABVD (75%) and BEACOPP
(25%), with advanced stages - ABVD (61.3%) and BEACOPP (38.7%). The median courses
of first-line therapy were 4 (1-10). Radiation therapy in first-line therapy was performed
in 4 patients (8.9%). The structure of response to first-line therapy were complete
response - 51.4%, partial response - 25.7%, disease stabilization - 2.9%, disease
progression - 20%. Fourteen and 8 patients received second and third-line therapy,
respectively. Autologous hematopoietic stem cell transplantation was performed in
6 patients - the only 42.8% of patients with relapsed / refractory HL. OS in the study
group was 81%, PFS - 38% (median PFS - 23 months). The level of CD4+ cells at the
onset of HL less than 250/µl was associated with a statistically significant worsening
of OS during 1 year (50% vs 100%, p=0.014). Factors such as gender, age, stage of
the disease, ECOG status, the presence of B-symptoms at the onset of the disease,
and the treatment regimen did not statistically significantly affect the two years
OS and PFS from the diagnosis of HL in patients with HIV.
Summary/Conclusion: The national multicenter study allowed characterization of HL
in patients with HIV and evaluation of the efficacy of first-line therapy, which was
found to be lower than in the general population. The level of CD4+ cells was the
only factor that affect 2-year OS. The obtained data can form the basis for further
prospective studies aimed at improving the results of HL treatment in HIV-infected
patients.
P1078: PATIENTS WITH HODGKIN LYMPHOMA DEVELOP ADEQUATE HUMORAL SEROLOGICAL RESPONSE
TO VACCINATION WITH TWO DOSES OF BNT162B2 AND THEIR IGG ANTIBODY LEVELS MARKEDLY INCREASE
AFTER A THIRD VACCINE DOSE
E. Dann1,2,*, T. Inbar1, T. Mashiach1, J. Eisa1, S. Ringelstein-Harlev1, N. Horowitz1
1Rambam Health Care Campus; 2The Ruth and Bruce Rappaport Faculty of Medicine, Technion,
Israel Institute of Technology, Haifa, Israel
Background: In patients (pts) with hematological malignancies, COVID-19 is considered
to be associated with a high risk of severe morbidity and mortality. While anti-COVID-19
vaccination of such pts has become the standard of care, pts undergoing lymphodepleting
therapy fail to generate protective serological response due to either the nature
of their underlying disease or exposure to therapy.
Aims: This study aimed to assess serological response to vaccination with BNT162b2
(Pfizer) as well as COVID-19-related morbidity and mortality in Hodgkin lymphoma (HL)
pts.
Methods: The above vaccine was available in Israel from January 2021 and all pts with
hematological malignancies were recommended to undergo vaccination with 2 doses of
this vaccine, injected 21 days apart. Six months later a 3rd dose was recommended
and in another 3 months a 4th dose was available for pts at risk. Serology tests were
performed at least 2 weeks after the 2nd vaccination. The SARS-CoV-2 IgG II Quant
(Abbott©) assay was used to measure levels of IgG antibodies (Abs) against the SARS-CoV-2
spike protein. A result was considered positive if the IgG level was ≥150 AU/ml, which
was defined as an adequate serological response.
Results: The current non-interventional single-center study evaluated the outcome
of 55 HL pts (median age 46 years, 53% females); 51% of pts had advanced HL. Study
participants received 1-9 lines of therapy (median 1 line). Six pts had COVID-19 prior
to vaccination, 49 were vaccinated: 9 with 2 doses, 36 with 3 doses and 4 with 4 doses
of BNT162b2. Following initial 2 vaccine doses and after a 3rd dose Ab levels >150
AU/ml were developed in 85% and 89.5% of pts, respectively. At a median of 95 days
post-2nd vaccination, Ab levels were 2024 (1-29400) and 4 (0-7539) in 48 patients
with no background disease versus 7 pts treated with lymphodepleting drugs or having
a background disease, respectively. During the follow-up, 5 vaccinated pts were diagnosed
with COVID-19 when the Delta variant was prevalent and 9 - during the Omicron wave.
Notably, similar Ab levels were observed in those infected with Omicron and in non-infected
pts, reflecting the genetic drift of this variant. A further analysis was performed
to compare findings in a subgroup of 7 pts who had an additional background disease
along with HL, such as chronic lymphocytic leukemia, s/p kidney transplantation, solid
tumor, or those who were heavily pretreated, including therapy with bendamustine,
versus the values observed in the rest 48 pts. The median age in the former subgroup
was 58 (31-80) years, which was significantly older than in the remaining pts [median
45 (18-78) years] and median Ab levels were 4 (0-7539) AU/ml and 2024 (1-2940) AU/ml,
respectively. Notably, after the 3rd vaccination, the median Ab level in both groups
was 7000 AU/ml.
Summary/Conclusion: The results of the current study show that at least 85% of HL
pts develop a high titer of anti-spike antibodies after vaccination with 2 BNT162b2
doses. These titers substantially increased post the 3rd vaccine dose. Only a minority
of HL pts who had additional background diseases or were heavily pretreated, failed
to develop an adequate serological response; however, some of them had high Ab titers
post-3rd and 4th vaccinations. In this study, morbidity and mortality rates of HL
pts infected with COVID-19 were lower than those reported in pts with other lymphoma
types.
P1079: COMPARISON OF NOVEL SALVAGE REGIMENS AND TRADITIONAL SALVAGE CHEMOTHERAPY IN
RELAPSED AND REFRACTORY CLASSIC HODGKIN LYMPHOMA
D. Ermann1,*, V. Vardell2, E. Zacholski3, A. Fegley3, D. Modi4, K. Fedak4, S. Sundaram5,
S. Rajeeve5, G. Goyal6, H. Hatic7, P. Torka8, S. Ba Aqeel8, A. Borogovac9, K. MacDougall9,
S. Kothari10, A. Kress11, E. Travers12, N. Chilakamarri13, E. Brem14, L. Fitzgerald15,
C. Wagner16, B. Hu15, D. Stephens15, H. Shah15
1Hematology/Oncology, Huntsman Cancer Institute, University of Utah; 2Internal Medicine,
University of Utah, Salt Lake City; 3Clinical Pharmacology, Virginia Commonwealth
University, Richmond; 4Hematology/Oncology, Karmanos Cancer Institute, Detroit; 5Hematology/Oncology,
Tisch Cancer Institute, Mount Sinai, New York; 6Hematology; 7Hematology/Oncology,
University of Alabama, Birmingham; 8Hematology/Oncology, Roswell Park Comprehensive
Cancer Center, Buffalo; 9Hematology/Oncology, University of Oklahoma, Oklahoma City;
10Hematology; 11Hematology/Oncology, Yale Comprehensive Cancer Center, New Haven;
12Hematology, University of Kentucky, Lexington; 13Internal Medicine, Pomona Valley
Medical Center, Pomona; 14Hematology/Oncology, University of California, Irvine; 15Hematology;
16Clinical Pharmacology, Huntsman Cancer Institute, University of Utah, Salt Lake
City, United States of America
Background: Traditional salvage chemotherapy followed by autologous stem cell transplant
(ASCT) is the standard second line approach for treatment of relapsed/refractory (r/r)
Classical Hodgkin Lymphoma (cHL) and can cure approximately 50-60% of patients (pts).
Novel agents such as brentuximab vedotin (BV) and check-point inhibitors (CPIs) have
high response rates in the r/r setting and have recently been used as salvage regimens
to induce remissions before ASCT. Limited data exists comparing efficacy of these
two approaches.
Aims: Our aim was to compare outcomes of pts receiving salvage chemotherapy (CT) as
opposed to novel treatments (NT) for their first salvage regimen for r/r cHL.
Methods: Adult pts with r/r cHL who received their first salvage regimen (SR1) between
January 2018 and June 2020 were retrospectively identified across 9 academic centers
in the United States. Baseline characteristics were compared across CT and NT cohorts
based on SR1. Endpoints were to compare complete response (CR), overall response rate
(ORR), and event free survival (EFS) between the CT and NT cohorts. ORR was defined
as CR+ partial response (PR). Events were defined as stable disease (SD) or progressive
disease (PD) after SR1, less than a complete response (CR) at 90 days post-ASCT, PD
after ASCT, or death.
Results: In total, 120 pts were identified with a median age of 33 years (range 18-85).
68% had advanced disease and 13% were early stage unfavorable (NCCN) at diagnosis.
88% received ABVD based front-line regimen. Many pts had poor prognostic characteristics
including 43% with primary refractory disease, 52% with relapse <12 months from diagnosis,
38% with extranodal disease, and 26% with B-symptoms at time of relapse (Table 1).
65% of pts (n=78) received CT and 35% (n=42) received NT for SR1. 90% of CT pts received
Ifosfamide, Carboplatin, and Etoposide (ICE) as SR1. Regimens used for NT treated
pts included BV + Bendamustine (43%), BV alone (36%), BV + CPI (12%), BV + CT (5%),
and other CPI (4%). No significant difference was found in the ORR and CR rates according
to pts treated with CT vs. NT of 67% vs. 69% and 47% vs. 55%, respectively. Of the
104 patients who received ASCT, 88% received CT and 83% received NT for SR1. At a
median follow-up of 32 months, 1-year EFS was 57% vs 69% and 2-year EFS was 56% vs
66% between CT and NT cohorts (p=0.25) (Figure 1).
All pts who progressed after CT for SR1 (n=31) received NT for SR2; whereas 73% (n=11)
of those progressing after NT for SR1 received CT for SR2. ORR for CT in SR2 was 91%
vs 70% for NT (p=.48). The most common SR2 CT regimen was ICE (82%), and NT regiment
for SR2 was BV alone (40%), BV + Bendamustine (31%), BV + CPI (23%), and other CPI
(6%). 79% vs 89% remained progression-free post-ASCT at their last follow-up after
receiving CT vs. NT at last salvage respectively (p=0.15).
Of the 16 pts who did not proceed to ASCT, 7 died within an average of 19.5 months
from the receipt of SR1. Interestingly, pts who did not proceed to ASCT had a significantly
improved 2-year EFS if they received NT (n=7) at SR1 (71% vs. 13%) when compared to
CT at SR1 (n=9) (p=0.03).
Image:
Summary/Conclusion: There was a numerical trend towards better CR and EFS for novel
therapy compared to traditional chemotherapy for first salvage, however the outcomes
were not statistically significant. This demonstrates that CT is still a useful salvage
therapy that can effectively get patients to an ASCT for curative intent. Prospective
studies comparing novel therapy to traditional chemotherapy for salvage are warranted.
P1080: COMPARISON OF NIVOLUMAB 40 MG EFFICACY VERSUS 3 MG/KG IN PATIENTS WITH RELAPSED
AND REFRACTORY CLASSIC HODGKIN LYMPHOMA
L. Fedorova1,*, K. Lepik1, N. Mikhailova1, E. Kondakova1, Y. Komarova1, M. Popova1,
P. Kotselyabina1, E. Borzenkova1, V. Baykov1, I. Moiseev1, A. Kulagin1
1RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russia
Background: Nivolumab 3 mg/kg was shown to be effective in patients with relapsed
and refractory classic Hodgkin lymphoma (r/r cHL). However, previously obtained data
on pharmacokinetics, as well as financial toxicity of therapy, raised the issue of
studying necessity of efficacy of reduced doses of PD-1 inhibitors. Efficacy of nivolumab
40 mg has previously been demonstrated in patients with r/r cHL in a prospective phase
2 study (Lepik KV et al., 2020). Continued accumulation of experience in the use of
low-dose PD-1 inhibitors in patients with r/r cHL is required to support previous
data.
Aims: To compare the efficacy of nivolumab (Nivo) 40 mg therapy with 3 mg/kg for patients
with r/r cHL.
Methods: The Nivo40 trial (NCT03343665) expanded prospective cohort of patients (group
1, n=50) treated with Nivo 40 mg was compared with the retrospective group 2 (n=116)
of patients treated with Nivo 3 mg/kg. Patients characteristics are demonstrated in
the Table 1.
The response was evaluated every 3 months by PET-CT using LYRIC criteria. Adverse
events (AE) were analyzed by NCI CTCAE 4.0.3.
Overall response rate, progression-free survival (PFS) and overall survival (OS) were
compared between group 1 and 2. During the survival analysis the PFS was censored
by the time of additional therapy initiation.
Variable
Nivo 40 mgN=50
Nivo 3 mg/kgN=116
p
Median age, years (range)
36 (20-54)
38 (14-65)
0.129
Male/female, n (%)
17/33 (34/66)
56/60 (48/52)
0.089
Primary chemoresistance, n (%)
38 (76)
74 (64)
0.124
Early relapse, n (%)
7 (14)
14 (12)
0.731
Prior autologous stem cell transplantation, n (%)
17 (34)
44 (38)
0.630
Prior brentuximab vedotin, n (%)
18 (36)
62 (53)
0.039
Therapy lines before Nivo therapy, n (range)
4 (1-8)
5 (2-10)
0.011
B symptoms at Nivo therapy initiation, n (%)
26 (52)
71 (61)
0.269
Disease stage at Nivo therapy initiation, n (%)
2
8 (16)
11 (9)
0.294
3
5 (10)
7 (6)
4
37 (74)
97 (84)
Progression at Nivo therapy initiation, n (%)
46 (92)
92 (79)
0.272
ECOG status at Nivo therapy initiation, n (%)
0-1
31 (62)
70 (60)
0.758
2
14 (28)
29 (25)
3
4 (8)
14 (12)
4
1 (2)
2 (2)
Results: Median follow up was 44 (11-55) months in group 1 and 60 (6-70) months in
group 2. Median Nivo cycles was 19 (2-49) and 20 (1-32) respectively. The best response
to Nivo therapy was detected at 6 (2-24) and 6 (1-27) cycles respectively.
Overall response rate was 66% in group 1 and 67% in group 2. The structure of response
in group 1 was: complete response (CR) in 38% of patients, partial response (PR) in
28%, stable disease (SD) in 6%, indeterminate response (IR) in 22% and progressive
disease (PD) in 6%; in group 2: CR in 34%, PR in 33%, SD in 5%, IR in 20% and PD in
8%. Median OS was not achieved in both groups, 3-year OS was 97,8% and 96,5% respectively
(p=0.356). Median PFS was 21,9 months (95%CI: 16,75-27,05) in group 1 and 18,8 months
(95%CI: 13,44-24,16) in group 2, 3-year PFS was 25,6% and 27% respectively (p=0.356).
Additional therapy after Nivo monotherapy was started in 78% of patients after Nivo
40 mg and in 84% after Nivo 3 mg/kg. Allogeneic stem cell transplantation after Nivo
therapy was performed in 5 (10%) patients in group 1 and in 26 (22%) patients in group
2.
Any grade adverse events were detected in 66% of patients in group 1 and in 79% in
group 2 (p=0.068), 3-4 grade AE were detected in 10% and 19% respectively (p=0.151).
Summary/Conclusion: Nivolumab 40 mg therapy is comparable to the standard dose of
3 mg/kg in terms of overall response rate as well as survival in patients with r/r
cHL. However, a direct comparison of different doses of nivolumab in a prospective
study is required.
P1081: PET-ADAPTED THERAPY AFTER THREE CYCLES OF ABVD FOR ALL STAGES OF HODGKIN LYMPHOMA:
LONG TERM FOLLOW UP OF THE GATLA LH-05 TRIAL
A. Pavlovsky1,*, N. L. Fiad1, M. V. Prates1, I. Fernandez1, N. Kurgansky1, A. Cerutti1,
F. Sackmann1, F. Negri Aranguren1, P. Negri Aranguren1, G. Remaggi1, L. Ferrari1,
R. Mariano1, L. Guanchiale1, J. Maradei1, F. Giuliani1, E. Roveri1, A. Enrico1, S.
Zabaljauregui1, M. D. R. Cabrejo1, C. Gumpel1, A. I. Varela1, M. Riddick1, S. Pavlovsky1
1GATLA, Buenos Aires, Argentina
Background: PET-CT adapted treatment for first line Hodgkin Lymphoma has been widely
studied in the last decades. Long-term follow-up is important to judge both effcacy
and safety of this approach.
Aims: Evaluate the efficacy of the PET3 adapted treatment in patients with classical
Hodgkin’s lymphoma (HL) in Ann Arbor stages I-IV.
Methods: We analyzed updated follow-up data on all patients (pts) treated within the
LH-05 GATLA trial. Newly diagnosed pts with HL Stages I-IV were included. All patients
received 3 ABVD and were evaluated with a PET-CT-3 (PET3). Pts with a negative PET3
(DS 1 and 2) were consideredin metabolic CR and received no further therapy. Pts with
DS 3 and 4 completed 6 ABVD and IFRT on PET-CT positive areas. Pts with progressive
disease (DS 5) after 3 ABVD received salvage chemotherapy. We present the updated
results of 490 pts with a median follow-up of 10 years. With a median age of 35 yrs.,
300 presented with localized stage and 190 with advanced stage.
Results:
In LH-05, of all pts, 338 (69%) achieved CR with negative PET3, 152 (31 %) were PET3
positive. With a median follow up of 120 months the PFS and OS for all pts at 5 years
is 79.2%and 94.3% respectively. Pts with negative PET3 had an PFS of 89% and 80% for
localizedand advanced stage, compared to 63% for all pts with positive PET3 (p <0.0001).
We performed a multivariate analysis for PFS which included age, stage, IPS, bulky
disease, extranodalareas and the result of the PET3. This last parameter, together
with age, were the only ones with statistical significance (p=0.001 and 0.046 respectively).
Stage at diagnosis was not significant.
With long term follow up the OS at 5 years is 97.3% and 87.3% for all PET3 negative
vs PET3 positive pts.
When comparing the results LH-05 with our previous clinical trial (LH-96) there is
no difference in PFS and OS at 5 years but in LH-05 only 31% received more than 3
cycles of ABVD and IFRT compared to 61% and 100% in LH-96. This PET adapted approach
reduces exposure to chemo andradiotherapy with no negative effect on long term outcome.
Image:
Summary/Conclusion:
This long term follow up data support the PET-CT adapted approach for all stages ofHL
after a short course of ABVD. In the Cox regression model, PET-CT at completion of
treatment was the most significant factor associated to PFS.
Treatment with 3 cycles of ABVD can be adequate for pts with negative PET3 regardlesstheir
stage at diagnosis. Nevertheless, this long term follow up demonstrated that there
is still a room for improvement trying to identify PET3 negative pts that will relapse
and escalating treatment in PET3 positive pts to improve outcome. GATLA is designing
a trial with the aim to improve these two different risk groups.
P1082: SAFETY AND EFFICACY ANALYSIS IN OLDER PATIENTS TREATED WITHIN THE GATLA LH-05
PROTOCOL: PET-ADAPTED THERAPY AFTER 3 CYCLES OF ABVD FOR ALL STAGES OF HODGKIN LYMPHOMA
N. L. Fiad1,*, M. V. Prates1, I. Fernandez1, N. Kurgansky1, A. Cerutti1, F. Negri
Aranguren1, L. Guanchiale1, F. Sackmann1, J. Maradei1, A. Enrico1, P. Negri Aranguren1,
A. Pavlovsky1
1GATLA, Buenos Aires, Argentina
Background: Treatment for classic Hodgkin lymphoma (cHL) in older age population continues
to be a challenge. Comorbidity prevalence, increase toxicity to the standard treatments
and the lack of inclusion in clinical trials all contribute to this phenomenon. The
obtained results are inferior in comparison to young adults and there is shortage
of evidence regarding effective therapeutic strategies. Recently, the GATLA LH-05
protocol has published the long-term follow-up of the results of the PET/CT adapted
strategy after 3 cycles of ABVD, regardless of the presentation stage and no upper
age limit.
Aims: To assess the effectiveness and safety of the interim PET/CT adapted treatment
in cHL patients older than 60 years old.
Methods: A retrospective analysis of the LH-05 database was performed. Patients were
included ≥ aged 60 with recent diagnosis of cHL stage I-IV and HIV negative. All patients
received 3 cycles of ABVD and were evaluated with PET-TC (PET3). Those patients with
negative PET (Deauville score 1 and 2) were considered to be in complete remission
(CR) and they finalized the treatment. Patients with DS 3 and 4 completed 6 ABVD cycles
and involved-field radiotherapy in hypermetabolic areas in interim PET3. Patients
with DS 5 were considered to have progressive disease. Progression-free survival (PFS)
and overall survival (OS) were evaluated. Kaplan-Meier method and Log-rank test were
used for survival analysis.
Results: Of a total of 490 patients included in the GATLA LH-05 protocol, 59 met the
inclusion criteria. The mean age was of 66 years (range: 60-89), 90% presented a PS<2.
The most frequent histological subtype was Nodular Sclerosis (54%) and 75% presented
in localized stage (I-II). All patients received initial treatment with 3 cycles of
ABVD and were evaluated by PET: 81% presented negative PET (DS 1-2) and 19% were positive.
No GIII-IV toxicity or treatment-related deaths were recorded. With a median follow-up
of 10 years, median PFS and OS were not reached. At 36 and 60 months, PFS was 86%
and 78,9%, and OS was 90% and 85.5%, respectively. Negative PET3 patients had a PFS
of 85.4%, while positive PET3 patients had a PFS of 43% (p=0,0001). In the multivariable
analysis that included age (>60 vs. <60), stage (localized vs. advanced), IPS (<2
vs. >2), extranodal areas, bulky disease and PET3 result, only age and PET3 result
had significant impact in PFS (p=0.046 and p=0,001 respectively). PFS of PET/TC- positive
patients was significantly lower than in the cohort of patients younger than 60. OS
at 36 and 60 months in PET negative patients was 97.5% and 94.5% respectively, versus
63.6% and 53% in PET/TC positive patients.
Image:
Summary/Conclusion: With the PET/TC adapted treatment after 3 cycles of ABVD in 59
patients > 60 years old, 81% of patients achieved negative PET and therefore received
no further treatment. These patients had an excellent result with a PFS of 85.4% at
3 years, similar to the younger patient population. However, a significant reduction
in PFS was observed in PET3 positive patients > 60 years old compared to younger ones.
Implementation of this PET/TC guided strategy, regardless of stage at diagnosis, resulted
in reduced exposure to chemotherapy and radiotherapy, contributing to the absence
of severe treatment-related morbidity and mortality.
P1083: BRENTUXIMAB VEDOTIN, ETOPOSIDE, SOLUMEDROL, HIGH DOSE ARA-C & PLATINUM FOLLOWED
BY HDT & APBSCT FOR REFRACTORY/RELAPSED HODGKIN LYMPHOMA PATIENTS: LONG-TERM RESULTS
OF THE GELTAMO GROUP BRESHAP STUDY
R. Garcia-Sanz1,*, C. Martínez2, F. De la Cruz3, A. P. González4, A. Rodríguez5, B.
Sánchez-González6, E. Domingo-Domenech7, M. Moreno8, J. López9, J. L. Piñana10, M.
Bastos11, M. Canales12, A. Gutiérrez13, M. J. Rodríguez-Salazar14, A. Navarro1, A.
Sureda7
1hematology, Hospital Universitario, Salamanca; 2hematology, Hospital Clinic, Barcelona;
3hematology, Hospital Virgen del Rocío, Sevilla; 4hematology, Hospital Universitario
Central de Asturias, Oviedo; 5hematology, Hospital 12 de Octubre, Madrid; 6hematology,
Hospital del Mar; 7hematology, Hospital Duran i Reynals, Barcelona; 8hematology, Hospital
Germans Trias i Pujol, Badalona; 9hematology, Hospital Ramón y Cajal, Madrid; 10hematology,
Hospital Clínico, Valencia; 11hematology, Hospital Gregorio Marañón; 12hematology,
Hospital La Paz, Madrid; 13hematology, Hospital Son Espases, Palma de Mallorca; 14hematology,
Hospital Universitario de Canarias, Tenerife, Spain
Background: For Refractory/Relapsed Hodgkin Lymphoma (RRHL) patients, the combination
of BRentuximab vedotin, Etoposide, Solumedrol, High dose Ara-C and Platinum (BRESHAP)
as induction therapy prior to high-dose therapy and Autologous Stem Cell Transplantation
(APBSCT) induces an overall (OR) and complete remission (CR) rate of 93% and 71% of
patients with an initial 75% Time to Tumor Progression at three years (Garcia-Sanz
et al, Ann Oncol 2019). In these patients, the primary efficacy endpoint is usually
the CR pre-APBSCT, but long-term results including time to tumor progression (TTP)
or overall survival are challenges that should be evaluated in these recently adopted
strategies.
Aims: In this work, we evaluate long-term results of the phase II trial with the combination
of BV and ESHAP [BRESHAP] as 2nd line therapy for RRHL prior to APBSCT (ClinicalTrials.gov
#NCT02243436).
Methods: Data update with an additonal follow-up period of four years compared with
the initial publication in the EHA-2018 congress
Results: The trial included 66 patients with relapsed or refractory classical HL (cHL)
who had failed to one prior line of therapy were eligible. There were 35 females &
31 males, with a median age of 36 years (18-66). Forty patients were primary refractory,
16 early relapses (CR ≤1 year) and 10 late relapses (CR >1 year). During the follow-up,
excluding progressions, there were 39 Severe Adverse Events (SAEs) reported in 22
patients (hospitalizations and AEs around transplant were not considered SAEs), which
means that no new SAEs were reported after the initial publication of the results.
Transplant was done after BRESHAP in 61 patients with no problems reported about the
engraftment in the long-term evaluation. As reported, pre-transplant OR was 93% (including
a 71% CR).
At a mean follow-up of 75 months, 16 patients have progressed and 3 died without progression,
providing 7-year time to treatment failure (TTP) and progression free survival (PFS)
of 74% and 70%, respectively. Eight patients have died: 5 due to progression, and
the 3 due to complications in the absence of relapse or refractory disease: pneumonia,
sepsis and pulmonary embolism. The overall survival (OS) at seven year was 88% (figure
2). There were three variables related to a lower PFS and TTP: CR after BRESHAP, bone
marrow infiltration at relapse/refractoriness and B symptoms at inclusion in the trial.
In the multivariate analysis, only response to BRESHAP maintained the independent
predictive capacity. Respect to OS, the only prognostic factor was CR to 2nd line
therapy
Image:
Summary/Conclusion: BRESHAP followed by high dose therapy and autologous peripheral
blood stem transplant is a safe and effective strategy for patient with refractory
or relapsed Classical Hodgkin lymphoma with excellent long-term results.
P1084: CHECK-POINT INHIBITORS IN PATIENTS WITH RELAPSE/REFRACTORY HODGKIN’S LYMPHOMA:
A RETROSPECTIVE ANALYSYS BY THE RETE EMATOLOGICA PUGLIESE (REP).
F. Gaudio1,*, G. Loseto2, V. Bozzoli3, P. R. Scalzulli4, A. M. Mazzone5, L. Tonialini6,
V. Fesce7, G. Quintana8, G. De santis9, P. Masciopinto10, E. Arcuti10, F. Clemente2,
S. Scardino3, G. Tarantini9, D. Pastore8, L. Melillo7, V. Pavone6, P. Mazza5, A. M.
Carella4, N. Cascavilla4, N. Di Renzo3, A. Guarini2, P. Musto1,10
1Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico; 2Hematology Unit,
Giovanni Paolo II IRCCS Cancer Institute Oncology Hospital, Bari; 3Hematology and
Stem Cell Transplant Unit, “Vito Fazzi” Hospital, Lecce; 4Hematology Unit, IRCCS “Casa
Sollievo della Sofferenza”, S. Giovanni Rotondo (FG); 5Hematology Unit, Department
of Hematology-Oncology, Moscati Hospital, Taranto; 6Hematology and Transplant Unit,
Cardinal Panico Hospital, Tricase (LE); 7Hematology Unit, Azienda Ospedaliero Universitaria-Ospedali
Riuniti, Foggia; 8Hematology Unit, A. Perrino Hospital, Brindisi; 9Hematology Unit,
Dimiccoli Hospital, Barletta; 10Department of Emergency and Organ Transplantation,
“Aldo Moro” University, Bari, Italy
Background: The majority of patients with either limited stage or advanced-stage classical
Hodgkin lymphoma (cHL) can be cured with chemotherapy; however, 5–10% will have refractory
disease to frontline therapy and approximately 10–30% will relapse. In recent years,
checkpoint/PD-1 inhibitors have significantly changed the prognosis of patients with
relapsing refractory cHL, demonstrating exceptional results in heavily pretreated
patients. There is limited data on the real-world experience with PD-1 inhibitors
in cHL and it is unknown whether fewer selected patients treated with these agents
receive benefits similar to those observed in published trials.
Aims: We performed a retrospective multicentre analysis of the Rete Ematologica Pugliese
(REP) of 66 patients with relapsing refractory cHL who received PD-1 inhibitors in
the non-trial setting.
Methods: Forty-three patients (65%) were treated with nivolumab and 23 (35%) with
pembrolizumab. Median age at diagnosis was 36 years (range, 18–81) and median age
at initiation of PD-1 inhibitor therapy was 43 years (range 19–86).
Forty-four (67%) and 23 (35%) patients underwent autologous stem cell transplantation
(SCT) and allogeneic SCT, respectively, prior to checkpoint inhibitor therapy.
The median lines of treatment attempted prior to PD-1 inhibitor therapy was 4 (range,
3 to 7). All patients were treated with brentuximab vedotin prior to receiving checkpoint
inhibitor therapy.
Results: The best overall response rate to PD-1 inhibitor therapy was 70%: 47% complete
remission (CR) and 23% partial remission (PR). Five patients (8%) achieved stable
disease (SD), while 14 patients (21%) showed progressive disease (PD). In patients
treated with nivolumab the response was: CR in 23 (54%), PR in 9 (21%), SD in 4 (9%),
PD in 7 (16%) patients, respectively; in those treated with pembrolizumab CR occurred
8 (35%), PR in 6 (26%), SD in 1 (4%), PD in 8 (35%) patients, respectively. Twenty-four
immune-related adverse events were documented (4 gastrointestinal, 4 hepatic, 6 fever,
4 hematological, 3 dermatological, 3 allergic rhinitis). Toxicity resolved in all
patients and there were no deaths attributed to checkpoint inhibitor therapy.
After a median follow-up of 26 months (range 3-72 months), 54 patients (82%) are alive
and 12 (18%) died. The cause of death was attributed to disease progression in 9 patients,
and sepsis in 3 patients. After PD-1 inhibitor therapy, 22 patients (33%) relapsed
or progressed. The overall survival and progression free survival at five years were
65% and 54%, respectively.
Summary/Conclusion: We demonstrate similar response rates, clinical outcome and toxicity
profiles compared to clinical trials. These results support the effectiveness and
tolerability of PD-1 inhibitors therapy in relapsing refractory cHL in a real-world
setting.
P1085: EVALUATION OF GONADAL FUNCTION IN YOUNG WOMEN DIAGNOSED WITH HODGKIN AND NON-HODGKIN
LYMPHOMA
A. N. Georgopoulou1,*, A. Giannakou1, T. P. Vassilakopoulos1, M. Siakantaris1, N.
A. Georgopoulos2, S. Kalantaridou3, E. Lalou1, K. Keramaris1, E. Loukari1, M. Arapaki1,
I. Konstantinou1, A. Machairas1, A. Kopsaftopoulou1, I. Mammali2, I. Vassilopoulos1,
D. Galopoulos1, M. K. Angelopoulou1
1Department of Hematology and BMT of the National and Kapodistrian University of Athens,
Laikon, laikon general hospital, Athens; 2Department of Endocrinology of the University
Hospital of Patras, University Hospital of Patras, Patras; 3B Obstetrics and Gynecology
Department of the National and Kapodistrian University of Athens, Areteion Hospital,
Athens, Greece
Background: ABVD chemotherapy (CT) is the standard treatment approach for Hodgkin
lymphoma (HL), with a negative interim PET. R-DA-EPOCH and R-CHOP are the most frequently
used regimens used in primary mediastinal/high grade B-NHL and DLBCL, respectively.
Young women are often affected by these types of lymphoma and may be cured in >80%
of the cases. Thus, treatment-related complications are being increasingly recognized,
among which, gonadal insufficiency with its major psychological consequences. This
is more prevalent in female patients, in whom collection and cryopreservation of oocytes/ovarian
tissue are not applied in everyday clinical practice. Published data are scarce on
this subject. Moreover, little is known about the kinetics of gonadal function and
sex hormones during chemotherapy to safely guide contraceptive measures.
Aims: The aim of this study is the prospective evaluation of gonadal function in young
women with malignant lymphoma who are receiving CT. We here present our preliminary
results on 50 patients with HL and B-NHL.
Methods: This is a prospective study of gonadal function in female patients≤40 years.
Hormonal measurements were performed at pre-specified time points: before treatment
(t0), during CT(t1), at the end of CT(t2) and every six months(t6,t12) thereafter.
The following hormones were measured: follicle-stimulating hormone (FSH), lutenizing
hormone (LH), progesterone (PG), estradiol (Ε2), anti-Mullerian hormone (ΑΜΗ). The
study included:32 HL [median age: 29 years, 32 ABVD] and 18 B-NHL patients [median
age:27 years, 9 RDAEPOCH, 7 RCHOP, 2 other]. FSH reflects gonadal function in women
(increased levels indicate gonadal dysfunction). AMH is considered to be the most
sensitive biomarker for gonadal reserve (decreasing values correlate with ovarian
insufficiency). E2 and progesterone are the major sex hormones.
Results: HL-ABVD: FSH constantly increased from the beginning, peaking in the middle
(fsh0-1 p<0.0001), remaining high until the end (fsh0-2 p<0.0001), without reaching
normal levels at 6 months (fsh0-6 p=0.002), finally normalizing at 12 months after
the end of treatment. Consistently with FSH, AMH sharply decreased during treatment:
[median values: 2,85IU/mL(t0), 0,45IU/mL(t1), 0,62IU/mL(t2), p<0.0001 for both]. A
rebound increase was observed at 6 months and slowly decreased again to normal values
at 12 months after the end of CT. Estradiol and progesterone were not affected throughout
the treatment.
B-NHL: Gonadal damage, reflected by the increase in FSH, was evident, though with
slightly different kinetics compared to HL: [median values: 4,1IU/mL(t0), 8,3IU/mL(t2),
fsh0-2 p=0,035]. FSH gradually increased during treatment, reaching a peak towards
the end. FSH slowly reached normal values after six months from the end of treatment.
In parallel to HL, AMH values sharply decreased during CT: [median values: 7,3IU/mL(t0),
0,17IU/mL(t1), 1,57IU/mL(t2), amh0-1 p=0.002, amh0-2 p=0,043). However, in contrast
to HL, no clear rebound was seen and AMH values remained extremely low, even at 18
months after the end of treatment (median values: 0,23IU/mL), highlighting the gonadotoxic
effect of CT. Estradiol and progesterone did not change significantly.
Image:
Summary/Conclusion: Gonadal function in female patients with malignant lymphomas is
affected during CT in both HL and B-NHL, though with different kinetics. In HL patients
treated with ABVD, gonadal function normalized at 6 months, whereas, in B-NHL gonadal
dysfunction remained even at 18 months after the end of CT, possibly indicating a
chemotherapy-dependent genotoxic effect.
P1086: FAVEZELIMAB (ANTI–LAG-3) AND PEMBROLIZUMAB CO-BLOCKADE IN ANTI–PD-1–NAIVE PATIENTS
WITH RELAPSED OR REFRACTORY CLASSICAL HODGKIN LYMPHOMA: AN OPEN-LABEL PHASE 1/2 STUDY
G. Gregory1,*, J. Timmerman2, D. Lavie3, P. Borchmann4, A. F. Herrera5, L. Minuk6,
V. Vucinic7, P. Armand8, A. Avigdor9, R. Gasiorowski10, Y. Herishanu11, C. Keane12,
J. Kuruvilla13, J. Palcza14, P. Pillai14, P. Marinello14, N. A. Johnson15
1School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia;
2UCLA Medical Center, Los Angeles, United States of America; 3Hadassah Medical Center,
Jerusalem, Israel; 4University Hospital of Cologne, Cologne, Germany; 5City of Hope,
Duarte, United States of America; 6CancerCare Manitoba, Winnipeg, Canada; 7University
of Leipzig Medical Center, Leipzig, Germany; 8Dana-Farber Cancer Institute, Boston,
United States of America; 9Sheba Medical Center, Ramat Gan, and Sackler Faculty of
Medicine, Tel Aviv University, Tel Aviv, Israel; 10Concord Hospital, University of
Sydney, Concord, Australia; 11Tel Aviv Sourasky Medical Center, Tel Aviv-Yafo, Israel;
12Princess Alexandra Hospital, Brisbane, Australia; 13Princess Margaret Cancer Centre,
Toronto, Canada; 14Merck & Co., Inc., Kenilworth, United States of America; 15Jewish
General Hospital, Montreal, Canada
Background: Programmed cell death 1 (PD-1) inhibitors are a standard of care in patients
with relapsed or refractory (R/R) classical Hodgkin lymphoma (cHL), but clinical interest
persists for new approaches to deepen and lengthen responses. Dual blockade of PD-1
and lymphocyte-activation gene 3 (LAG-3) has demonstrated antitumor activity in preclinical
models.
Aims: The multicohort phase 1/2 MK-4280-003 study (NCT03598608) evaluated the safety
and efficacy of favezelimab (MK-4280), a humanized Immunoglobulin G4 LAG-3 inhibitor,
plus pembrolizumab (a PD-1 inhibitor) in patients with R/R hematologic malignancies.
This analysis focused on anti–PD-1–naïve patients with R/R cHL (cohort 1).
Methods: This study included a safety lead-in phase (part 1) to determine the recommended
phase 2 dose (RP2D) followed by a dose-expansion phase (part 2). Eligible patients
in cohort 1 must have had R/R cHL after autologous stem cell transplantation (ASCT)
or been ineligible for ASCT and have had no prior anti–PD-1 therapy. In part 1, patients
from all cohorts received intravenous (IV) pembrolizumab 200 mg every 3 weeks (Q3W)
and favezelimab IV 200 mg or 800 mg Q3W. Dose-finding based on occurrence of dose-limiting
toxicities (DLTs) was determined using a modified toxicity probability interval design.
In part 2, patients received pembrolizumab + favezelimab at the established RP2D for
up to 2 years (35 cycles), or until documented disease progression, adverse events
(AEs), or withdrawal from the study. Primary end point was safety, including DLTs
and AEs. Secondary end point was objective response rate (ORR). Duration of response
(DOR), progression-free survival (PFS), and overall survival (OS) were exploratory
end points.
Results: Only 1 DLT (autoimmune hepatitis [grade 4]) was identified among the first
6 patients from all cohorts included in part 1 at the favezelimab 200 mg dose; thus,
the dose was escalated to 800 mg. No DLTs were observed in the 15 additional patients
treated at the 800 mg dose. The RP2D for the combination was defined as 800 mg Q3W
+ pembrolizumab 200 mg Q3W. In cohort 1, 30 patients were enrolled; median age was
40 years, 53% had ECOG PS 0, and 80% had no more than 3 prior lines of therapy. As
of the database cutoff (Nov 1, 2021), 9 of 30 (30%) patients had discontinued treatment
either due to adverse events (n=3) or disease progression (n=6). After a median follow-up
of 13.5 months, ORR for cohort 1 was 73% (95% CI, 54-88; complete response, 7 patients
[23%]; partial response, 15 patients [50%]). 28 of 30 patients (93%) had a reduction
from baseline in target lesions. Median DOR was not reached ([NR]; 95% CI, 0+ to 23+
months) and 6 patients (51%) had response ≥12 months. The median PFS was 19 months
(95% CI, 8-NR) and the 12-month PFS rate was 57%. The median OS was NR (95% CI, NR-NR)
and the 12-month OS rate was 94%. Treatment-related AEs (TRAE) occurred in 26 patients
(87%); most common (≥10%) were hypothyroidism (27%), fatigue (20%), infusion-related
reactions (20%), headache (17%), and arthralgia, hyperthyroidism, myalgia, and nausea
(10% each). Grade 3 or 4 TRAEs occurred in 6 patients (20%) and 10% of patients discontinued
due to TRAEs. No treatment-related deaths occurred.
Summary/Conclusion: Favezelimab 800 mg + pembrolizumab 200 mg Q3W demonstrated an
acceptable safety profile and effective antitumor activity in anti–PD-1–naïve patients
with R/R cHL. Further studies are merited to compare the activity of this combination
to that of pembrolizumab alone.
P1087: FAVEZELIMAB (ANTI–LAG-3) AND PEMBROLIZUMAB CO-BLOCKADE IN PATIENTS WITH RELAPSED
OR REFRACTORY CLASSICAL HODGKIN LYMPHOMA WHO PROGRESSED AFTER ANTI–PD-1 THERAPY: AN
OPEN-LABEL PHASE 1/2 STUDY
A. F. Herrera1,*, D. Lavie2, N. A. Johnson3, A. Avigdor4, P. Borchmann5, C. Andreadis6,
A. Bazargan7, G. Gregory8, C. Keane9, T. Inna10, V. Vucinic11, P. L. Zinzani12, H.
Zhang13, P. Pillai13, P. Marinello13, J. Timmerman14
1City of Hope, Duarte, United States of America; 2Hadassah Medical Center, Jerusalem,
Israel; 3Jewish General Hospital, Montreal, Canada; 4Sheba Medical Center, Ramat Gan,
and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; 5University
Hospital of Cologne, Cologne, Germany; 6UCSF Helen Diller Family Comprehensive Cancer
Center, San Francisco, United States of America; 7University of Melbourne, Melbourne,
and St Vincent’s Hospital, Fitzroy; 8School of Clinical Sciences at Monash Health,
Monash University, Melbourne; 9Princess Alexandra Hospital, Brisbane, Australia; 10Rambam
Health Care Campus, Haifa, Israel; 11University of Leipzig Medical Center, Leipzig,
Germany; 12IRCCS Azienda Ospedaliero-Universitaria di Bologna Istituto di Ematologia
“Seràgnoli” and Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale,
Università di Bologna, Bologna, Italy; 13Merck & Co., Inc., Kenilworth; 14UCLA Medical
Center, Los Angeles, United States of America
Background: PD-1 inhibitors are a standard of care for relapsed or refractory (R/R)
classical Hodgkin lymphoma (cHL), but optimal therapy is yet to be defined for patients
whose disease progresses after anti–PD-1 treatment. Dual blockade of PD-1 and lymphocyte-activation
gene 3 (LAG-3) has demonstrated antitumor activity in preclinical models.
Aims: This multicohort phase 1/2 study (NCT03598608) evaluated safety and efficacy
of favezelimab (MK-4280), a humanized IgG4 LAG-3 inhibitor, plus the PD-1 inhibitor
pembrolizumab in patients with R/R hematologic malignancies. Cohort 2 focused on patients
with R/R cHL refractory to anti–PD-1 therapy.
Methods: This study included a safety lead-in phase (part 1) to determine recommended
phase 2 dose (RP2D) followed by a dose-expansion phase (part 2). Eligible patients
in cohort 2 had R/R cHL, relapsed after or were ineligible for autologous stem cell
transplantation (ASCT), and progressed after ≥2 doses of anti–PD-1 therapy (within
12 weeks of last dose). In part 1, patients from all cohorts received intravenous
(IV) pembrolizumab 200 mg every 3 weeks (Q3W) and favezelimab IV 200 mg or 800 mg
Q3W. Dose-finding based on occurrence of dose-limiting toxicities (DLT) was determined
using a modified toxicity probability interval design. In part 2, patients received
pembrolizumab + favezelimab at the established RP2D for up to 2 years (35 cycles),
or until documented disease progression, adverse events (AEs), or withdrawal from
study. Primary end point was safety, including DLTs and AEs. Secondary end point was
objective response rate (ORR). Duration of response (DOR), progression-free survival
(PFS), and overall survival (OS) were exploratory end points.
Results: Only 1 DLT (autoimmune hepatitis [grade 4]) was observed among the first
6 patients from all cohorts included in part 1 at the favezelimab 200 mg dose; thus,
the dose was escalated to 800 mg. No DLTs were observed in the 15 additional patients
at the 800 mg dose. Favezelimab RP2D was defined as 800 mg Q3W + pembrolizumab 200 mg
Q3W. In cohort 2, 33 patients were enrolled; median age was 37 years, 64% had ECOG
PS 0, and 94% had at least 4 prior lines of therapy. At database cutoff (Nov 1, 2021),
20 patients had discontinued treatment due to adverse events (n=7); disease progression
or clinical progression (n=11); or withdrawal/physician decision (n=2). After a median
follow-up of 16.5 months, ORR for patients receiving favezelimab 800 mg (n=29) was
31% (95% CI, 15-51; complete response, 2 [7%]; partial response, 7 [24%]). 19 of 29
responders (66%) had an anti–PD-1–based regimen as their most recent line of therapy
at study entry. 23 of 29 patients (79%) had a reduction from baseline in target lesions.
Median DOR for patients who received favezelimab 800 mg was not reached ([NR]; 95%
CI, 0+ to 14+ months). For all patients in cohort 2, the median PFS was 9 months (95%
CI, 5-15); 12-month PFS rate was 39%. Median OS was 26 months (95% CI, 26-NR); 12-month
OS rate was 91%. Treatment-related AEs (TRAE) occurred in 28 patients (85%); most
common (>10%) were hypothyroidism (18%), nausea and fatigue (15% each), and arthralgia
and diarrhea (12% each). Grade 3 or 4 TRAEs occurred in 6 patients (18%) and 18% of
patients discontinued treatment due to TRAEs. No treatment-related deaths occurred.
Summary/Conclusion: Favezelimab 800 mg + pembrolizumab 200 mg Q3W showed a tolerable
safety profile and effective antitumor activity in heavily pretreated patients with
R/R cHL whose disease had progressed after anti–PD-1 therapy, suggesting that the
combination may reinduce a response in these patients.
P1088: UPDATED DATA FROM A PHASE 1/2 STUDY OF BRENTUXIMAB VEDOTIN COMBINED WITH CHEMOTHERAPY
IN PEDIATRIC PATIENTS WITH ADVANCED STAGE CLASSICAL HODGKIN LYMPHOMA
F. Locatelli1,*, F. Luisi2, M. Pianovski3, M. A. Salvino4, F. Fagioli5, S. Epelman6,
L. Britto De Abreu Lima7, R. Norris8, V. Odone Filho9, M. Zecca10, C. Favre11, R.
Kobayashi12, Y. Koga13, Y. Sidi14, X. Zhou14, X. Bai14, F. Campana14, E. J. Leonard14,
A. R. Franklin15
1Department of Pediatric Haematology and Oncology and Cell and Gene Therapy, IRCCS
Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome, Italy; 2Pediatria,
GRAACC/UNIFESP, São Paulo; 3Department of Pediatric Oncology, Hospital Erastinho,
Liga Paranaense de Combate ao Câncer, Jardim das Américas, Curitiba, Parana; 4Hematology,
IDOR, Salvador, Hospital São Rafael, Salvador, Brazil; 5Department of Sciences of
Public Health and Pediatrics, Regina Margherita Children’s Hospital, Turin, Italy;
6Pediatric Oncology Department, Hospital Santa Marcelina, São Paulo; 7Hematology,
INCA - Instituto Nacional de Câncer, Rio de Janeiro, Brazil; 8Department of Pediatrics,
Cincinnati Children’s Hospital Medical Center, Cancer and Blood Diseases Institute,
Cincinnati, OH, United States of America; 9ITACI – Instituto De Tratamento Do Câncer
Infantil – Departament De Pediatria – Faculdade De Medicina Da Universidade De São
Paulo, São Paulo, Brazil; 10Oncoematologia Pediatrica, Fondazione IRCCS Policlinico
San Matteo, Pavia; 11Dipartimento di Oncoematologia, Azienda Ospedaliero Universitaria
Ospedale Pediatrico Meyer, Firenze, Italy; 12Department of Hematology/Oncology for
Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo; 13Department of Pediatrics,
Kyushu University Hospital, Fukuoka-shi, Fukuoka-Ken, Japan; 14Takeda Development
Center Americas, Inc. (TDCA), Lexington, MA; 15Center for Cancer and Blood Disorders,
Children’s Hospital Colorado, Aurora, CO, United States of America
Background: Brentuximab vedotin is a CD30-directed monoclonal antibody-drug conjugate
approved for use in the first-line adult classical Hodgkin lymphoma (cHL) setting
in combination with doxorubicin, vinblastine, and dacarbazine (A+AVD). The safety,
tolerability, recommended dosing, immunogenicity, and antitumor activity of A+AVD
have not previously been evaluated as a first-line therapy in treatment-naïve pediatric
patients with cHL.
Aims: To provide updated safety and efficacy data from an open-label phase 1/2 study
of A+AVD in previously untreated pediatric patients with stage III or IV cHL (NCT02979522).
Methods: Patients with newly diagnosed stage III or IV cHL who were aged 5 to <18
years were treated with A+AVD on days 1 and 15 of 28-day cycles, for a maximum of
6 cycles. In phase 1, patients (n = 8) received brentuximab vedotin 48 mg/m2 + AVD;
dose-limiting toxicity (DLT) was assessed from day 1 of cycle 1 to day 56. In phase
2, 51 additional patients received the same treatment regimen. Progression-free survival
(PFS) was defined as the time between first dose and time of disease progression,
and event-free survival (EFS) was defined as the time between first dose and time
of treatment failure; EFS events were disease progression, treatment withdrawal, and
death. All patients had been on study for at least 2 years at the data cutoff (September
24, 2021). The data presented refer to phases 1 and 2 combined.
Results: All patients (n = 59) in phases 1 and 2 completed 6 cycles of A+AVD. Among
6 DLT-evaluable patients in phase 1, no DLTs were observed. The maximum tolerated
dose of brentuximab vedotin was not reached and the recommended dose was determined
to be 48 mg/m2. Treatment-emergent adverse events (TEAEs) occurred in all patients;
the most common TEAEs were vomiting (85%), nausea (75%), and neutropenia (58%). Fourteen
patients (24%) experienced treatment-emergent peripheral neuropathy (PN); of these,
10 patients had PN resolved by end of treatment (EOT), 13 patients had PN resolved
by last contact, and one patient had ongoing grade 1 paraesthesia at last contact.
No on-study deaths occurred. At the end of cycle 2, the PET-negative rate (Deauville
score 1, 2, or 3) was 90%. Objective response rate (complete response [CR] + partial
response) according to independent review facility was 88%, with 76% of patients achieving
a CR. Median duration of response and duration of CR were not estimable. Median PFS,
EFS, and overall survival were not reached, suggesting promising efficacy of A+AVD
in pediatric patients. Overall PFS, EFS and overall survival data will be presented
at the meeting.
Summary/Conclusion: Brentuximab vedotin 48 mg/m2 plus AVD every two weeks was generally
well tolerated with an acceptable safety profile, and demonstrated an efficacy benefit
in CD30+ pediatric patients with previously untreated advanced cHL. These data support
A+AVD as a suitable frontline therapy option in this patient population.
P1089: BRENTUXIMAB VEDOTIN, NIVOLUMAB, DOXORUBICIN, AND DACARBAZINE (AN+AD) FOR ADVANCED
STAGE CLASSIC HODGKIN LYMPHOMA: PRELIMINARY SAFETY AND EFFICACY RESULTS FROM THE PHASE
2 STUDY (SGN35 027 PART B)
H. Lee1, I. W. Flinn2, J. Melear3, R. Ramchandren4, J. Friedman5, J. M. Burke3, Y.
Linhares6, M. Raval3, R. Chintapatla7, T. A. Feldman8, H. Yimer3, M. Islas-Ohlmayer3,
A. Dean3, V. Rana9, M. D. Gandhi3, J. Renshaw3, A. L. Gillespie-Twardy3, L. Ho10,*,
M. Puhlmann10, W. Guo10, C. A. Yasenchak11
1MD Anderson, Houston; 2Sarah Cannon Research Institute and Tennessee Oncology, Nashville;
3US Oncology Research, The Woodlands; 4University of Tennessee Medical Center, Nashville;
5University Hospitals Seidman Cancer Center, Cleveland; 6US Oncology Research, Woodlands;
7Kadlec Clinic, Kennewick; 8Lymphoma Division, John Theurer Cancer Center at Hackensack
Meridian Health, Hackensack; 9University of Colorado Health Hematology and Oncology,
Colorado Springs; 10Seagen, Bothell; 11Willamette Valley Cancer Institute and Research
Center, Eugene, United States of America
Background: Brentuximab vedotin (BV) and nivolumab are both active and well-tolerated
in patients (pts) with classical Hodgkin lymphoma (cHL) and were previously studied
in first salvage (overall response rate [ORR] 85%; complete response [CR] 67%) (Advani
2021) and as firstline (1L) therapy in older adults (ORR 95%; CR 79%) (Yasenchak 2019).
The combination of BV plus nivolumab demonstrated promising activity as a frontline
treatment option for pts over 60 yrs of age with cHL (Friedberg 2018). Additionally,
in pts with non-bulky Stage I or II cHL, treatment with BV plus doxorubicin and dacarbazine
(AD) resulted in a CR rate of 97% at end of treatment (EOT), as well as a promising
4-year progression-free survival (PFS) estimate of 91%. There were no cases of ≥Grade
3 peripheral neuropathy and only 9% were Grade 2 (Abramson 2021).
Aims: This abstract will present the preliminary safety and efficacy results from
Part B of the SGN35-027 study, which enrolled pts with Ann Arbor Stage II cHL with
bulky mediastinal disease (defined as a single node or nodal mass ≥10 cm as determined
by CT imaging), or Stage III or IV cHL.
Methods: SGN35-027 (NCT03646123) is an open-label, multiple part, multicenter, phase
2 clinical trial. Pts received up to 6 cycles of AN+AD (consisting of BV 1.2 mg/kg,
nivolumab 240 mg, doxorubicin 25 mg/m2, and dacarbazine 375 mg/m2). All study drugs
were administered by IV infusion on Days 1 and 15 of each 28-day cycle. The primary
endpoint was CR rate at EOT. Key secondary endpoints included safety, tolerability,
ORR, and PFS. Disease response and progression was assessed by investigators using
the Lugano Classification Revised Staging System for nodal non-Hodgkin and Hodgkin
Lymphomas (Cheson 2014) and Lymphoma Response to Immunomodulatory Therapy Criteria
(LYRIC) (Cheson 2016) at Cycle 2 and EOT.
Results: In Part B, 58 pts were enrolled and 57 pts received at least 1 dose of study
treatment. Of these 57 pts, 30 (53%) were male, 27 (47%) were female, 50 (88%) were
white, 46 (81%) were not of Hispanic or Latino/a or Spanish origins, and 54 (95%)
were <65 years old. Median age was 35 yrs (range: 19-78 yrs); 30% had Stage II cHL
with bulky mediastinal disease, while 18% and 51% had Stage III and IV cHL, respectively.
Of 58 pts enrolled, 52 (90%) completed treatment and 4 (7%) discontinued due to AEs.
The most frequently reported TEAEs were nausea, fatigue, and diarrhea (70%, 51%, and
46% of pts, respectively). Grade 3 or higher TEAEs were reported in 30 pts (53%),
with neutropenia and increased ALT in 5 pts (9%) being the most frequent. Treatment-related
AEs occurred in 98% of pts; the most frequent were nausea, fatigue, and peripheral
sensory neuropathy (65%, 46%, and 39% of pts, respectively). Treatment-related SAEs
occurred in 8 pts (14%), of which pneumonitis was the most common (3 pts [5%]). Immune-mediated
AEs were observed in 18 pts (32%), and hypothyroidism (4 pts [7%]) was the most frequently
reported. No febrile neutropenia was observed, and there were no Grade 5 AEs.
Preliminary results show at EOT, 52 pts had a response to treatment (ORR 93% [95%
CI:82.7-98.0]), with 49 pts having a complete response (CR rate: 88% [95% CI: 75.9-94.8]).
Summary/Conclusion: Preliminary results demonstrate that AN+AD has promising clinical
activity and is well-tolerated, with no new safety signals observed. The omission
of bleomycin and vinblastine may have contributed to the absence of certain AEs, such
as febrile neutropenia. AN+AD may provide another active treatment option for patients
with 1L advanced cHL.
P1090: CAMRELIZUMAB (CAM) COMBINED WITH GEMOX CHEMOTHERAPY RESULTS IN HIGH COMPLETE
METABOLIC RESPONSE RATES IN RELAPSED/REFRACTORY CLASSIC HODGKIN LYMPHOMA (CHL): A
PHASE II TRIAL
Y. Xie1,*, Y. Tang1, W. Zheng1, L. Ping1, N. Lin1, M. Tu1, C. Zhang1, Z. Ying1, W.
Liu1, L. Deng1, M. Wu1, L. Mi1, T. Du1, X. Wang1, J. Zhu1, Y. Song1
1Key laboratory of Carcinogenesis and Translational Research (Ministry of Education),
Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing, China
Background: Approximately 30-35% of patients with classic Hodgkin Lymphoma will prove
refractory to frontline therapy or relapse subsequently. Traditional second-line chemotherapy
regimens result in complete response rates about 20-40%. Achievement of complete metabolic
response (CMR) assessed by PET/CT imaging prior to hematopoietic stem cell transplant
(HSCT) predicts favorable progression free survival (PFS) and overall survival (OS).
PD‐1 antibody has shown remarkable efficacy in relapsed or refractory hodgkin lymphoma
(R/R HL).
Aims: we designed a clinical trial to evaluate the efficacy and safety of PD‐1 antibody
camrelizumab combined with GEMOX chemotherapy in R/R HL patients. The preliminary
results had been obtained.
Methods: This is an open‐label, single‐center, non‐randomized, phase 2 study (NCT04239170).
The main inclusion criteria included age ≥ 18, ECOG < 2, histopathology confirmed
classical HL, no more than 3 lines of previous chemotherapy treatment, having measurable
lesion(s) assessed by Lugano 2014 criteria and preparing to receive autologous stem
cell transplantation (ASCT). Patients were treated with 2 cycles of camrelizumab (CAM,
200 mg IV, q2w) followed by 2 cycles (28-day cycle) of CAM (IV, day 1 and day 15)
combined with standard GEMOX (gemcitabine 1000 mg/m2 and oxaliplatin 100mg/m2, IV,
day 1 and day 15). Patients achieved CR were transferred to stem cell mobilization/collection
stage; patients achieved PR/SD could have another cycle of CAM plus GEMOX, patients
who achieved PR/CR underwent stem cell mobilization/collection. For patients waiting
more than 4 weeks for ASCT, 1-2 cycles of CAM monotherapy were allowed. The primary
endpoint is the proportion of patients who achieved PET‐CT‐confirmed complete response
(CR) according to Lugano 2014 criteria.
Results: From March 2020 to December 2021, 30 patients were enrolled. One patient
withdrew informed consent. The median age of remaining 29 patients was 34 years old
(ranged from 22 to 63), with a male‐to‐female ratio of 16:13. At the first tumor response
evaluation, 69% (20/29) patients achieved CR and 17% (5/29) patients achieved partial
response (PR), resulting an overall response rate (ORR) of 86%. Only one patient had
progression disease (PD). The patients who achieved PR or stable disease (SD) received
an additional cycle of CAM plus GEMOX treatment, and one patient who had PR in the
first evaluation was found to achieve CR. Up to now, collection of the autologous
hematopoietic stem cells was completed in thirteen patients. With a cutoff date of
February 10, 2022, the 1-year (progression-free survival) PFS rate was 74% and the
1-year (overall survival) OS rate was 100%, median PFS and OS were not reached with
too few events (Figure). Most adverse events (AEs) were of grade 1 to 2. Common AEs
included reactive capillary endothelial proliferation (RCEP), alanine aminotransferase
(ALT)/ aspartate aminotransferase (AST) elevation, vomiting, nausea, hyperuricemia
(all > 30%). Hematologic AEs included neutropenia (> 34%). No grade 4 or above AEs
occurred. Grade 3 AE occurred in 6 patients, including 1 pulmonary infection, 1 nausea,
1 fever, 1 hypertriglyceridemia, 1 ALT elevation, and 1 neutropenia. Serious adverse
events included 1 case of interstitial pneumonia (grade 2) and 1 case of fever (grade
3) resulting in hospitalization.
Image:
Summary/Conclusion: Camrelizumab combined with GEMOX chemotherapy showed encouraging
clinical efficacy and tolerable toxicity in R/R HL patients and merited further study.
P1091: FEASIBILITY OF ASCT AFTER ANTI-PD-1 THERAPY FOR R/R CLASSICAL HODGKIN LYMPHOMA
N. Mochkin1,*, V. Sarzhevskiy1, Y. Protopopova2, E. Demina1, V. Melnichenko1, V. Bogatyrev1,
A. Samoylova1, A. Mamedova1, A. Rukavitsyn1, A. Bannikova1, E. Smirnova1, N. Shorokhov1
1FSBI National Medical Surgical Center n.a. N.I. Pirogov; 2I.M. Sechenov First Moscow
State Medical University (Sechenov University), Moscow, Russia
Background: Immunotherapy with checkpoint inhibitors (anti-PD-1) is highly effective
in relapsed/refractory (R/R) classical Hodgkin Lymphoma (cHL) and leads to more than
70% overall response rate. However, long-term progression-free survival rate is not
sufficient. Consolidation with ASCT after anti-PD-1 could be a very promising option
even among previously chemorefractory patients. Nevertheless, there is not enough
experience about the safety and feasibility of ASCT after anti-PD-1 in the context
of possible immune-related adverse events (AE).
Aims: to assess the safety and feasibility of ASCT after anti-PD-1 therapy in patients
with R/R cHL.
Methods: We retrospectively analysed patients with cHL who previously underwent ASCT
from November 2018 until December 2021 after treatment with anti-PD-1. A median patient
age was 34 years (range, 20-55), 22 patients were female, 21 - male. Patients received
anti-PD-1 as monotherapy (n=17), combination of anti-PD-1 and chemotherapy (n=4),
anti-PD-1 monotherapy followed by combination of anti-PD-1 and chemotherapy (n=14),
anti-PD-1 monotherapy followed by salvage therapy (n=3) and other (n=2). A median
number of anti-PD-1 cycles was 6 (range, 1-45). The median time from the last dose
of anti-PD1 was 69 days (range, 14-365). Conditioning regimen for ASCT consisted of
BeEAM (n=12), BeAC (n=30) and other (n=1). Safety assessment was performed by Common
Terminology Criteria for Adverse Events (CTCAE) (v5.0).
Results: Forty-three eligible patients were enrolled in this analysis. A median time
to engraftment was 10.5 days (range, 9-26). A median time to platelet count > 20x109/L
was 13.5 days (range, 7-43). A median use of G-CSF after high-dose chemotherapy followed
by ASCT was 9 days (range, 0-26). PEG-G-CSF (empegfilgrastim 7.5 mg/ml once) was used
in 10 patients (23.3%). Plerixafor was used in 6 patients (14%). Neutropenic fever
was observed in 32 patients (74.4%). Documented infections were reported in 19 patients
(44.2%) among which were 5 cases of clostridial colitis (11.6%). A median intravenous
administration of antibiotics was 8 days (range, 0-23). Grade 3-4 mucositis and enteropathy
was reported in 2 patients (4.6%) and 6 (14%), respectively. Other non-hematological
toxicity was observed in 10 patients (23.4%): 8 patients (18.6%) developed engraftment
syndrome (that was successfully treated with steroids), grade 4 autoimmune toxic myocarditis
was reported in 1 patient (2.3%). One death due to grade 5 autoimmune myocarditis
in combination with grade 5 autoimmune pneumonitis was reported (2.3%). A median duration
of hospitalization was 25 days (range, 14-72). Transplant-related mortality (TRM)
was reported in 2 patients (4.6%).
Image:
Summary/Conclusion: The obtained data on safety of ASCT after anti-PD-1 treatment
generally correspond to the data of international studies. Toxicity profile tends
to be very similar to the published data about ASCT without anti-PD-1 previous treatment,
especially in terms of TRM. However, we observe a high incidence of engraftment syndrome
that can lead to fulminant immune-related AE (myocarditis and pneumonitis) probably.
Attention should be paid to the defining of optimal “wash-out” period after last dose
of anti-PD-1 and early defining of possible life-threatening immune related AE (especially
myocarditis and pneumonitis).
P1092: NODULAR LYMPHOCYTE PREDOMINANT HODGKIN LYMPHOMA (NLPHL): HISTOLOGYCAL AND CLINICAL
REVIEW IN A SINGLE CENTRE.
E. Rámila1,*, N. Papaleo2, A. Arrieta2, C. Blazquez2, M. Solorzano2, M. Vidal2, X.
Vilaseca1, J. Piedra1, J. I. Martinez1
1Department of Hematology; 2Department of Pathology, Hospital Universitari Parc Taulí
Sabadell (Barcelona), Sabadell, Spain
Background: NLPHL constitutes a subtype of Hodgkin lymphoma with distinct clinical
characteristics and evolution.
Aims: To review the histopatologycal diagnosis and clinical data in patients with
NLPHL in our institution between 1999-2021.
Methods: We retrospectively reviewed the histology of lymph node biopsy and clinical
data, treatment and outcome of 21 adult patients consecutively diagnosed of a NLPHL
between 1999 and 2021.
Results: Twenty-one adult patients with diagnosis of NLPHL (3 of which presented transformation
areas to diffuse large B-cell lymphoma (t-DLBCL) in the biopsy), were identified.
After the histological review of the biopsies 1 patient was reclassified as diffuse
large B cell lymphoma (DLBCL) and another patient as NLPLH with t-DLBCL. Six (30%)
patients showed a typical histological pattern (A-B) in the biopsy whereas 14 (70%)
presented histopathological variants (pattern C-F) including the 4 patients with transformation
areas to DLBCL. Thus, the clinical data of a total number of 20 patients with NLPLH
(4 of them with transformation areas) were reviewed.
Twelve (60%) patients were males and 8 (40%) female with a median age of 46 years
(range 17-79). Regarding the clinical stage at diagnosis, 9 (45%), 2 (10%), 5 (25%)
and 4 (20%) were in favourable IA stage, favourable IIA stage, unfavourable I-IIA
stage (according the German Study Hodgkin Group and European Organisation for Research
and Treatment of Cancer criteria) and advanced III-IV stage respectively. Three of
the 4 patients in advance stage presented variant histological patterns.
Among the 9 patients in favourable clinical IA stage the treatment administered was
the following: 2 of them didn’t receive any further treatment (in one by rejection
and other patient was in surveillance after lymph node resection), 5 received Radiotherapy
and 2 received combined treatment (chemotherapy with ABVD (doxorubicin, bleomycin,
vinblastine, dacarbazine) and radiotherapy). The 2 patients in favourable IIA stage
received combined treatment. Among the 5 patients in unfavourable I-II clinical stage,
3 received combined treatment (ABVD and radiotherapy), 1 patient was treated with
ABVD and 1 with R-CHOP (rituximab, ciclophosphamide, doxorubicin, vincristine and
prednisone) due to areas of t-DLBCL in the biopsy. Patients (4) in advanced stage
were treated with chemotherapy: 2 with ABVD, 1 with CVP (ciclophosphamide, vincristine
and prednisone) and 1 with rituximab and platinum based regimen (transformation areas
to DLBCL in the biopsy and previous episode of DLBCL).
Regarding to responses, 15 (83.3%) of the 18 treated patients achieved a complete
remission and 3 (16.7%) were refractory (2 of them received second line therapy incuding
rituximab with a complete remission). One patient relapsed; he was refractory to ABVD
and achieved a complete remission with R-CHOP.
With a median follow-up of 5.9 years (range 0.8-18) the overall survival is 90%; 2
patients have died (1 of them, a 79 years old patient with comorbidities, due to lymphoma).
Summary/Conclusion: We have seen a 95% of concordance with the initial diagnosis after
a histological review in our patients with NLPLH. 20% of the biopsies showed transformation
areas to DLBCL. Most of the patients (80%) presented in a localized stage. Refractory
patients responded well to antiCD20 treatment. The prognosis is good with an overall
survival of 90%.
P1093: A NEW PROGNOSTIC MODEL FOR INDIVIDUAL OUTCOME PREDICTION IN ADVANCED-STAGE
HODGKIN LYMPHOMA.
R. Rask Kragh Jørgensen1,2,*, S. Eloranta3, M. Tang Severinsen1,2, A. Kiesbye Øvlisen1,
K. Bjøro Smeland4, C. Kiserud4, J. Haaber Christensen5, M. Hutchings6, R. Bo-Dahl
Sørensen7, P. Kamper8, I. Glimelius3,9, K. E. Smedby3,10, F. Bergström3,11, T. C.
El-Galaly1,2, L. Hjort Jakobsen1,2,12
1Department of Hematology, Clinical Cancer Research Centre, Aalborg University Hospital;
2Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; 3Department
of Medicine Solna, Division of Clinical Epidemiology, Karolinska Institutet, Stockholm,
Sweden; 4Department of Oncology, Oslo University Hospital, Oslo, Norway; 5Department
of Hematology, Odense Universitets Hospital, Odense; 6Department of Hematology, Copenhagen
University Hospital, Copenhagen; 7Department of Hematology, Zealand University Hospital,
Roskilde; 8Department of Hematology, Aarhus University Hospital, Aarhus, Denmark;
9Department of Immunology, Genetics and Pathology, Unit of Oncology, Uppsala University,
Uppsala; 10Department of Hematology, Karolinska University Hospital; 11Department
of Mathematics, Stockholm University, Stockholm, Sweden; 12Department of Mathematical
Sciences, Aalborg University, Aalborg, Denmark
Background: Patients diagnosed with advanced-stage Hodgkin lymphoma (aHL) are commonly
risk stratified based on prognostic scores like the International Prognostic Score
(IPS). These scores are often based on dichotomized clinical predictors, leading to
loss of information and thus inaccurate prognoses.
Aims: This study investigated if a machine learning approach utilizing real world
data from aHL patients can outcompete the IPS in predicting overall survival (OS).
Methods: This study included patients with classical aHL registered in the Danish
Lymphoma Registry (LYFO). We developed a new prognostic model for predicting OS using
stacking, which combined several predictive survival models into a single model. The
models used in the stacking, were Cox porportional hazard (CPH), Flexible parametric
survival (FPS), Random Survival forest and IPS models, with different combinations
of covariates included depending on the model. The time-varying area under the curve
(AUC), integrated Brier score (IBS), and concordance index (C-Index) were used to
measure performance of the new model as well as the IPS and a simplified version containing
three risk-factors (IPS-3). The performance measures were computed using 10-fold cross-validation.
Results: A total of 1,110 aHL patients were included and the 5-year OS was 81% (95%
CI, 79-84%). The IBS for the stacking, IPS, and IPS-3 model was 0.078, 0.099, and
0.096, respectively, while the C-Index was 0.86, 0.73, and 0.71. The time-varying
AUC was consistently higher for the stacking-model compared to IPS-models within the
first five years after diagnosis (Figure).
According to the IPS-model, 143 (12.9%) patients were high-risk (IPS 5-7). The 5-year
OS for these patients was 62% (95% CI, 54-71%). We defined an alternative high-risk
group by including the 143 patients with the lowest predicted 5-year OS based on the
stacking-model. This high-risk group had a 5-year OS of 40% (95% CI, 32-50%).
Image:
Summary/Conclusion: The new prognostic model for aHL based on machine-learning technique
(stacked model) demonstrated a substantial improvement in predictive performance compared
to the IPS-models. The model also detected a high-risk group with worse OS than the
IPS high-risk group. The model is planned for international validation.
P1094: CHARACTERISTICS, MANAGEMENT, AND OUTCOMES OF PATIENTS WITH T-CELL LARGE GRANULAR
LYMPHOCYTIC LEUKEMIA AT A LARGE TERTIARY CARE CENTER
H. Audil1,*, M. Shah1, K. Rabe1, W. Ding1, P. Hampel1, N. N. Bennani1, T. Call1, C.
Hanson1, Y. Wang1, A. Koehler1, S. Schwager1, J. Leis2, S. Slager1, S. Kenderian1,
A. Al-Kali1, G. Nowakowski1, M. Shi1, S. Parikh1
1Mayo Clinic, Rochester; 2Mayo Clinic, Phoenix, United States of America
Background: Large granular lymphocytic leukemia (LGLL) comprises 2-6% of chronic lymphoproliferative
disorders. T-LGLL accounts for approximately 85% of cases, while NK-LGLL represents
15% of cases. There is a significant knowledge gap regarding diagnosis, management,
and outcomes of LGLL.
Aims: Our aims were to describe the clinical characteristics, time to first therapy
(TTFT), type of first line therapy, response to treatment, and overall survival (OS)
in patients with LGLL seen at our institution.
Methods: We identified LGLL patients from the Mayo Clinic Chronic Lymphoproliferative
Disorders Database seen between 1/1995-7/2021. At least 3 of the following 4 criteria
were used to diagnose LGLL (T-LGLL or NK-LGLL): (1) a distinct T-cell or NK-cell population
by flow cytometry; (2) a clonal T-cell or NK-cell population; (3) intrasinusoidal
cytotoxic T-cell or NK-cell infiltrates in bone marrow, spleen, or liver; and (4)
persistence of the abnormal T-cell or NK-cell population or unexplained cytopenia
for more than 6 months (Salama, BCJ, 2022). Baseline clinical characteristics, TTFT,
first line therapies, and OS were analyzed for all patients. The Mayo Clinic IRB approved
this study.
Results: We identified 217 patients who met inclusion criteria for LGLL diagnosis;
196 (90.3%) patients had T-LGLL and 21 (9.7%) patients had NK-LGLL. The median age
at diagnosis was 65 years [range 21-86], and 120 (55.3%) were male. 159 (73.3%) patients
had evidence of cytopenia at the time of diagnosis. Frequently associated clinical
characteristics included dependence on red cell or platelet transfusions (44.3%),
colony-stimulating factors (20.0%), or both (21.7%); splenomegaly (32.9%); history
of autoimmune disease (24.9%); history of hematologic disorder (18.4%) or malignancy
(5.6%); and constitutional symptoms (11.1%).
The median follow-up was 8.4 years; 133 patients died, and 111 patients were treated
for LGLL during this period. The median TTFT after diagnosis was 47.0 months (42.8
for T-LGLL and 68.0 for NK-LGLL) with a median of 1 (range 0-11) line of treatment.
First-line therapy consisted of methotrexate in 33 (29.7%) patients, cyclophosphamide
in 25 (22.5%), cyclosporine in 6 (5.4%), chlorambucil in 6 (5.4%), rituximab in 5
(4.5%), steroids alone in 17 (15.3%), and splenectomy in 7 (6.3%); 5 (4.5%) patients
received combination therapy and 2 (1.8%) received IVIG. Alemtuzumab, cladribine,
fludarabine, lenalidomide, and vincristine were first-line therapy in 1 (0.9%) patient
each. Response to first-line therapy (Lamy, Blood, 2011) consisted of complete response
(CR) in 31%, partial response (PR) in 33%, and treatment failure or progressive disease
in 36% patients. The median OS was 11.0 years: 11.0 years for T-LGLL and 8.1 years
for NK-LGLL (p = 0.90; Figure 1).
Image:
Summary/Conclusion: In this retrospective study of LGLL, we utilized stringent pathology
inclusion criteria to allow for analyses of a homogenous cohort of patients seen at
our institution. One in 4 patients with LGLL had a concomitant autoimmune condition,
and 25% had a concomitant hematologic condition or hematologic malignancy at the time
of initial diagnosis. Approximately a third of all patients who needed LGLL treatment
did not achieve a response to frontline therapy, suggesting that newer treatments
are needed to improve outcomes of this rare disease.
P1095: TREATMENT ATTRIBUTES FOR 3RD LINE FOLLICULAR LYMPHOMA TREATMENT DECISION-MAKING:
PHYSICIAN PERSPECTIVES FROM A SURVEY ACROSS WESTERN EUROPE AND THE UNITED STATES
P. C. Johnson1,*, A. Bailey2, N. Milloy2, E. Clayton2, R. G. Quek3, Q. Ma3
1Department of Medicine, Division of Hematology & Oncology, Massachusetts General
Hospital Cancer Center & Harvard Medical School, Boston, United States of America;
2Oncology, Adelphi Real World, Bollington, United Kingdom; 3Health Economics & Outcomes
Research, Regeneron Pharmaceuticals Inc., Sleepy Hollow, United States of America
Background: Follicular lymphoma (FL) is the second most common subtype of non-Hodgkin
lymphoma (NHL), and is the most common of the clinically indolent NHLs. Treatment
(tx) selection must integrate a myriad of clinical and patient factors, including
disease resistance, cumulative toxicities and treatment efficacy, as well as patient
quality of life (QoL) and the costs of therapies. However, real-world data regarding
physician perspectives on tx selection is lacking, especially at later lines of therapy.
Aims: This study aims to assess physician perspectives regarding key tx selection
attributes including those beyond efficacy and safety, as well as physician perception
of patient’s tx preference, when selecting a 3rd line (3L) FL tx.
Methods: Descriptive data were drawn from the Adelphi FL Disease-Specific Programme™,
a point-in-time study that was fielded June 2021-Jan 2022. Haematologists, haem-oncologists
and medical oncologists (med-onc) in EUR [France, Germany, Italy, Spain, United Kingdom
(UK)] and the US completed online surveys regarding self-reported demographics and
the top 7 tx attributes contributing to their rationale for 3L FL tx selection from
a comprehensive prespecified list of 28 attributes derived from expert opinions. Physicians
also provided their perception on patient’s preference regarding tx attributes for
choosing 3L tx.
Results: Of 251 physicians surveyed in EUR (France: n=46; Germany: n=40; Italy: n=43;
Spain: n=41; UK: n=31) and the US (n=50), 49% were haem-oncologists, 41% haematologists
and 10% med-oncs. 52% primarily worked at academic hospitals.
Progression free survival (74.1%), overall survival (70.1%), duration of response
(59.8%), evidence-based efficacy overall / overall response rate (57.4%) and ability
to achieve complete response (46.2%) were most frequently physician reported top 7
attributes for 3L tx selection.
Notably, long-term safety and impact on patients’ QoL were reported by 38.6% and 34.3%
of physicians, respectively.
Fewer physicians reported cost (4.8%), patient acceptability of lag time between leukapheresis
and CAR-T infusion (7.2%), patient acceptability of frequency of administration (8.0%),
less monitoring needed (8.8%) and fewer outpatient / inpatient consultations needed
(5.6% and 7.6% respectively) as their top 7 3L tx attributes considered.
Frequency of some 3L tx selection attributes reported differed substantially between
EUR and the US including long-term safety (35.8% vs 50.0%), impact on patients’ QoL
(37.8% vs 20.0%), and suitability for patients aged over 60 years (10.9% vs 22.0%).
Summary/Conclusion: Treatment efficacy and improved survival were the most important
attributes for physicians’ 3L FL tx selection whilst safety, impact on patient QoL,
frequency of inpatient / outpatient consultations, monitoring requirements, and cost
were amongst those attributes considered less important by physicians.
Despite literature indicating that patient QoL and cost of tx should both be key considerations
for physicians when choosing FL tx, our study highlights these are less commonly reported
attributes for treatment selection. Future study is warranted to further explore reasons
for physician treatment selection.
P1096: TREATMENT PATTERNS AND OUTCOMES IN RELAPSED/REFRACTORY MANTLE CELL LYMPHOMA
A. Bock1,*, K. Poonsombudlert2, M. Larson3, J. Gile1, R. Tawfiq1, S. Maliske2, M.
Maurer3, J. Cerhan4, X. Andrade-gonzalez1, A. Saliba1, J. Paludo1, D. Inwards1, S.
Ansell1, T. Witzig1, T. Habermann1, B. Link2, S. Ayyappan2, G. Nowakowski1, U. Farooq2,
Y. Wang1
1Division of Hematology, Mayo Clinic, Rochester; 2Division of Hematology, University
of Iowa, Iowa City; 3Division of Biostatistics; 4Division of Epidemiology, Department
of Quantitative Health Sciences, Mayo Clinic, Rochester, United States of America
Background: Mantle cell lymphoma (MCL) is an incurable B-cell lymphoma with heterogeneous
clinical presentation and no established standard of care. In the last two decades,
the pattern of frontline therapy for MCL has evolved, and multiple new agents became
available for relapsed/refractory (R/R) MCL. The pattern of treatment, its evolution
with time, and the association with treatment outcomes in R/R MCL are not well understood.
Aims: To characterize second line (2L) treatment patterns and outcomes in patients
with R/R MCL initially diagnosed in the rituximab era.
Methods: Patients with newly diagnosed MCL between August 2002 and April 2015 and
followed through 2021 were identified from the Mayo Clinic/University of Iowa Molecular
Epidemiology Resource (MER) prospective cohort study. Patients who initiated therapy
for R/R MCL after relapse or progression to first line (1L) therapy were included
in this analysis. Clinical characteristics and 1L and 2L therapies were abstracted
from MER and by chart review; treatment outcomes were analyzed.
Results: Among a total of 343 MCL patients with a median follow-up of 11.8 years,
188 had documented progression/relapse to 1L therapy, of which 170 had 2L treatment
information available for this analysis. At the time of 2L therapy, 123 (72%) patients
had an age >60, 136 (80%) were male, 132 (78%) had stage III/IV, and simplified MIPI
was 0-3 (low) in 37 (35%), 4-5 (intermediate) in 40 (37%), and 6-11 (high) in 30 (28%;
simplified MIPI missing in 63 patients). The median time from diagnosis to 2L was
27 months (range 0.5-141).
The patterns of 1L and 2L treatment are shown in Figure 1. 1L treatment included HiDAC-based
(n=31, 13 had autologous stem cell transplant [ASCT]), R-CHOP-like (n=79, 45 had ASCT),
R-Bendamustine (BR, n=22, 2 had ASCT), other systemic (n=29), and non-systemic (n=9).
15 patients (9%) received rituximab maintenance after induction or ASCT. 2L treatment
included salvage therapy followed by ASCT (n=13; salvage with HiDAC-based n=2, R-CHOP-like
n=5, BR n=3, platinum-based n=2, other n=1) or Allogeneic SCT (AlloSCT, n=5), HiDAC-based
n=9, R-CHOP-like n=6, BR n=33, BTK inhibitor (BTKi, n=21), other targeted therapy
(lenalidomide, bortezomib, mTOR inhibitors, n=26), other systemic (n=47), and non-systemic
(n=10).
The median follow-up from 2L therapy was 8.2 years. The objective response rate to
2L therapy was 71% (45% complete response). The median progression-free survival (PFS)
and overall survival (OS) were 1.1 and 4.0 years, respectively. The 2-year PFS rate
and 5-year OS rate were 55% (95% CI 36-84) and 71% (95% CI 49-100), respectively for
patients treated with BTKi at 2L, and 42% (95% CI 28-63) and 46% (95% CI 32-68), respectively,
for those treated with BR. In patients intended to undergo salvage therapy followed
by ASCT (n=18, n=13 transplanted) or AlloSCT (n=5, all transplanted), the 2-year PFS
rate and 5-year OS rate were 80% (95% CI 52-100) and 60% (95% CI 41-88), and 80% (95%
CI 52-100) and 61% (95% CI 42-88), respectively. Patients treated with HiDAC-based,
R-CHOP-like, other targeted therapy, or other systemic therapy had poorer outcomes
(2-year PFS 0-27%, 5-year OS 0-35%).
Image:
Summary/Conclusion: Second line treatment for MCL was very heterogenous in terms of
treatment and outcomes and was likely impacted by 1L treatment and other clinical
factors. The evolution of 1L and R/R MCL treatment patterns and the associated patient
characteristics and outcomes requires further exploration in larger datasets, given
extensive heterogeneity in practice patterns.
P1097: MEK-INHIBITORS IN TREATMENT OF LANGERHANS CELL HISTIOCYTOSIS
E. Burtsev1,*, G. Bronin1
1HSCT and BMT, Morozov Children’s Hospital, Moscow, Russia
Background: There are increasing data of targeted therapy efficacy of different types
of Langerhans cell histiocytosis (LCH) with inhibitors of BRAF-specific serin-threonine
kinase (BRAF-inhibitors) in cases with BRAFV600e mutation published last years. At
the same time there are lack of published data of MAPK/ERK pathway inhibitors (MEK-inhibitors)
use in pediatric patients with BRAF-negative forms of LCH.
Aims: Aim of the study is to evaluate efficacy and safety of MEK-inhibitor (cobimetinib)
in eight pediatric BRAFV600e-negative refractory LCH patients.
Methods: The study included 8 children with various forms of LCH. BRAFV600e mutation
was not found in any case included in the trial. All patients received therapy according
to the LCH IV protocol and were diagnosed with progression of LCH during or after
termination of the treatment. Monotherapy with cobimetinib was used as a second line
therapy in all cases. The response to the targeted therapy was assessed in accordance
with the international scale Response Evaluation Criteria in Solid Tumors (RECIST
v.1.1). The assessment of the toxicity was performed in accordance with the international
scale of Common Terminology Criteria for Adverse Events (CTCAE v.5.0).
Results: Complete response to cobimetinib was not achieved in any patient. Partial
response was established in 62,5% cases. One patient was diagnosed with disease progression
in three months after termination of the therapy. The incidence of adverse events
was high (75%). The most common side effects were diarrhea (75%), rush (50%) and electrolyte
disturbances (37,5%).
Summary/Conclusion: Cobimetinib monotherapy is effective in BRAF V600e- negative refractory
pediatric LCH patients. The response to the treatment can be delayed. All cases of
the toxicity were dose dependant and successfully resolved after dose correction.
Further research is needed to define duration of treatment and optimal dosage regimens.
P1098: OUTCOME OF FOLLICULAR LYMPHOMA PATIENTS IN MAINTENANCE TREATMENT WITH ANTICD20
MONOCLONAL ANTIBODIES IN SARS-COV2 ERA: RESULTS FROM A MULTICENTER, RETROSPECTIVE-
PROSPECTIVE ITALIAN STUDY.
A. Castellino1,*, C. Castellino1, C. Boccomini2, M. Clerico3, P. Nicoli4, A. Vanazzi5,
F. Fanelli6, T. Perrone7, F. Marchesi8, F. Cocito9, M. Merli10, S. Bigliardi11, B.
Mecacci12, V. Bozzoli13, G. Margiotta-Casaluci14, E. Meli15, A. Anastasia16, L. Farina17,
O. Annibali18, D. Gottardi19, M. Zanni20, A. Conconi21, C. Ciochetto22, N. Cenfra23,
F. Rotondo24, S. Ratotti25, A. Cuneo26, C. Selleri27, S. Galimberti28, M. Massaia1
1Hematology Unit, AO Santa Croce e Carle di Cuneo, Cuneo; 2Hematology Unit, Citta’
della Salute e della Scienza; 3Hematology Unit, Citta della Salute e della Scienza,
Torino; 4Hematology Unit, San Luigi Gonzaga Hospital and University, Orbassano; 5Hematology
Unit, Istituto Europeo di Oncologia, Milano; 6Hematology Unit, San Camillo Hospital,
Roma; 7Hematology Unit, Policlinico Hospital, Bari; 8Hematology Unit, IRCCS Regina
Elena National Cancer Institute, Roma; 9Hematology Unit, San Gerardo Hospital, Monza;
10Hematology Unit, Ospedale di Circolo ASST Sette Laghi, Varese; 11Hematology Unit,
Ospedale Civile, Sassuolo; 12Hematology Unit, University of Siena, Siena; 13Hematology
Unit, Vito Fazzi Hospital, Lecce; 14Hematology Unit, Department of Translation Medicine,
AOU Maggiore della Carità, Novara; 15Hematology Unit, ASST Grande Ospedale Metropolitano
Niguarda, Milano; 16Hematology Unit, Ospedali Civili, Brescia; 17Hematology Unit,
Istituto Nazionale Tumori, Milano; 18Hematology Unit, Università Campus Bio-Medico,
Roma; 19Hematology Unit, Ospedale Mauriziano, Torino; 20Hematology Unit, SS Arrigo
e Biagio Hospital, Alessandria; 21Hematology Unit, Ospedale degli Infermi, Biella;
22Hematology Unit, ASL TO4, Ivrea; 23Hematology Unit, Ospedale Santa Maria Goretti,
Latina; 24Hematology Unit, Ospedale Papardo, Messina; 25Hematology Unit, IRCCS Fondazione
San Matteo, Pavia; 26Hematology Unit, Sant’Anna Hospital, Ferrara; 27Hematology Unit,
AOU S. Giovanni di Dio e Ruggi D’Aragona Salerno, Salerno; 28Hematology Unit, Santa
Chiara Hospital and University, Pisa, Italy
Background: Maintenance in FL patients (pts) improves progression free survival (PFS).
SARS-Cov2 pandemic posed unique challenges for immunocompromised pts.
Aims: The aim is to evaluate the outcome of FL pts in maintenance with antiCD20-MoAb
during SARS-Cov2 pandemic and how suspension of therapy affected lymphoma outcome
and the risk of SARS-Cov2 infection and its morbidity and mortality.
Methods: This is an observational, multicenter, retrospective and prospective study.
Results: A total of 420 from 18 Italian Hematological Centers were included in the
analysis. Median age was 62 years old (range 27-91 years), 216 pts (51%) were male.
Main clinical characteristics of the population were: histological grade 1-2 vs 3A
in 288 (69%) vs 109 (26%), while not valuable in 23 (5%) pts; limited I-II vs advanced
III-IV stage in 57 (14%) vs 361 (86%) pts, not reported in 2 cases. FLIPI score was
low vs intermediate vs high in 71 (17%) vs 151 (36%) vs 192 (46%) patients, respectively,
not valuable in 6 cases. All 420 patients included were in maintenance treatment with
antiCD20 MoAb at the time of the onset of SARS-Cov2 pandemic (March 2020): 333 (79%)
pts were receiving maintenance after a first line, while 87 (21%) after a second line.
342 (81%) pts were receiving Rituximab, while 75 (18%) Obinutuzumab, 3 patients did
not start the planned maintenance because of pandemic spread. Status of disease after
induction was complete remission (CR) in 374 (89%), partial response (PR) in 41 (10%),
progressive disease (PD) in 1, not evaluated in 4 pts, respectively. At the end of
maintenance was CR in 265 (63%), PR in 19 (4%), stable disease (SD) in one and PD
in 14 (3%) patients, respectively, maintenance is stiil ongoing in 121 (29%) pts.
Because of SARS-Cov2 pandemic from March 2020 consequences on maintenance treatment
were: temporary suspension in 122 (29%), definitively interruption in123 (29%), no
modification in 175 (42%) of pts, respectively. Median number of maintenance treatment
administered at the time of SARS-Cov2 pandemic onset was 2 (range 1-12), median number
of courses administered at the time of analysis was 8 (range 0-12), in patients who
modified treatment because of pandemic median number of performed courses was 7 (range
0-11) and median number of lost cycles were 2 (range 1-12). Pts were divided into
two groups according to type of approach to maintenance during pandemic: pts who interrupted
maintenance (temporary or definitively): groups A (245 (58%) pts) vs pts who did not
modified maintenance: group B (175 (42%) pts). No differences in clinical characteristics,
type of therapy and response were observed between the two groups. 29(7%) relapses
were observed: 16 (7%) vs 13 (7%) in group A vs B, respectively. 70 (17%) pts experienced
SARS-Cov2 positivity: 47 (19%) vs 23 (13%) in group A vs B, respectively. 53 (76%)
pts had symptomatic COVID syndrome and 43 (61%) were hospitalized, with no differences
between the two groups. Anti-SARS-Cov2 vaccine was administered in 349 patients, serology
assessment was done in 46% of cases, showing 21 (13%) reactive vs 138 (87%) not reactive
pts, with no differences between the two groups. 21 (30%) pts died because of COVID:
9 (19%) vs 12 (52%) in groups A vs B, respectively.
Summary/Conclusion: Suspension of maintenance treatment during SARS-Cov2 pandemic
did not show a protection in terms of SARS-Cov2 positivity and morbidity. A trend
in lower mortality is suggested. No differences in terms of relapse rate were observed,
but longer follow up is needed.
P1099: THE SPECTRUM OF SECOND PRIMARY MALIGNANCIES AND CAUSE-SPECIFIC MORTALITY AMONG
PATIENTS WITH FOLLICULAR LYMPHOMA IN THE UNITED STATES: A POPULATION-BASED STUDY.
K. Chamarti1,*, V. Rudraraju2, S. Kiani1
1Department of Internal Medicine, Texas Tech University Health Sciences Center at
Permian Basin, Odessa, United States of America; 2Department of Internal Medicine,
Jawaharlal Nehru Medical College, Belgaum, India
Background: Follicular lymphoma (FL) is the second most common type of non-Hodgkin
lymphoma (NHL). FL accounts for 35 percent of NHLs and has an estimated incidence
of 3.18 cases per 1,00,000 people in the United States. The clinical course of FL
is variable, and it has median overall survival of 10 to 12 years. In the era of targeted
therapies with anti-CD20 antibodies, there is data lacking on the spectrum of second
primary malignancies (SPMs) and cause-specific mortality in patients with follicular
lymphoma.
Aims: In the current study, we aim to identify the risk of SPM and cause specific
mortality in FL patients in the era of targeted therapies with anti CD-20 antibodies
using a national registry from the United States.
Methods: Using the National Cancer Institute’s Surveillance, Epidemiology, and End
Results (SEER)-18, we conducted a retrospective study with patients diagnosed with
FL (ICD-0-3 code 9690/3) between 2012 and 2018. We calculated the risk of SPMs developing
≥ 2 months using standardized incidence ratios (SIRs) and cause-specific mortality
by standardized mortality ratios (SMRs) and absolute excess risk (AER)/10,000 population.
Results: A total of 6,215 patients with follicular lymphoma (FL) were included in
this study. The median age at diagnosis was 66 years (range 3 to 85 years) and the
median follow-up duration was 31 months (range 0 to 81 months). A total of 355 patients
(5.7%) developed 376 SPMs, of which 56% were solid organ malignancies, and 44% were
hematologic neoplasms. The latency period for SPM development was 20 months (range
2 to 80 months). The SPM risk for the cohort was significantly elevated (SIR= 1.99,
95% confidence interval [CI] = 1.79-2.2, P<0.05) when compared with the general population.
The SIR was higher in females (SIR= 2.16, 95% CI=1.85-2.51) when compared to males
(SIR= 1.86, 95% CI= 1.61- 2.13). The SIR for age groups were as follows: age < 65
years (SIR= 2.84, 95% CI= 2.43-3.31), age > 65 years (SIR= 1.59, 95% CI= 1.38-1.82).
The risk for hematopoietic SPMs was higher (SIR= 8.66, 95% CI= 7.33-10.15) than solid
organ tumors (SIR= 1.30, 95% CI= 1.13-1.49). Specific SPMs with the highest risk included
NHL (SIR= 16.24, 95% CI= 13.62-19.22), Hodgkin lymphoma (SIR= 14.31, 95% CI= 5.25-31.15),
followed by thyroid cancer (SIR= 4.63, 95% CI= 2.53-7.76), kidney and renal pelvis
cancer (SIR= 1.43, 95% CI= 1.55-4.14), and lung cancer (SIR= 2.16, 95% CI= 1.63-2.79).
The risk of developing Hodgkin lymphoma, thyroid, and kidney cancers was high in the
first year of FL diagnosis. While, the risk for NHL was elevated throughout the follow-up
period. At last follow-up, 724 (11.5%) patients had died, of which 65% were due to
malignant neoplasms. The risk of mortality due to all malignant cancers combined was
statistically significant (SMR= 6.26, 95% CI= 2.00-2.31), largely due to mortality
from NHL (SMR= 139.5, 95% CI= 126-154, AER= 286.8), leukemia (SMR= 5.23, 95% CI= 2.99-8.49,
AER= 9.63), and infectious diseases (SMR= 2.58, 95% CI= 1.04-5.31, AER=3.19).
Summary/Conclusion: Our study reveals that despite therapeutic advances in FL, hematologic
neoplasms (including NHL and leukemia) remain the major cause of death in our cohort.
Patients with FL are at higher risk of SPM than the general population, most notably
for NHL, Hodgkin lymphoma, followed by thyroid, kidney, and lung cancer. These patients
may benefit from cancer-specific screening during follow-up.
P1100: BRUIN MCL-321: A PHASE 3 OPEN-LABEL, RANDOMIZED STUDY OF PIRTOBRUTINIB VS INVESTIGATOR
CHOICE OF BTK INHIBITOR IN PATIENTS WITH PREVIOUSLY TREATED, BTK INHIBITOR NAÏVE MCL
(TRIAL IN PROGRESS)
T. A. Eyre1, N. N. Shah2, S. Le Gouill3, M. Dreyling4,*, E. Vandenberghe5, W. Jurczak6,
Y. Wang7, C. Y. Cheah8, M. Gandhi9, C. Chay10, J. P. Sharman11, D. J. Andorsky12,
M. Yin13, M. Balbas13, J. Kherani13, M. L. Wang14
1Oxford University Hospitals NHS Foundation Trust, Churchill Cancer Center, Oxford,
United Kingdom; 2Medical College of Wisconsin, Milwaukee, United States of America;
3Service d’hématologie clinique du CHU de Nantes, INSERM CRCINA Nantes-Angers, NeXT
Université de Nantes, Nantes, France; 4Medicine III, LMU University Hospital, Munich,
Germany; 5Hope Directorate St. James Hospital, Dublin, Ireland; 6Maria Sklodowska-Curie
National Research Institute of Oncology, Krakow, Poland; 7Division of Hematology,
Mayo Clinic, Rochester, United States of America; 8Linear Clinical Research and Sir
Charles Gairdner Hospital, Perth, Australia; 9Virginia Cancer Specialists, Fairfax;
10Messino Cancer Centers, Asheville; 11Willamette Valley Cancer Institute and Research
Center, US Oncology Research; 12Rocky Mountain Cancer Centers, US Oncology Research,
Eugene; 13Loxo Oncology at Lilly, Stamford; 14University of Texas MD Anderson Cancer
Center, Houston, United States of America
Background: Covalent Bruton’s Tyrosine Kinase (BTK) inhibitors (BTKi) have transformed
the management of relapsed mantle cell lymphoma (MCL), but these treatments are not
curative and the majority of patients will require additional treatment. Covalent
BTKi share pharmacologic liabilities (e.g. low oral bioavailability, short half-life)
that collectively may lead to suboptimal BTK target coverage especially in rapidly
proliferating tumors with high BTK protein turnover such as MCL. To address these
limitations, pirtobrutinib, a highly selective, non-covalent BTKi that inhibits both
wild type (WT) and C481-mutated BTK with equal low nM potency was developed. In the
phase 1/2 BRUIN study, pirtobrutinib achieved pharmacokinetic exposures that exceeded
its BTK IC96 at trough, was well tolerated, and demonstrated promising efficacy in
heavily pretreated, poor-prognosis MCL patients, most of whom had prior treatment
with a covalent BTKi.
Aims: The purpose of this randomized study is to determine whether pirtobrutinib is
superior to investigator’s choice of covalent BTKi in patients with previously treated
MCL.
Methods: BRUIN MCL-321 is a randomized, open-label, global phase 3 study comparing
pirtobrutinib monotherapy versus investigator’s choice of covalent BTKi monotherapy
(ibrutinib, acalabrutinib, or zanubrutinib) in patients with previously treated, BTKi
naïve MCL. Approximately 500 patients will be randomized 1:1. Randomization will be
stratified by sMIPI risk (low/intermediate vs high), comparator BTKi (ibrutinib vs
acalabrutinib/ zanubrutinib), and number of prior lines of therapy (1 vs ≥ 2).
Eligible patients are adults aged ≥18 years with a confirmed diagnosis of MCL (cyclin
D1 overexpression, and ≥ 1 B-cell marker) who have received ≥ 1 prior line of systemic
therapy for MCL that did not include a prior BTKi. Patients must have measurable disease
per Lugano criteria and must have progressed on or relapsed following the most recent
line of therapy prior to study enrollment. Key exclusion criteria include a history
of current or prior central nervous system (CNS) involvement, significant cardiovascular
disease, stroke, or intracranial hemorrhage within 6 months of randomization, and
allogeneic stem cell transplant (SCT), autologous SCT or chimeric antigen receptor
(CAR) T-cell therapy within 60 days of randomization.
The primary endpoint is progression-free survival (PFS) per Lugano criteria assessed
by an independent review committee (IRC), with the goal of demonstrating superiority
of pirtobrutinib over investigator’s choice of covalent BTKi. Secondary endpoints
include overall response rate (ORR), duration of response (DoR), investigator-assessed
PFS per Lugano criteria, overall survival, event-free survival, time to treatment
failure, time to next treatment, PFS2 (time from randomization to disease progression
on next line of treatment or death from any cause), safety and tolerability, and patient
reported outcomes. This global study is currently enrolling patients (NCT04662255).
Results: This study is a Trial in Progress. The results will be presented at a later
date.
Summary/Conclusion: This study is a Trial in Progress. The conclusions will be presented
at a later date.
P1101: PIRTOBRUTINIB, A HIGHLY SELECTIVE, NON-COVALENT (REVERSIBLE) BTK INHIBITOR
IN PREVIOUSLY TREATED MANTLE CELL LYMPHOMA: UPDATED RESULTS FROM THE PHASE 1/2 BRUIN
STUDY
T. A. Eyre1,*, M. L. Wang2, N. N. Shah3, A. J. Alencar4, J. N. Gerson5, M. R. Patel6,
B. Fakhri7, E. Vandenberghe8, W. Jurczak9, X. N. Tan10, K. L. Lewis10, T. Fenske3,
Y. Wang11, C. C. Coombs12, I. Flinn13, D. Lewis14, S. Le Gouill15, M. Gandhi16, C.
Chay17, M. L. Palomba18, J. A. Woyach19, J. M. Pagel20, N. Lamanna21, J. P. Sharman22,
D. J. Andorsky23, J. B. Cohen24, M. A. Barve25, P. Ghia26,27, M. Yin20, P. L. Zinzani28,
C. Ujjani29, Y. Koh30, K. Izutsu31, E. Lech-Maranda32, J. Kherani20, C. Tam33, S.
Sundaram34, B. Nair20, D. E. Tsai20, M. Balbas20, A. R. Mato18, C. Y. Cheah10
1Oxford University Hospitals NHS Foundation Trust, Churchill Cancer Center, Oxford,
United Kingdom; 2MD Anderson Cancer Center, Houston; 3Medical College of Wisconsin,
Milwaukee; 4Sylvester Comprehensive Cancer Center, Miami; 5University of Pennsylvania,
Philadelphia; 6Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota;
7University of California San Francisco, San Francisco, United States of America;
8Hope Directorate St. James Hospital, Dublin, Ireland; 9Maria Sklodowska-Curie National
Research Institute of Oncology, Krakow, Poland; 10Linear Clinical Research and Sir
Charles Gairdner Hospital, Perth, Australia; 11Division of Hematology, Mayo Clinic,
Rochester; 12University of North Carolina at Chapel Hill, Chapel Hill; 13Sarah Cannon
Research Institute, Nashville, United States of America; 14University Hospitals Plymouth
NHS, Plymouth, United Kingdom; 15Service d’hématologie clinique du CHU de Nantes,
INSERM CRCINA Nantes-Angers, NeXT Université de Nantes, Nantes, France; 16Virginia
Cancer Specialists, Fairfax; 17Messino Cancer Centers, Asheville; 18Memorial Sloan
Kettering Cancer Center, New York; 19The Ohio State University Comprehensive Cancer
Center, Columbus; 20Loxo Oncology at Lilly, Stamford; 21Herbert Irving Comprehensive
Cancer Center, Columbia University, New York; 22Willamette Valley Cancer Institute
and Research Center, US Oncology Research; 23Rocky Mountain Cancer Centers, US Oncology
Research, Eugene; 24Winship Cancer Institute, Emory University, Atlanta; 25Mary Crowley
Cancer Research, Dallas, United States of America; 26Università Vita-Salute San Raffaele
and IRCCS Ospedale San Raffaele, Milan, Italy; 27Medicine III, LMU University Hospital,
Munich, Germany; 28Institute of Hematology “Seràgnoli” University of Bologna, Bologna,
Italy; 29Fred Hutchinson Cancer Research Center, Seattle, United States of America;
30Internal Medicine, Seoul National University Hospital, Seoul, South Korea; 31National
Cancer Center Hospital, Tokyo, Japan; 32Institute of Hematology and Transfusion Medicine,
Warsaw, Poland; 33Peter MacCallum Cancer Center, Royal Melbourne Hospital, and University
of Melbourne, Melbourne, Australia; 34Hematology and Medical Oncology, Tisch Cancer
Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
Background: Covalent BTK inhibitors (BTKi) have transformed the management of mantle
cell lymphoma (MCL), but these treatments are not curative and the majority of patients
(pts) will require additional treatment. Covalent BTKi share pharmacologic liabilities
(e.g. low oral bioavailability, short half-life) that collectively may lead to suboptimal
BTK target coverage, for example in rapidly proliferating tumors with high BTK protein
turnover such as MCL. To address these limitations, pirtobrutinib, a highly selective,
non-covalent (reversible) BTKi that inhibits both wild type (WT) and C481-mutated
BTK with equal low nM potency was developed.
Aims: BRUIN is a multicenter phase 1/2 study (NCT03740529) of oral pirtobrutinib monotherapy
in pts with advanced B-cell malignancies who have received ≥2 prior therapies.
Methods: Pirtobrutinib was dose escalated in a standard 3 + 3 design in 28-day cycles.
The primary objective for phase 1 was to determine the recommended phase 2 dose (RP2D)
and the primary objective of phase 2 was overall response rate (ORR); secondary objectives
included duration of response (DoR), progression-free survival (PFS), overall survival
(OS), safety and tolerability, and pharmacokinetics. Efficacy evaluable pts included
all dosed pts who underwent their first response evaluation or discontinued therapy.
Response was assessed every 8 weeks from cycle 3, and every 12 weeks from cycle 13
and was measured according to Lugano Classification. Safety was assessed in all pts
(CLL/SLL and NHL).
Results: As of 27 September 2020, 323 pts (170 CLL/SLL, 61 MCL, 26 WM, 26 DLBCL, 13
MZL, 12 FL, 9 RT, and 6 other NHL [other transformation, B-PLL and hairy cell leukemia])
were treated on 7 dose levels (25-300mg QD). Median age was 69 (range 50-87) years
for MCL pts. Among the 61 MCL pts, median number of prior lines of therapy was 3 (range,
1-8) and a majority of them had received a prior BTKi (93%), an anti-CD20 antibody
(98%) or chemotherapy (92%). No DLTs were reported and MTD was not reached (n=323).
200mg QD was selected as the RP2D. Fatigue (20%), diarrhea (17%) and contusion (13%)
were the most frequent treatment-emergent adverse events regardless of attribution
or grade seen in ≥10% of pts (n=323). The most common adverse event of grade ≥3 was
neutropenia (10%). Treatment-related hemorrhage and hypertension occurred in 5 (2%)
and 4 (1%) pts, respectively. Five (1%) pts discontinued due to treatment-related
adverse events. At the efficacy cutoff date, 52 prior BTKi treated MCL pts were efficacy
evaluable with an ORR of 52% (95% confidence interval 38-66; 13 complete response
(CR) [25%], 14 partial response (PR) [27%], 9 stable disease (SD) [17%]), 11 progressive
disease (PD) [21%] and 5 [10%] discontinued prior to first response assessment). Median
follow up was 6 months (range 0.7-18.3+). Responses were observed in 9/14 pts (64%)
with prior autologous or allogeneic stem cell transplant, and 2 of 2 with prior CAR-T
cell therapy.
Summary/Conclusion: Pirtobrutinib demonstrated promising efficacy in heavily pretreated,
poor-prognosis MCL following multiple prior lines of therapy, including a covalent
BTKi. Pirtobrutinib was well tolerated and exhibited a wide therapeutic index. Updated
data, including approximately 60 new pts with MCL and an additional 10 months since
the prior data cut will be presented.
P1102: A PHASE 1 STUDY OF PARSACLISIB IN COMBINATION WITH RITUXIMAB, BENDAMUSTINE
+ RITUXIMAB, OR IBRUTINIB IN PATIENTS WITH PREVIOUSLY TREATED B-CELL LYMPHOMA (CITADEL-112):
PRELIMINARY SAFETY RESULTS
J.-M. Sancho1,*, A. Lopez-Guillermo2, P. Abrisqueta3, A. Kumar4, R. Cordoba5, M. Tani6,
W. Zhao7, E. Rappold7, P. Langmuir7, M. Mims8
1Clinical Hematology Department, Hospital Germans Trias i Pujol, Institut Català d’Oncologia;
2Clinic Barcelona; 3Department of Hematology, Vall d’Hebron Institute of Oncology
(VHIO), Hospital Universitari Vall d’Hebron, Barcelona, Spain; 4University of Arizona,
Tucson, United States of America; 5Lymphoma Unit, Department of Hematology, Fundacion
Jimenez Diaz University Hospital, Health Research Institute IIS-FJD, Madrid, Spain;
6Haematology Unit, Santa Maria delle Croci Hospital, Ravenna, Italy; 7Incyte Corporation,
Wilmington; 8Department of Medicine, Hematology/Oncology, Baylor College of Medicine,
Houston, United States of America
Background: Rituximab-based chemoimmunotherapy regimens are backbone treatment (Tmt)
for both indolent (follicular [FL], marginal zone [MZL]) and aggressive (diffuse large
B-cell [DLBCL], mantle cell [MCL]) B-cell lymphomas. Standard of care (SoC) for relapsed
or refractory (R/R) disease includes anti-CD20 in combination with chemotherapy and
targeted therapies, such as Bruton’s tyrosine kinase inhibitors (eg, ibrutinib) and
phosphoinositide 3-kinase (PI3K) inhibitors. Parsaclisib is a potent and highly selective
next generation PI3Kδ inhibitor that is currently being investigated in hematological
malignancies.
Aims: CITADEL-112 (NCT03424122) is an open-label phase 1 study evaluating the safety
and tolerability of adding parsaclisib to investigator choice SoC Tmt rituximab (RIT),
RIT + bendamustine (BEN), or ibrutinib (IBR) in patients (pts) with R/R B-cell lymphoma.
Methods: Enrolled pts were ≥18 years and had histologically confirmed DLBCL, FL, MCL,
or MZL, ECOG PS 0–2, were R/R to ≥1 (≥2 for FL) prior systemic therapy, and ineligible
for stem cell transplant. Pts received parsaclisib 20 mg orally once daily (QD) for
8 weeks then 20 mg once weekly (QW) in combination with either: RIT 375 mg/m2 IV QW
for 4 doses in cycle 1 (± cycle 2) (Tmt A); RIT 375 mg/m2 IV on day 1 + BEN 90 mg/m2
on day 1 and day 2 of each 28-day cycle for ≤6 cycles (Tmt B); or IBR 560 mg QD (Tmt
C). Pts received treatment until disease progression, unacceptable toxicity, or withdrawal.
Results: At data cutoff (May 14, 2021), 50 pts were treated (16 pts each in Tmt A
and C, 18 pts in Tmt B) and 13 pts were ongoing treatment (3 pts in Tmt A, 8 pts in
Tmt B, 2 pts in Tmt C). Most pts had received ≥2 prior systemic treatments (81.3%,
61.1%, and 68.8% in Tmt A [range 1–4], B [range 1–4], and C [range 1–7], respectively).
The most common reasons for discontinuation were progressive disease (56.3%, 38.9%,
and 50.0%) and adverse events (AEs) (12.5%, 11.1%, and 6.3% in Tmt A, B, and C, respectively).
One pt in Tmt B experienced a dose-limiting toxicity of grade 4 neutropenia for >14
days. All pts experienced at least 1 treatment-emergent AE (TEAE); in Tmt A, 75.0%
had grade ≥3 and 37.5% had serious TEAEs; Tmt B, 83.3% had grade ≥3 and 27.8% had
serious TEAEs; and Tmt C, 62.5% had grade ≥3 and 43.8% had serious TEAEs. Common any-grade
TEAEs (≥30%) included neutropenia (62.5%), diarrhea (37.5%), and anemia (31.3%) in
Tmt A; neutropenia (50.0%), abdominal pain, asthenia, diarrhea, and nausea (each 33.3%)
in Tmt B; neutropenia (50.0%) and increased ALT and increased AST (each 37.5%) in
Tmt C. Most common grade ≥3 TEAEs (≥15%) were neutropenia (50.0%) and diarrhea (18.8%)
in Tmt A, and neutropenia (38.9% and 25.0%) in Tmt B and Tmt C, respectively. Serious
TEAEs occurring in >1 pt were COVID-19, diarrhea, and pneumonia (n = 2 each) in Tmt
A, and atrial fibrillation (n = 2) in Tmt C. TEAEs with fatal outcome were reported
in 2 pts in Tmt A (COVID-19 and COVID-19 pneumonia [n = 1], interstitial lung disease
[n = 1]) and 1 pt in Tmt C (COVID-19, acute kidney injury). Parsaclisib dose interruption
or dose reduction due to TEAEs occurred in 75.0% and 18.8% of pts, respectively, in
Tmt A; 66.7% and 27.8% of pts, respectively, in Tmt B; and 56.3% and 18.8% of pts,
respectively, in Tmt C.
Summary/Conclusion: Parsaclisib 20 mg QD for 8 weeks followed by 20 mg QW can be safely
combined with RIT, RIT + BEN, or IBR in pts with R/R B-cell lymphomas. The tolerability
profile of the combination regimens was manageable, with no unexpected safety concerns.
P1103: INMIND: A PHASE 3 STUDY OF TAFASITAMAB PLUS LENALIDOMIDE AND RITUXIMAB VERSUS
PLACEBO PLUS LENALIDOMIDE AND RITUXIMAB FOR RELAPSED/REFRACTORY FOLLICULAR LYMPHOMA
(FL) OR MARGINAL ZONE LYMPHOMA (MZL)
L. H. Sehn1,*, K. Hübel2, S. Luminari3, A. Salar4, B. E. Wahlin5, A. K. Gopal6, C.
Bonnet7, S. Paneesha8, M. Trneny9, H. Mashegu10, C. Lihou10, D. Li10, C. W. Scholz11
1BC Cancer Centre for Lymphoid Cancer and The University of British Columbia, Vancouver,
Canada; 2Department of Internal Medicine I Oncology and Hematology, University Hospital
Cologne, Cologne, Germany; 3Hematology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio
Emilia, Italy; 4Department of Haematology, Hospital del Mar-IMIM, Barcelona, Spain;
5Department of Medicine, Unit of Hematology, Karolinska Institute, Stockholm, Sweden;
6Division of Medical Oncology, University of Washington Medicine, Seattle, United
States of America; 7Clinical Hematology, Centre Hospitalier Universitaire, University
of Liège, Liège, Belgium; 8University Hospitals Birmingham NHS Foundation Trust, Birmingham,
United Kingdom; 9First Department of Medicine, First Faculty of Medicine, Charles
University, General Hospital, Prague, Czechia; 10Incyte Corporation, Wilmington, United
States of America; 11Department of Hematology and Oncology, Vivantes Klinikum Am Urban,
Berlin, Germany
Background: Patients (pts) with indolent non-Hodgkin lymphoma (NHL) subtypes FL or
MZL respond to first-line treatment but relapse is common; there is no standard treatment
for pts with relapsed/refractory (R/R) FL or MZL. Tafasitamab (TAFA) is an Fc-engineered
humanized monoclonal antibody (mAb) against CD19, which is broadly expressed in FL
and MZL and regulates proliferation via B-cell receptor signaling. In preclinical
studies, TAFA has shown activity against NHL cell lines in combination with rituximab
(anti-CD20 mAb) and lenalidomide (LEN). TAFA monotherapy demonstrated promising clinical
activity in a phase 2a study in pts with R/R NHL (NCT01685008), with an ORR of 29%
(n/N=10/34) in pts with FL and 33% (n/N=3/9) in pts with MZL. In an ongoing phase
2, single-arm study (L-MIND, NCT02399085), TAFA + LEN followed by TAFA alone demonstrated
an ORR of 57.5% (n/N=46/80) with CR of 40% (n/N=32/80) and median DOR of 34.6 mo (95%
CI: 26.1–NR), in pts with R/R diffuse large B-cell lymphoma (FDA approved indication).
These observations suggest a potential clinical benefit of TAFA + LEN and rituximab
for pts with R/R FL or MZL.
Aims: Phase 3 double-blind, placebo-controlled, randomized study designed to investigate
whether TAFA + LEN and rituximab provides improved clinical benefit compared with
LEN and rituximab in pts with R/R FL or MZL.
Methods: Pts will be randomized 1:1 to receive TAFA (12 mg/kg IV on days 1, 8, 15,
and 22 of 28-day cycle [cycles 1–3], then days 1 and 15 [cycles 4–12]) + LEN (20 mg
PO QD [or starting dose 10 mg PO QD if creatinine clearance ≥30 to <60 mL/min], days
1–21/cycle for 12 cycles) and rituximab (375 mg/m2 IV on days 1, 8, 15, and 22 of
cycle 1, then day 1 of cycles 2–5), or placebo (0.9% saline solution IV) + LEN and
rituximab. Stratification will include progression of disease within 24 mo (FL; yes
vs no), refractoriness to prior anti-CD20 mAb therapy (FL; yes vs no), and number
of prior lines of therapy (FL or MZL; <2 vs ≥2). Radiological (PET) assessment will
be performed at baseline, every 12 (± 2) weeks in year 1, 16 (± 2) weeks in years
2–3, and 24 (± 3) weeks in years 4–6; additional assessment performed within 4 (±
2) weeks if progressive disease confirmed before end-of-treatment (EOT) visit. The
primary study endpoint is PFS (investigator assessed [INV] by Lugano 2014 criteria)
for pts with FL. Key secondary endpoints are PFS (INV) in the overall population (FL
and MZL), PET-CR rate (INV) at EOT (90 days after last treatment) and OS in pts with
FL. Other secondary endpoints include PET-CR rate (INV) at EOT and OS in the overall
population, and ORR (INV), DOR (INV), health related quality of life, safety, and
minimum residual disease-negativity rate at EOT in FL and the overall population.
Inclusion criteria include age ≥18 y, histologically confirmed FL (grade 1, 2, or
3a) or MZL (nodal, splenic, or extranodal), documented R/R disease, ≥1 prior systemic
anti-CD20 therapy (including anti-CD20 refractory disease), ECOG PS ≤2, adequate systemic
organ function, and high tumor burden (per GELF criteria). Exclusion criteria include
prior rituximab + LEN treatment, history of radiotherapy for other diseases (≥25%
of bone marrow), nonhematologic malignancy, congestive heart failure (LVEF <50%),
active systemic infection, known CNS lymphoma, or severe immunocompromised state.
inMIND (NCT04680052, EudraCT2020-004407-13) is currently enrolling pts; planned enrollment
is 528 pts with R/R FL and 60–90 pts with R/R MZL across North America, Europe, and
Asia Pacific.
Results: Not applicable.
Summary/Conclusion: Not applicable.
P1104: A PHASE 1 STUDY EVALUATING SAFETY AND EFFICACY OF PARSACLISIB IN COMBINATION
WITH BENDAMUSTINE + OBINUTUZUMAB IN PATIENTS WITH RELAPSED OR REFRACTORY FOLLICULAR
LYMPHOMA (CITADEL-102)
M. Hamadani1,*, M. Coleman2, R. Boccia3, J. Duras4, M. Hutchings5, P. L. Zinzani6,
R. Cordoba7, M. B. Oreiro8, V. Williams9, M. Stouffs9, P. Langmuir9, J.-M. Sancho10
1Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee; 2Clinical
Research Alliance Inc, Westbury; 3Center for Cancer and Blood Disorders, Bethesda,
United States of America; 4Department of Haematooncology, University Hospital Ostrava
and Faculty of Medicine, University of Ostrava, Ostrava, Czechia; 5Department of Haematology
and Phase 1 Unit, Rigshospitalet, Copenhagen, Denmark; 6Institute of Hematology “Seràgnoli”,
University of Bologna, Bologna, Italy; 7Lymphoma Unit, Department of Hematology, Fundación
Jimenez Diaz University Hospital; 8Instituto de Investigación Sanitaria Gregorio Marañón,
Madrid, Spain; 9Incyte Corporation, Wilmington, United States of America; 10Clinical
Hematology Department, Institut Català d’Oncologia-Hospital Germans Trias i Pujol,
Barcelona, Spain
Background: Patients (pts) with follicular lymphoma (FL) generally respond well to
first-line CD20-targeted therapies, such as obinutuzumab or rituximab-based regimens.
However, many pts relapse and studies suggest that each subsequent relapse is associated
with shorter durations of response to the next treatment. Parsaclisib is a potent
and highly selective next generation PI3Kδ inhibitor. The combination of bendamustine
+ obinutuzumab is approved for pts with relapsed/refractory (R/R) FL. We hypothesized
that adding parsaclisib may improve clinical benefit with a manageable safety profile
in this pt population.
Aims: CITADEL-102 (NCT03039114) is an open-label, phase 1, dose-finding study that
investigated safety and efficacy of parsaclisib in combination with bendamustine +
obinutuzumab in pts with R/R FL following rituximab-containing regimens.
Methods: Pts enrolled were ≥18 years with histologically confirmed CD20-positive FL,
R/R to any prior rituximab-containing regimen, ECOG PS 0–2, ≥1 measurable lesion,
and ≤4 prior therapies. Pts received parsaclisib 20 mg orally once daily (QD) for
8 weeks then 20 mg once weekly (QW); bendamustine 90 mg/m2 infusion on days 1 and
2 of cycles 1–6; and obinutuzumab 1000 mg infusion on days 1, 8, and 15 of cycle 1,
and day 1 of cycles 2–6, and on every second cycle of cycles 8–30 in pts having complete
response/complete metabolic response (CR/CMR), partial response/partial metabolic
response (PR/PMR), or stable disease/no metabolic response. Part 1 (safety run-in)
used a 3 + 3 design with dose de-escalation to identify the maximum tolerated dose
(MTD) of parsaclisib in combination with bendamustine + obinutuzumab. In Part 2 (dose
expansion), the safety and efficacy of this combination were further evaluated. The
primary study endpoint was safety and tolerability; secondary endpoints included efficacy
outcomes (ORR, DOR, PFS, and OS).
Results: A total of 26 pts were enrolled and treated; median (range) age was 65.0
(44–80) years, 25 (96.2%) had ECOG PS ≤1, 11 (42.3%) had ≥2 prior systemic therapies,
and 6 (23.1%) had received prior bendamustine. Median (range) parsaclisib exposure
was 10.6 (0.4–32.8) months. Main reasons for treatment discontinuation included adverse
events (AEs) (8 pts, 30.8%) and progressive disease (6 pts, 23.1%). All pts experienced
treatment-emergent AEs (TEAEs); most common any-grade TEAEs (≥10 pts) were pyrexia
(53.8%), neutropenia (50%), diarrhea (46.2%), thrombocytopenia, and nausea (each 38.5%).
Grade ≥3 TEAEs were experienced by 88.5% of pts; most common grade ≥3 TEAEs (≥2 pts)
were neutropenia (34.6%), febrile neutropenia (23.1%), thrombocytopenia (19.2%), ALT
and AST increase (each 11.5%), and diarrhea, neutrophil count decreased, and rash
maculopapular (each 7.7%). One of 6 evaluable pts in Part 1 had a DLT of grade 4 QTc
elongation. The MTD was not reached, and parsaclisib 20 mg QD for 8 weeks then 20 mg
QW was the selected dosage for dose expansion in Part 2. Treatment discontinuation
due to TEAEs was 30.8%, 7.7%, and 15.4% for parsaclisib, bendamustine, and obinutuzumab,
respectively. One fatal TEAE (COVID-19 pneumonia) occurred. ORR (95% CI) as reported
by the investigator was 76.9% (56.4–91.0), with 17 pts (65.4%) achieving CR/CMR and
3 pts (11.5%) achieving PR/PMR as the best overall response. Median DOR, PFS, and
OS were not reached.
Summary/Conclusion: Parsaclisib in combination with bendamustine + obinutuzumab appears
to have a manageable safety profile and demonstrated promising efficacy in pts with
R/R FL.
P1105: FINAL RESULTS OF THE PHASE I/II HOVON124/ECWM-R2 STUDY INCLUDING 2-YEAR RITUXIMAB
MAINTENANCE AFTER INDUCTION WITH IXAZOMIB, RITUXIMAB AND DEXAMETHASONE IN RELAPSED
WALDENSTRÖM’S MACROGLOBULINEMIA
M.-A. Dimopoulos1,*, K. Amaador2,3, M. C. Minnema4, K. Nasserinejad5, M. Kap5, E.
Kastritis1, M. Gavriatopoulou1, W. Kraan3,6, M. E. D. Chamuleau7, D. Deeren8, L. Tick9,
J. K. Doorduijn10, F. Offner11, L. H. Böhmer12, R. D. Liu2, S. T. Pals3,6, J. M. Vos2,3,
M. J. Kersten2,3
1Department of Clinical Therapeutics, National and Kapodistrian University of Athens,
School of Medicine, Athens, Greece; 2Department Of Hematology, Amsterdam UMC, University
of Amsterdam, Cancer Center Amsterdam; 3LYMMCARE (Lymphoma and Myeloma Center Amsterdam),
Amsterdam; 4Department of Hematology, University Medical Center Utrecht, University
Utrecht, Utrecht; 5HOVON Data Center, Department of Hematology, Erasmus MC Cancer
Institute, Rotterdam; 6Department of Pathology, Amsterdam UMC, University of Amsterdam,
Cancer Center Amsterdam; 7Department of Hematology, Amsterdam UMC, VU University,
Amsterdam, Netherlands; 8Department of Hematology, AZ Delta, Roeselare, Belgium; 9Department
of Hematology, Maxima Medical Center, Eindhoven; 10Department of Hematology, Erasmus
MC Cancer Institute, Rotterdam, Netherlands; 11Department of Hematology, University
Hospital Gent, Gent, Belgium; 12Department of Hematology, Haga Teaching Hospital,
The Hague, Netherlands
Background: In the phase I/II HOVON124/ECWM-R2 trial, induction treatment with the
combination of ixazomib, subcutaneous (s.c.) rituximab and dexamethasone (IRd) showed
promising efficacy with manageable toxicity in patients with relapsed Waldenström’s
Macroglobulinemia (WM).
Aims: Here we report the final analysis of the trial after two years of rituximab
maintenance and with a median follow-up (FU) on study of 45.6 months (range, 12.4-72.2).
Methods: In total, 59 patients were enrolled (median age, 69 years; range, 46-91 years)
of which 48 patients completed at least six cycles of induction with IRd; 41 patients
(median age 66 years, 66% male) with at least a minimal response (MR) continued to
2 years of rituximab maintenance (1400 mg s.c., q 3 months) starting 3 months after
the last induction cycle. The primary endpoint of the study was overall response rate
(ORR, ≥ MR)) after 8 induction cycles and was 71% (Kersten/Amaador et al, JCO, 2022).
Secondary endpoints included progression-free survival (PFS), overall survival (OS)
and ORR after 2 years of rituximab maintenance and improvement of response after maintenance.
Results: In total, 22 (54%) out of 41 patients completed 2 years of rituximab maintenance.
The median number of cycles was 8 (range, 1-8). Nineteen patients did not complete
maintenance treatment due to progression (n=16), excessive toxicity (n=1), non-compliance
(n=1), and other unknown reason (n=1). The best ORR after maintenance was 85% (2%
complete response [CR], 24% very good partial response [VGPR], 39% partial response
[PR], and 20% MR). Improvement of response after maintenance occurred in 9 (22%) patients;
VGPR to CR in one (2%), PR to VGPR in 6 (15%), and MR to PR in 2 (5%). A further decrease
in IgM levels was seen after 2 years of maintenance (IgM 3.62 g/dl at baseline; 1.3 g/dl
after induction and 0.42 g/dl after end of maintenance, p-value<0.001; Figure 1A),
accompanied by a further increase in Hb levels (Hb 10.5 to 14.3 g/dl, p-value<0.001;
Figure 1B). The median progression-free survival (PFS) was 23.6 months (95% CI, 13.4
to 43.2; Figure 1C), and median overall survival (OS) was not reached; at 45 months,
85% of patients were alive (95% CI, 0.72 to 0.92; Figure 1D). The median time to best
response was 7 months, the median duration of response (DOR) was 34.8 months, and
the median time to next treatment (TTNT) was 36 months[MM(1]. After a median FU of
45.6 months, 29 patients had received subsequent therapy and the median time to progression
after subsequent therapy was 53.7 months[MM(2]. During maintenance, rituximab was
administered i.v. instead of s.c. in 7 (17%) patients. None of the patients treated
with s.c. rituximab developed systemic hypersensitivity reactions but 2 patients had
an injection site reaction (1 grade 1, 1 grade 2). Grade 3/4 toxicity was seen in
10% and 5% of patients, respectively (grade 3/4 neutropenia (n=1 and n=2), grade 3
chronic kidney disease (n=1), and grade 3 elevations of transaminases (n=2)). In the
41 patients who started maintenance, 8 SAEs were reported in 6 patients, of which
4 occurred during maintenance (mostly infections) and 4 (mostly secondary malignancies,
considered unrelated to study drug) during FU. Three patients died during maintenance
therapy due to progressive disease, intracranial bleeding, and acute myeloid leukemia,
respectively.
Image:
Summary/Conclusion: Rituximab maintenance after IRd induction is feasible and well
tolerated. The ORR improved in 22% of patients, confirming the efficacy of this chemo-free
regimen in combination with rituximab maintenance in relapsed/refractory WM.
P1106: PRELIMINARY RESULTS OF A PHASE 1 STUDY OF NOVEL BCL-2 INHIBITOR LISAFTOCLAX
(APG-2575) IN CHINESE PATIENTS (PTS) WITH RELAPSED OR REFRACTORY (R/R) NON-HODGKIN
LYMPHOMAS (NHLS)
M. Sun1,*, J. Qi1, Y. Song2, K. Zhou2, H. Liu3, A. Shen3, L. Geng3, F. Zhou4, J. Huang4,
Z. Chen5, H. Zhang5, M. Lu6, M. Ahmad6, L. Men5, D. Yang7, Y. Zhai6, J. Wang1
1State Key Laboratory of Experimental Hematology, National Clinical Research Center
for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy
of Medical Sciences and Peking Union Medical College, Tianjin; 2Department of Hematology,
Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou; 3First Affiliated Hospital
of University of Science and Technology of China (USTC) Anhui Provincial Hospital,
Hefei; 4Zhongnan Hospital, Wuhan University, Wuhan; 5Ascentage Pharma (Suzhou) Co.,
Ltd., Suzhou, China; 6Ascentage Pharma Group Inc., Rockville, United States of America;
7State Key Laboratory of Oncology in South China Collaborative Innovation Center for
Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
Background: Many B-cell malignancies evade apoptosis by overexpressing BCL-2 proteins.
Although indicated for the management of certain hematological malignances (HMs),
BCL-2 inhibitor (BCL-2i) venetoclax requires a slow dose ramp-up to reduce the risk
of tumor lysis syndrome (TLS) and is associated with severe neutropenia. Investigational
lisaftoclax is a novel, potent, selective BCL-2i, which was previously reported to
have activity against HMs in US and Australian pts. Lisaftoclax also offers a daily
(rather than weekly) ramp-up to the target dose.
Aims: The objective of this study is to evaluate the safety, tolerability, pharmacokinetics
(PK), pharmacodynamics (PD), and preliminary efficacy of lisaftoclax in Chinese pts
with R/R NHLs.
Methods: In this study, pts were dosed with lisaftoclax orally once daily in 28-day
cycles. A daily ramp-up schedule was used in pts with chronic lymphocyte leukemia
(CLL) or NHL with medium/high-risk TLS. A regular schedule was used in pts without
CLL and those with NHL with low-risk TLS.
Results: As of January 1, 2022, 40 pts (median age 55.5 [range 30-79] years; 72.5%
male) had been treated with lisaftoclax (dose range 20-800 mg), including 20 who remain
in the trial. The median number of prior therapies was 4 (range 0-14). NHL subtypes
included: 12 CLL, 9 follicular, 7 mantle cell (MCL), 4 marginal zone (MZL), 4 T-cell,
2 diffuse large B-cell, and 2 Waldenström macroglobulinemia. DLT, MTD, and laboratory/clinical
TLS were not observed, and the RP2D was 600 mg. Treatment-emergent adverse events
(TEAEs) were reported in 36 pts (90%), of which most were grade 1-2 (67.5%). Common
TEAEs (≥ 15% all grades) were hyperuricemia (38.6%), hypertriglyceridemia and anemia
(each 36.4%), thrombocytopenia (29.5%), hyperphosphatemia (27.3%), neutropenia (25%),
proteinuria (22.7%), leukopenia (18.2%), and diarrhea (15.9%). Common grade 3-4 TEAEs
(> 5%) were neutropenia (13.6%) and thrombocytopenia (9.1%). A total of 4 (10%) pts
experienced serious AEs, including 1 small intestinal obstruction, 1 pneumonia, 1
wound hemorrhage, and 1 case each of anemia and thrombocytopenia. One pt had dose
reduction because of grade 2 colitis, and 1 pt receiving 100 mg discontinued because
of grade 4 thrombocytopenia. With a medium treatment duration of 4 (range 4-20) cycles,
12 of 32 (37.5%) evaluable pts reached a complete response (CR; n = 4) or partial
response (PR; n = 8), including 7 with CLL, 2 MCL, 2 MZL, and 1 T-cell NHL. In pts
with CLL, the overall response rate (ORR) was 63.6%, CR 27.3%, and PR rate 36.4%.
At doses ≥ 200 mg, the ORR was 87.5%. A total of 8 of 11 pts had at least 1 adverse
prognostic factor (TP53 mutation/17p-/IGHV unmutated/complex chromosomal abnormalities).
Of 11 pts, 5 had disease that previously failed BTKi or CD20 monoclonal antibody therapy.
Systemic exposure increased across lisaftoclax doses from 20 to 800 mg, with an average
half-life of 4 to 6 hours. There was no significant accumulation following the once-daily
dosing.
Summary/Conclusion: Here, we report for the first time clinical data for lisaftoclax
in Chinese pts with R/R NHLs. Lisaftoclax was well tolerated at doses of up to 800 mg/day,
with no TLS observed (despite the daily ramp-up), and was active against CLL/SLL,
MZL, MCL, and T-cell NHL subtypes. Lisaftoclax may offer a more convenient treatment
alternative, with a daily ramp-up schedule that may be more pt friendly. ClinicalTrials.gov:
NCT03913949.
P1107: CLINICAL OUTCOMES OF SOLID ORGAN TRANSPLANT PATIENTS WITH EBV+ PTLD WHO FAIL
RITUXIMAB PLUS CHEMOTHERAPY: A MULTINATIONAL, RETROSPECTIVE CHART REVIEW STUDY
V. Dharnidharka1, D. Thirumalai2, U. Jaeger3,*, W. Zhao2, D. Dierickx4, P. Xun2, P.
Minga5, A. Sawas6, N. Sadetsky7, P. Chauvet8, E. Sundaram9, A. Barlev7, H. Zimmerman10,
R. U. Trappe10
1Washington University School of Medicine & St. Louis Children’s Hospital, St Louis;
2Atara Biotherapeutics, Thousand Oaks, United States of America; 3Medical University
of Vienna, Vienna, Austria; 4Universitair Ziekenhuis Leuven, Leuven, Belgium; 5Niguarda
Ca’ Granda Hospital, Milan, Italy; 6Herbert Irving Comprehensive Cancer Center, New
York; 7Atara Biotherapeutics, South San Francisco, United States of America; 8CHU
Lille, Lille, France; 9Nashville Bioscences, Nashville, United States of America;
10DIAKO Bremen, Bremen, Germany
Background: Post-transplant lymphoproliferative disease (PTLD) is a rare and often
aggressive disease i n the setting of immunosuppression following solid organ transplant
(SOT). Epstein-Barr virus (EBV) infection of B cells is responsible for about 50%
of cases, either due to reactivation of the virus after transplantation or primary
EBV infection. Although there is no approved therapy for patients (pts) with PTLD,
guidelines include reduction of immunosuppression (RIS) as a part of initial treatment
and may be sufficient for pts with early lesions. Rituximab, either as monotherapy
or in combination with chemotherapy (CT), is used in addition to RIS as initial treatment.
Pts with EBV+ PTLD following SOT who fail rituximab plus CT have poor outcomes with
limited treatment options. Published data on clinical outcomes of these pts remain
limited and not well documented.
Aims: To characterize the outcomes for pts diagnosed with EBV+ PTLD following SOT
who fail initial rituximab plus CT in a multi- national real-world setting.
Methods: We conducted a large multinational, multicenter, retrospective chart review
study of pts with EBV+ PTLD following allogeneic hematopoietic cell transplantation
(HCT) or SOT who received rituximab or rituximab plus CT between January 2000‒December
2018 and were refractory (failed to achieve complete response [CR] or partial response
[PR]) or relapsed at any point after such therapy. Data were collected from 29 centers
across North America (United States and Canada) and the European Union. This analysis
includes pts with EBV+ PTLD following SOT who were refractory/relapsed to rituximab
plus CT. The Kaplan-Meier method was utilized to estimate the overall survival (OS).
Results:: A total of 86 pts with EBV+ PTLD following SOT who failed rituximab plus
CT were included in the analysis; 65 (75.6%) pts were refractory while 21 (24.4%)
relapsed after initial response of CR or PR. Median age at PTLD diagnosis was 43 years
(range 1‒78) and median time to PTLD onset from transplant was 1.7 years (range 0.1‒27.9).
Median follow up time was 12.9 months from the date of PTLD diagnosis. PTLD histological
subtypes were 66 (76.7%) monomorphic, 18 (20.9%) polymorphic, and 2 (2.3%) early lesions.
The most common PTLD subtype was diffuse large B-cell lymphoma (DLBCL) (58, 67.4%).
Of the 86 pts, 49 (57%) received CT following rituximab monotherapy while 37 (43%)
pts received CT concurrently with rituximab. Overall, 63 (73.3%) pts died. PTLD-specific
mortality was observed in 41 (65.1%) pts, treatment-related mortality in 10 (15.9%)
pts, mortality due to organ rejection/failure in 2 (3.2%) pts, mortality due to other
causes in 7 (11.1%) pts, and mortality due to unknown causes in 3 (4.8%) pts. Median
OS was 15.5 months (95% confidence interval [CI]: 8.3‒22.9) from PTLD diagnosis, and
was 4.1 months (95%CI: 1.9‒8.5) from the earliest date when pts became refractory
or relapsed following rituximab plus CT.
Image:
Summary/Conclusion: The prognosis for pts with EBV+ PTLD following SOT who fail rituximab
plus CT remains poor, with an estimated median OS of about 4 months and a majority
of pts dying from PTLD and related treatment. In this specific population, there remains
a significant unmet medical need for effective and well-tolerated therapies.
P1108: HIGH COMPLETE RESPONSE RATE FOLLOWING POINT-OF-CARE ANTI CD19 CAR T-CELL THERAPY
IN PATIENTS WITH RELAPSED/REFRACTORY FOLLICULAR LYMPHOMA
S. Fried1,2,*, M. J. Besser3,4, E. Shkury1,2, R. Yerushalmi1,2, N. Shem-Tov1,2, I.
Danylesko1,2, E. Jacoby2,5, O. Itzhaki4, R. Shouval1,2,6, M. Kedmi1,2,7, A. Shimoni1,2,
A. Nagler1,2, A. Avigdor1,2
1Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center,
Tel Hashomer; 2Sackler School of Medicine; 3Department of Clinical Microbiology and
Immunology, Sackler School of Medicine, Tel Aviv University, Tel-Aviv; 4Ella Lemelbaum
Institute for Immuno Oncology; 5Department of Pediatric Hematology-Oncology, Safra
Children’s Hospital, Chaim Sheba Medical Center, Tel Hashomer, Israel; 6Adult BMT
Service, Memorial Sloan Kettering Cancer Center, New York, United States of America;
7The Mina and Everard Goodman faculty of life sciences, Bar Ilan University, Ramat
Gan, Israel
Background: Patients with relapsed/refractory follicular lymphoma (R/R FL) often experience
multiple relapses and require various lines of therapy. The ELARA and ZUMA-5 trials
demonstrated high response rates along with acceptable safety profiles. We perform
a phase 1b/2 single-center clinical trial of autologous point-of-care (POC) academic
anti-CD19 chimeric antigen receptor (CAR) T-cells for patients with R/R FL treated
with at least 2 lines of systemic therapy (NCT02772198).
Aims: To report outcomes of POC CAR T-cell therapy in patients with R/R FL.
Methods: Adults with R/R FL underwent a single leukapheresis procedure. Fresh peripheral
blood mononuclear cells were isolated, activated, and transduced with a gammaretrovirus
encoding for a CD19 CAR (based on an FMC63-derived ScFv, a CD28 costimulatory domain,
and a CD3-ζ signaling domain). Lymphodepletion included fludarabine 25 mg/m2 over
3 days (days −4 to −2) and cyclophosphamide 900 mg/m2 once (day −2), followed by infusion
of 1×106/kg CAR T-cells in the inpatient setting. Primary endpoints were response
(by PET-CT, per Lugano criteria) at day 28, best response, and safety. Secondary endpoints
included overall survival, progression-free survival (PFS), and production feasibility.
Last follow-up was as of 02/2022.
Results: All 19 patients enrolled received CAR T-cell infusion in a median of 11 days
(IQR 10-11) after leukapheresis. The median age was 61 years (IQR 52-66). Five (26%)
patients had Karnofsky performance status < 90%. Disease stage at enrollment was III-IV
in 16 (84%) patients. Two (11%) patients had bulky disease; 8 (42%) had LDH > upper
limit of normal; and 16 (84%) had Follicular Lymphoma International Prognostic Index
≥ 3. Disease status at enrollment was progressive disease (n=14, 74%), stable disease
(n=3, 16%), or partial response (PR; n=2, 11%). Twelve patients (64%) were refractory
to last treatment. Disease grade at most recent lymph node biopsy was 1 (n=3, 16%),
2 (n=11, 58%), or 3a (n=5, 26%). The median time from FL diagnosis was 3.9 years (IQR
2.5-4.6). Sixteen (84%) patients had progression of disease within 24 months of initial
therapy. The number of prior therapies was ≥ 4 in 6 (32%) patients; and 5 (26%) patients
underwent prior autologous transplantation.
Grade III-IV cytokine release and immune effector cell-associated neurotoxicity syndromes
occurred in 1 (5%) and 4 (21%) patients, respectively. One patient was infected with
COVID-19 on the 5th day following cell infusion and was admitted to the intensive
care unit. One patient had grade 3 atrial fibrillation. Severe neutropenia (absolute
neutrophil count <500/µL), thrombocytopenia (platelets <50K/µL) and anemia (hemoglobin
<10g/dl) occurred in 15 (79%), 5 (26%), and 7 (37%) patients, respectively. No bleeding
events or death were recorded following cell infusion.
Response was evaluated in all patients. Overall response rate on day 28 was 84% (79%
complete response [CR]). One patient with PR on day 28 achieved a CR after a year
of follow-up. Three patients (16%) continued to progress following CAR infusion.
All patients were alive at the last follow-up (median follow-up, 11.5 months [IQR
4-21]). One-year PFS was 74% (95% CI, 53-100). The median duration of response (DOR)
was not reached (95% CI, 12.5-not reached). Estimated DOR at 1-year was 89% (95% CI,
71-100).
Image:
Summary/Conclusion: Point-of-Care anti-CD19 CAR T-cell therapy, performed following
a very short production time, induced high CR rate with an acceptable safety profile
in a cohort of patients with high-risk R/R FL.
P1109: TRIAL IN PROGRESS: PHASE 1B/2 TRIAL OF TAZEMETOSTAT IN COMBINATION WITH VARIOUS
TREATMENTS IN PATIENTS WITH RELAPSED OR REFRACTORY HEMATOLOGIC MALIGNANCIES
A. Bessudo1,*, D. Greenwald2, M. H. Kazemi3, A. Mahindra4, L. Shunyakov5, M. Sondhi6,
H. O’Connor6, Y. Chen6, J. Yang6, G. Salles7
1cCare, Encinitas; 2UCLA, Santa Barbara; 3Astera Cancer Care, East Brunswick; 4Scripps
MD Anderson Cancer Center, La Jolla; 5Central Care Cancer Center, Bolivar; 6Epizyme,
Inc., Cambridge; 7Memorial Sloan Kettering Cancer Center, New York, United States
of America
Background: Enhancer of zeste homolog 2 (EZH2), an epigenetic regulator, suppresses
key cellular checkpoints and differentiation pathways to maintain the oncogenic characteristics
of germinal center B cells. In addition to activity in diffuse large B-cell lymphoma
(DLBCL) and mantle cell lymphoma (MCL), EZH2 inhibitors induced cell cycle arrest
and apoptosis in preclinical studies of multiple myeloma (MM). Tazemetostat (TAZ),
an oral selective EZH2 inhibitor, is approved for treatment of relapsed or refractory
(R/R) follicular lymphoma. Preclinical studies demonstrated antiproliferative and
synergistic effects of TAZ + lenalidomide (LEN), a Bruton tyrosine kinase inhibitor
(BTKi), and daratumumab, pomalidomide, and dexamethasone (mAbPD) in DBLCL, MCL, and
MM cell lines, respectively. LEN is effective for DLBCL and is approved in combination
with tafasitamab-cxix, a CD19-directed cytolytic antibody (CD19 antibody [Ab]), for
R/R DLBCL. Acalabrutinib, a BTKi, is approved in MCL, and mAbPD showed clinical activity
in R/R MM.
Aims: The aim of this study is to examine the efficacy and safety of TAZ + CD19 Ab,
LEN, BTKi, or mAbPD in patients (pts) with R/R hematologic malignancies.
Methods: This trial is a 2-part, multicenter, open-label, safety, efficacy, signal-finding,
multi-arm phase 1b/2 study of TAZ plus various treatment regimens for hematologic
malignancies (NCT05205252). Phase 1b is a dose escalation safety run-in; phase 2 is
a dose expansion study. Currently, there are 4 treatment arms (Table). Written informed
consent will be obtained upon enrollment. Eligible pts are adults (aged ≥18 y) with
an ECOG performance status 0–1 (phase 1b) or 0–2 (phase 2) and measurable disease
by Lugano classification for arms 1–3 or IMWG 2016 criteria for arm 4. Pts with severe
concurrent disease are excluded from this study. During the phase 1b safety run-in
portion of the study, pts will be enrolled in 3 dose escalation cohorts of TAZ 400 mg,
600 mg, and 800 mg. Pts in all novel treatment combinations (arms 1, 3, 4) will start
at the lowest TAZ dose of 400 mg, and treatment combinations previously studied (arm
2) will start at TAZ 800 mg. All pts will receive TAZ orally twice daily in continuous
cycles. All combination partners will be administered per their respective package
insert or investigator brochure. The phase 1b portion endpoint summarizes treatment-emergent
dose-limiting toxicities and adverse events to inform the recommended phase 2 dose
for each arm. The primary endpoint for all arms in phase 2 is objective response rate;
key secondary endpoints include progression-free survival, duration of response, overall
survival, and safety.
Results: As of February 9, 2022, no pts are enrolled at 1 active site; additional
sites are in startup.
Image:
Summary/Conclusion: This phase 1b/2 study will provide insights into the efficacy
and safety of TAZ + CD19 Ab, LEN, BTKi, or mAbPD in pts with R/R hematologic malignancies.
P1110: EFFICACY AND SAFETY OF IMMUNOCHEMOTHERAPY IN TREATMENT OF FOLLICULAR NON-HODGKIN’S
LYMPHOMA DURING COVID-19 PANDEMIC: A STUDY OF KROHEM, THE CROATIAN COOPERATIVE GROUP
FOR HEMATOLOGIC DISEASES
D. Galusic1,2, S. Basic - Kinda3, A. Pijuk1, V. Milunovic4, B. Dreta3,*, N. Franjic5,
B. Coha6, J. Sincic – Petricevic7, P. Gacina8, V. Pejsa9, M. Lucijanic9, I. Aurer3,10
1Division of Hematology, Department of Internal Medicine, University Hospital of Split;
2School of Medicine, University of Split, Split; 3Division of Hematology, Department
of Internal Medicine, University Hospital Centre Zagreb; 4Division of Hematology,
Department of Internal Medicine, University Hospital Merkur, Zagreb; 5Division of
Hematology, Department of Internal Medicine, University Hospital Centre Rijeka, Rijeka;
6Department of Internal Medicine, Dr. “Josip Bencevic” General Hospital, Slavonski
Brod; 7Division of Hematology, Department of Internal Medicine, University Hospital
Centre Osijek, Osijek; 8Division of Hematology, Department of Internal Medicine, Sestre
Milosrdnice University Hospital Center; 9Division of Hematology, Department of Internal
Medicine, University Hospital Dubrava; 10School of Medicine, University of Zagreb,
Zagreb, Croatia
Background: Follicular lymphoma (FL) is a systemic neoplasm of the lymphoid tissue
arising from B cell proliferation. The novel monoclonal anti-CD20 antibody obinutuzumab
in combination with chemotherapy has been widely accepted as the first choice in front
line treatment of FL. Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2),
responsible for coronavirus disease 2019 (COVID-19) is causing increased mortality
among patients with lymphoproliferative disorders compared with the general population.
Furthermore, there are some concerns in terms of morbidity and mortality for patients
with FL because of their immunocompromised status induced by recent exposure to cytotoxic
chemotherapy, especially bendamustine and anti-CD20.
Aims: To investigate efficacy and safety of immunochemotherapy protocols for patients
with newly diagnosed FL during COVID-19 pandemic.
Methods: We retrospectively investigated medical data of all patients with newly diagnosed
FL grade 1, 2 or 3A from Croatian hematologic registry in period from April 2019 to
March 2021. Only patients which required systemic treatment were included in the analysis.
All patients received obinutuzumab (G) in combination with either CHOP, bendamustine
(B) or CVP chemotherapy protocol. Treatment response was evaluated using international
lymphoma response criteria.
Results: We analyzed a total of 114 FL patients treated with G-chemotherapy. Mean
age was 62.4 ±10.5 years. Majority of patients were female (71/114 (62.3%)). FL grade
I was present in 45/114 (39.5%), grade II in 28/114 (24.6%), grade III in 27/114 (23.7%)
and not specified (but not IIIB) in 14/114 (12.3%) patients. A total of 61/114 (53.5%)
patients were treated with G-B, 49/114 (43%) with G-CHOP and 4/114 (3.5%) with G-CVP
immunochemotherapy. Similar rates of adverse events were observed in patients treated
with G-CHOP and G-B Median follow up was 17 months. Overall response rate was 94%,
complete remission (CR) in 68% and partial remission (PR) in 25% of patients. Median
overall survival (OS) and progression free survival (PFS) were not reached with 12-months
rates of 94% and 92%, respectively. Patients treated with G-CHOP had statistically
significantly superior OS and PFS compared to patients treated with G-B (P=0.002 and
P=0.006, respectively, Fig. 1). More favorable survival course associated with G-CHOP
in comparison to G-B persisted in multivariate analysis (P=0,026, HR=15,12) after
adjustment for age, sex, FLIPI grade and SARS-CoV-2 infection. Total of 12 patients
died during the follow up and COVID-19 was cause of death in 5 patients. During the
follow-up SARS-CoV-2 infection was diagnosed in 20/114 (17,5%) patients with overall
mortality rate of 25%. All of the 7 patients treated with G-CHOP recovered from SARS-CoV-2
infection and mortality rate in infected group of patients treated with G-B was 33%
(4/12 patients).
Image:
Summary/Conclusion: Increased COVID-19 mortality in patients with lymphoproliferative
disorders was observed in this study. Our group of patients had reduced OS and PFS
compared to the GALLIUM trial and SARS‐CoV‐2 infection was the most pronounced risk
factor for death. Even though in some studies bendamustine has shown to be less toxic
and more effective than CHOP in FL, there are some important pandemic aspects that
must be considered. Bendamustine exposure seems to be associated with worse outcome
in case of the infection with SARS-CoV-2. These intriguing differences could play
important role in treatment approach in COVID-19 pandemic. Future studies investigating
hematological malignancies in COVID-19 pandemic are warranted.
P1111: COMPARATIVE EFFICACY AND SAFETY OF TISAGENLECLEUCEL AND AXICABTAGENE CILOLEUCEL
IN RELAPSED/REFRACTORY FOLLICULAR LYMPHOMA
M. Dickinson1,*, J. Martinez-Lopez2, E. Jousseaume3, C. Anjos4, H. Yang5, X. Chai5,
T. Wang5, R. Ramos4, C. Lobetti-Bodoni3, S. Schuster6, N. Fowler7
1Peter MacCallum Cancer Centre, Royal Melbourne Hospital and the University of Melbourne,
Melbourne, VIC, Australia; 2Hospital Universitario 12 de Octubre, Complutense University,
CNIO, Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain; 3Novartis
Pharmaceuticals AG, Basel, Switzerland; 4Novartis Pharmaceuticals Corporation, East
Hanover, NJ; 5Analysis Group Inc., Boston, MA; 6Lymphoma Program, Abramson Cancer
Center, University of Pennsylvania, Philadelphia, PA; 7The University of Texas MD
Anderson Cancer Center, Houston, TX, United States of America
Background: The chimeric antigen receptor T-cell (CAR-T) therapies, tisagenlecleucel
(tisa-cel) and axicabtagene ciloleucel (axi-cel) have demonstrated clinical benefits
in treating relapsed/refractory follicular lymphoma (r/r FL).
Aims: To compare the efficacy and safety outcomes of tisa-cel and axi-cel in r/r FL
using matching-adjusted indirect comparison (MAIC).
Methods: Individual patient-level data (IPD) in ELARA (tisa-cel; NCT03568461; 03/2021
data-cut; N=97 [infused set]; N=90 [efficacy-evaluable set]) were weighted to match
the patient population in ZUMA-5 (axi-cel; NCT03105336; 03/2020 data-cut; N=124 [infused
set]; N=84 [efficacy-evaluable set]). Because patients from ZUMA-5 did not use bridging
chemotherapy, the primary analysis compared a patient subgroup not exposed to bridging
chemotherapy in ELARA (N=53 [infused set]; N=50 [efficacy-evaluable set]) with patients
in ZUMA-5. Baseline characteristics available in both trials were adjusted in MAIC
including age, sex, ECOG performance status, Ann Arbor stage, FLIPI, tumor burden
per GELF criteria, number of prior therapy lines, refractory status to the most recent
regimen, progression of disease within 24 months from first anti-CD20 monoclonal antibody-containing
therapy, and prior autologous stem cell transplant. Efficacy outcomes, including overall
response rate (ORR), complete response (CR) rate, and progression-free survival (PFS),
were compared among the efficacy-evaluable set, defined as patients with ≥12 months
of follow-up in both trials. Selected safety outcomes, including cytokine release
syndrome (CRS), neurological events (NE), and tocilizumab and corticosteroid use for
CRS, were compared between the infused populations. A sensitivity analysis was conducted
to compare patients with and without bridging chemotherapy in ELARA with ZUMA-5.
Results: After adjusting for differences in baseline characteristics, ORR, CR rate,
and PFS were comparable between tisa-cel and axi-cel efficacy-evaluable patients (Figure
1A). In the primary analysis, the hazard ratio for PFS post-weighting was 0.90 (95%
confidence interval: 0.39, 2.06; p=0.81) (Figure 1B). The adverse event rates for
CRS and NE of any grade were 32.70% (p<0.01) and 47.70% (p<0.001) lower, respectively,
in patients infused with tisa-cel compared to those infused with axi-cel. The proportion
of patients who used tocilizumab and corticosteroids for CRS were 35.57% (p<0.001)
and 12.69% (p<0.01) lower, respectively, in patients infused with tisa-cel (Figure
1C). Similar results were observed in the sensitivity analysis among patients with
and without bridging chemotherapy in ELARA (Figure 1A-C).
Image:
Summary/Conclusion: The MAIC results indicated that tisa-cel and axi-cel were comparable
in response rates and PFS, while tisa-cel was associated with better safety outcomes
than axi-cel. These results confirm and reinforce the relevance of CAR-T therapies
for r/r FL. Future analyses using IPD from both trials and real-world data are warranted.
P1112: TREATING BING-NEEL SYNDROME USING A TAILORED APPROACH: EXPERIENCE OF A SPECIALIST
NEUROHAEMATOLOGY CLINIC
J. Khwaja1,*, D. Smyth2, A. Rismani1, C. Hoskote3, C. Kyriakou1, M. P. Lunn2, S. D’Sa1
1Department of Haematology, University College London Hospital; 2Centre for Neuromuscular
Disease; 3Department of Neuroradiology, National Hospital for Neurology and Neurosurgery,
London, United Kingdom
Background: Bing-Neel syndrome (BNS) is a rare complication of lymphoplasmacytic lymphoma
(LPL) comprising LPL infiltration in the central nervous system (CNS). Clinical and
radiological features are diverse; the diagnosis is confirmed by cerebrospinal fluid
(CSF) analysis using immunological and molecular techniques. Rarely, a tissue biopsy
is required. The pattern of presentation including systemic involvement and CSF features
inform treatment strategies, which include CNS-penetrating therapies.
Aims: To evaluate the diagnostic characteristics of patients with BNS and their influence
on therapy.
Methods: Data from patients referred between 2011-2021 for management of BNS to our
academic neurohaematology centre were retrospectively reviewed. Those with imaging
features alone or where it was not possible to distinguish from high-grade transformation
were excluded.
Results: Thirty-five patients (22 male, 13 female) were identified. Median age at
diagnosis of BNS was 65 years (range 48-85). All patients were symptomatic. In 12
patients (34%) BNS was the de novo presentation of the IgM-related disorder, of which
3 (25%) had no detectable bone marrow (BM) infiltration of LPL at diagnosis. Approximately
half (17; 49%) had previously received therapy for LPL; median time to BNS diagnosis
in these was 49 months (range 3-125). At BNS diagnosis, BM involvement with LPL ranged
from 0-95%. More than half (14/26; 54%) had ≤10% infiltrate and almost a fifth (4/26)
>60%.
All patients had leptomeningeal involvement and 8 (23%) additionally had parenchymal
CNS disease. The majority had kappa light-chain predominance: IgMκ (n=26), non-IgMκ
(n=5), IgMλ (n=3), one unknown. The BNS diagnosis was made on CSF analysis (n=28;
80%), leptomeningeal tissue biopsy (n=3; 9%) where CSF was non-informative, or by
expert opinion based on supportive clinical, radiological and non-definitive CSF features
(n=4; 11%). Of those with a diagnosis based on CSF studies, B-cell clonality was confirmed
by flow cytometry (27/28; 96%), MYD88L265P mutation (18/28; 64%) and immunoglobulin
gene rearrangement (12/28; 43%). In 22 samples with a full dataset, median CSF white
cell count was 25/ul (1-233), CSF protein 1.69g/l (0.35-6), CSF IgM 9.49mg/l (1.07-61.5).
The majority were treated with intensive regimens (rituximab, methotrexate (MTX),
cytarabine (ARA-C) ± thiotepa/idarubicin; n=30) due to the presence of CNS disease
bulk and clinical need, and less commonly ibrutinib (n=3), bendamustine-rituximab
(BR, n=1); one patient had intrathecal therapy (MTX, ARA-C) at the height of the COVID
pandemic. Of those who received 2 cycles of intensive chemotherapy, 3 had ≥4 cycles
followed by BCNU/thiotepa autologous stem cell transplant; 10 proceeded to ‘consolidation’
(indefinite) ibrutinib to limit intensive chemotherapy or tackle systemic disease.
At a median follow up of 26 months (range 1-121), median survival was not reached;
2-year overall survival was 91% (95% CI 74-97). Three patients died during treatment
(1 invasive fungal infection post COVID-19 during ibrutinib consolidation post MTX/ARA-C
based therapy) and 2 during MTX-ARA-C based therapy; 7 patients relapsed or progressed
and were treated with ibrutinib: 1 relapsed after ibrutinib use, 1 patient was intolerant
of ibrutinib and switched to BR.
Image:
Summary/Conclusion: Our cohort confirms that BNS may present with leptomeningeal disease
and/or parenchymal disease, de novo and without systemic disease. Overall outcomes
are excellent with intensive regimens, consolidated with or followed by ibrutinib;
however, there are treatment-related toxicities emphasising the need for a tailored
approach.
P1113: PROGNOSTIC ROLE OF GENETIC ABNORMALITIES IN MANTLE CELL LYMPHOMA
E. Kleina1,*, S. Voloshin1, J. Vokueva1, O. Petukhova1, L. Martynenko1, M. Bakai1,
J. Ruzhenkova1, S. Linnikov1, E. Kariagina2, O. Uspenskaia3, I. Ziuzgin4, S. Bessmelcev1,
S. Sidorkevich1, I. Martynkevich1
1Russian Research Institute of Hematology and Transfusiology, Federal Medical and
Biological Agency; 2St. Petersburg GBUZ “City Hospital No. 15; 3GBUZ Leningrad Regional
Clinical Hospital; 4”The N.N. Petrov National Medicine Research Institute of oncology”
Ministry of Health of Russia, Saint-Petersburg, Russia
Background: Mantle cell lymphoma (MCL) – B-cell lymphoma with a high frequency of
genome instability associated with a recurrent clinical course, short non-progressive
and overall survival. Special attention is paid to the “double-hit” MCL (simultaneous
presence of translocations of CCND1 and C-MYC genes), characterized mainly by a pleomorphic/blastoid
morphological variant, refractory clinical course and unfavorable prognosis.
Aims: To identify the spectrum and frequency of occurrence of genetic anomalies and
their effect on the course of MCL.
Methods: The results of a standard cytogenetic study (G-banding) in 51 and FISH studies
in 75 patients with MCL are presented.
Results: In a standard cytogenetic study, a pathological karyotype was found in 25/51
(49,0%) patients. Translocation t(11;14)(q13;q32) was detected in 23/51 (45,1%) patients.
As part of the complex karyotype, t(11;14) was determined in 16/51 (31,4%) cases.
The FISH study made it possible to additionally identify genetic aberrations in patients
with MCL. In patients with normal karyotype t(11;14) was found in 18/51 (35,3%) cases.
C-MYC gene rearrangements were detected in 16/75 (21,3%) cases. In this group, genetic
abnormalities were found in 7/11 (63,6%) patients (in 5 patients, mitoses were not
obtained). In all patients t(11;14) was detected either as a single genetic aberration
(in 1/7 (14,3%)) or as part of a complex karyotype (in 6/7 (85,7%)) in combination
with genetic abnormalities 7 and 17 chromosomes. In 8/16 (50,0%) patients, C-MYC gene
rearrangements were combined with a deletion of the TP53 gene. In 59/75 (78,7%) cases,
C-MYC gene rearrangements were not detected. Chromosomal aberrations were found in
18/39 (46,2%) patients, including complex karyotype changes in 10/18 (55,6%) patients.
Translocation t(11;14) - in 16/18 (88,9%) cases. TP53 gene deletion was detected in
10/59 (16,9%) patients. Data analysis showed a significant adverse effect of the complex
karyotype, TP53 deletion, C-MYC rearrangement on the clinical course of MCL compared
with the standard risk group. The median progression-free survival in high molecular
risk patients was 12,5 months; the median progression-free survival was not achieved
in the standard risk group.
Summary/Conclusion: An integrated approach to the genetic diagnosis of MCL using standard
cytogenetic and FISH studies makes it possible to identify a high-risk group: “double-hit”
and with complex changes in the karyotype. It was also shown that in the group of
patients with changes in the C-MYC gene, additional genetic anomalies were more often
detected, which are an unfavorable prognostic factor: a complex karyotype (the presence
of three or more aberrations) – 37,5% and 16,9%, and aberrations involving the gene
TP53 – 50,0% and 16,9%. Changes in these genes are associated with shorter progression-free
survival rates.
P1114: ZANDELISIB ON INTERMITTENT DOSING AS A SINGLE AGENT OR IN COMBINATION WITH
RITUXIMAB OR ZANUBRUTINIB IN RELAPSED/REFRACTORY (R/R) FOLLICULAR LYMPHOMA (FL): RESULTS
FROM A MULTI-ARM PHASE 1B STUDY
J. D. Soumerai1,*, D. Jagadeesh2, H. Salman3, F. Samaniego4, K. Patel5, A. Stathis6,
N. Reddy7, V. P. Kenkre8, A. Asch9, C. Diefenbach10, I. S. Lossos11, D. Persky12,
F. Awan13, W. Huang14, K. Vandever14, A. D. Zelenetz15
1Massachusetts General Hospital, Boston; 2Cleveland Clinic, Cleveland; 3Stony Brook
University, Stony Brook; 4The University of Texas MD Anderson Cancer Center, Houston;
5Swedish Cancer Institute, Seattle, United States of America; 6Oncology Institute
of Southern Switzerland, Bellinzona, Switzerland; 7Vanderbilt University Cancer Center,
Nashville; 8University of Wisconsin Carbone Cancer Center, Madison; 9Oklahoma University
Health Sciences Center, Oklahoma City; 10Perlmutter Cancer Center at NYU Langone Health,
New York; 11Sylvester Comprehensive Cancer Center, Miami; 12University of Arizona,
Tucson; 13University of Texas Southwestern Medical Center, Dallas; 14MEI Pharma, Inc.,
San Diego; 15Memorial Sloan Kettering Cancer Center, New York, United States of America
Background: PI3Kδ inhibitors administered daily in patients (pts) with FL were limited
by immune-related adverse events (AEs) due to regulatory T cell (Treg) suppression
following continuous on-target inhibition. Zandelisib is a structurally differentiated
oral PI3Kδ inhibitor with high specificity and target-binding affinity. We hypothesized
that zandelisib administered on an intermittent dosing (ID) schedule (one capsule
daily on days 1-7 of a 28-day cycle) would enable Treg repopulation during treatment
breaks and improve safety.
Aims: We conducted this analysis to assess the safety and efficacy of zandelisib administered
by ID as a single agent or in combination therapy in pts with R/R FL enrolled in a
3-arm phase 1b study in various B-cell malignancies.
Methods: Eligible pts ≥18 years with FL Grade I-IIIA, ECOG PS 0-2, progressive disease
after ≥1 prior therapy and no prior PI3Kδ inhibitor, provided consent prior to enrollment
(NCT02914938). A dose escalation stage assessed single-agent zandelisib daily continuously
and is not reported here (Soumerai et al, J Clin Oncol 2018;36:#7519). Subsequent
cohorts evaluated zandelisib 60 mg daily for two 28-day cycles then on ID as a single
agent (Group 1) or in combination with rituximab 375 mg/m2 weekly x4 in cycle 1 and
on Day 1 of cycles 3-6 (Group 2). Group 3 evaluated zandelisib 60 mg on ID from cycle
1 and zanubrutinib 80 mg twice daily. Treatment was continued until disease progression
or intolerance. Imaging scans were obtained after 2 and 6 cycles 6 and then every
6 months. Response was assessed by investigators using the Lugano Criteria.
Results: 69 FL pts were treated: 18 in Group 1, 19 in Group 2, and 32 in Group 3.
Enrollment began in September 2017 for Group 1 and 2 and June 2019 for Group 3, and
is completed in all 3 groups. Median age was 66 years (range 38-87), median prior
therapies was 2 (range 1-5), 40 pts (58%) received ≥2 prior therapies, 45 pts (65%)
were POD24, 21 pts (30.4%) were refractory to last therapy, and 28 pts (41%) had tumors
≥5 cm. With a median follow-up of 10.8 months (range 1.8-49.5), 6 pts (8.7%) have
discontinued therapy due to an AE. Grade 3 AEs of special interest (AESI) were rash
in 6 pts (8.7%), ALT increased in 6 (8.7%), diarrhea in 3 (4.3%), colitis in 2 (2.9%),
and AST increased in 3 (4.3%). No grade 4-5 AESI were reported. 1 pt in Group 3 had
reversible grade 4 drug rash with eosinophilia and systemic symptoms (DRESSS) syndrome.
Grade 3-4 neutropenia was observed in 11 pts (16%). The overall response rate (ORR)
in 65 evaluable pts was 84.6% (55/65), 95% CI 73.5-92.4, and the CR rate was 24.6%
(16/65), 95% CI 14.8-36.9. The ORR was 77.8% (14/18) in Group 1, 94.7% (18/19) in
Group 2, and 82.1% (23/28) in Group 3. The median duration of response (DOR) was 31.1
months in Group 1, 25.8 months in Group 2, and not yet mature in Group 3, with median
drug exposure of 17.1, 20.5, and 7.1 months, respectively (Figure).
Image:
Summary/Conclusion: Zandelisib was generally well tolerated when administered by ID
as a single agent or in combination with rituximab or zanubrutinib, with a low rate
(8.7%) of grade 3 AESI and discontinuations due to AEs. The high ORR and prolonged
DOR are encouraging. This profile supports further evaluation of zandelisib on ID
as a single agent and in combination in various B-cell malignancies, both in R/R disease
and in earlier lines of therapy.
P1115: A MULTICENTER PHASE 1/2 TRIAL OF EO2463, A MICROBIAL-DERIVED PEPTIDE THERAPEUTIC
VACCINE, AS MONOTHERAPY AND IN COMBINATION WITH LENALIDOMIDE AND RITUXIMAB, FOR TREATMENT
OF PATIENTS WITH INDOLENT NHL
P. L. Zinzani1,*, S. M. Ansell2, F. Bosch3, J. W. Friedberg4, J. P. Marolleau5, L.
Arcaini6, R. Garcia-Sanz7, A. K. Gopal8, C. Grande9, R. Merryman10, A. Pinto11, S.
D. Smith12, J. C. Villasboas2, D. Wallace13, J. Fagerberg14, J. G. Magalhaes14, P.
Armand10
1Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Bologna, Italy;
2Division of Hematology, Mayo Clinic, Rochester, United States of America; 3Department
of Hematology, Vall d’Hebron Institute of Oncology, Barcelona, Spain; 4University
of Rochester Medical Center, Rochester, United States of America; 5CHU Amiens, Amiens,
France; 6Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; 7Hospital Universitario
de Salamanca, Salamanca, Spain; 8Division of Medical Oncology, University of Washington,
Seattle, United States of America; 9Universidad de Navarra, Madrid and Pamplona, Spain;
10Dana-Farber Cancer Institute, Boston, United States of America; 11Istituto Nazionale
Tumori “Fondazione G.Pascale”- IRCCS, Naples, Italy; 12University of Washington/Fred
Hutchinson Cancer Research Center, Seattle; 13Department of Medicine, University of
Rochester, Rochester, United States of America; 14Enterome, Paris, France
Background: EO2463 is a therapeutic vaccine designed to activate existing commensal
bacteria-specific memory T cells that cross-react with B cell markers in order to
drive anti-tumor immune activity against B-cell malignancies. EO2463 contains four
microbial-derived, synthetically produced peptides (OMP72, OMP64, OMP65, and OMP66),
which correspond to cytotoxic CD8 T cell HLA-A2 restricted epitopes, and exhibit molecular
mimicry with the B cell markers CD20, CD22, CD37, and CD268 (BAFF-receptor), respectively.
In pre-clinical models, these peptides can generate strong immune responses and specifically
stimulate cross-reactive cytotoxic CD8 T cells to recognize the chosen B cell targets.
EO2463 also contains a CD4 helper peptide referred to as universal cancer peptide
2, derived from the human telomerase reverse transcriptase catalytic subunit. The
present study is a first-in-human clinical trial of this microbiome-derived peptide
therapeutic cancer vaccine approach in patients with follicular lymphoma (FL) and
marginal zone lymphoma (MZL).
Aims:
The primary objectives of the phase 1 part of the trial are to define the recommended
phase 2 dose (RP2D) for EO2463 monotherapy, and to confirm the safety of EO2463 at
the monotherapy RP2D in combination with lenalidomide (EL), rituximab (ER), and lenalidomide/rituximab
(ER2).
The primary objective of the phase 2 part of the trial is to estimate the objective
response rate (ORR) according to the Lugano Classification 2014 during EO2463 monotherapy
Methods: This four-cohort phase 1/2 trial will investigate EO2463 monotherapy, and
combinations of EO2463/lenalidomide (EL), EO2463/rituximab (ER), and EO2463/lenalidomide/rituximab
(ER2), for treatment of patients with FL and MZL. Cohort 1 is a safety lead-in dose-finding
in patients with relapsed/refractory (RR) disease, with a 3-by-3 design to establish
the recommended phase 2 dose (RP2D) for EO2463 monotherapy and to confirm the safety
of the RP2D for combination schedules of EL, and ER2. Maximal eighteen patients will
be included in cohort 1. After the recommended EO2463 monotherapy dose is established,
cohorts 2, 3, and 4 will open to accrual. Fifteen patient will be included in each
of these cohorts. Cohort 2 will investigate EO2463 monotherapy in patients with newly
diagnosed FL/MZL who are not in need of treatment; cohort 3 will investigate EO2463
monotherapy, followed by ER in patients with limited tumor burden who need treatment,
and cohort 4 will further investigate EL, followed by ER2 in the RR setting. EO2463
will be administered subcutaneous 4 times at 2-week intervals, followed by continued
booster administrations every 4 weeks for 9 (Cohorts 2 and 3) or 12 (Cohorts 1 and
4) months. Inclusion/exclusion criteria, and the design and schedule of the intense
immune and safety monitoring will be presented.
Results: The safety lead-in dose-finding is currently ongoing, and no safety concerns
have been observed thus far.
Summary/Conclusion: This is a first in human clinical trial to evaluate safety, determine
the recommended phase 2 dose and estimate efficacy of a novel microbial-derived peptide
therapeutic vaccine, EO2463, in patients with indolent non-hodgkin lymphoma as monotherapy
and in combination with lenalidomide and rituximab.
P1116: SAFETY, PHARMACOKINETIC (PK), PHARMACODYNAMIC (PD) AND ACTIVITY OF THE HIGHLY
SELECTIVE PHOSPHOINOSITIDE 3-KINASE INHIBITOR DELTA (PI3KΔ) INHIBITOR IOA-244 IN PATIENTS
WITH FOLLICULAR LYMPHOMA (FL)
C. Carlostella1,*, M. Lahn2, T. Hammett2, L. van der Veen2, Z. Johnson2, C. Pickering2,
A. Santoro1
1IRCSS Humanitas Research Hospital, Milan, Italy; 2iOnctura, Geneva, Switzerland
Background: IOA-244 is a highly selective inhibitor of PI3Kδ, and has a favourable
safety and ADME profile in patients (pts) with solid malignancies. In pts with uveal
melanoma, IOA-244 also reduced circulating Tregs counts at the biologically effective
dose (BED) range.
Aims: As part of the First-in-human (FiH) dose escalation study, pts with FL received
IOA-244 at the BED range established in patients with solid malignancies to confirm
its favourable safety and PK profile in pts with haematologic malignancies.
Methods: IOA-244 was investigated in a two-part FIH study. Part A explored the continuous
daily dosing of IOA-244 at 10, 20, 40 and 80 mg in pts with solid malignancies and
at 20 mg and 80 mg in pts with FL. Part B consists of expansion cohorts of specific
tumour indications, including pts with lymphoma. Primary objective: safety of the
anticipated BED, or the recommended phase 2 dose (RP2D). Secondary objectives: PK;
PD (e.g., inhibition of CD63 expression on basophils, changes in immune cell subsets
in peripheral blood); Lugano-based responses; PFS and OS
Results: While Part A for solid malignancies have previously been reported (Di Giacomo
et al 2021), we here report for the first time the safety, PK, PD and activity of
pts with FL. This part of the study is still ongoing in patients with FL and is anticipated
to be completed in 2022. Pts at the first cohort of 20 mg QD daily (4/4; 2 female
and 2 males) had no DLT and no dose interruptions. Transient platelet reduction (G3)
and AST/ALT elevation (G2) were observed in 1/4 pts, which improved while pt was on
therapy. The PK and ADME profile was consistent with the ones observed in pts with
solid malignancies. One patient had FL and DLBCL as prior co-primary malignancy with
7 prior lines of therapy. The other 3 pts had 1-3 prior lines of therapy. The best
response was SD (1/3 pts) and all pts progressed after completing 2 cycles. LDH was
initially elevated in 2/4 pts which subsequently decreased on therapy. 3/4 pts had
low Treg counts at baseline. Additional pts are being recruited at the RP2D for solid
malignancies, at 80 mg QD.
Reference:
Di Giacomo et al. Annals of Oncology (2021) 32 (suppl_7): S1428-S1457. 10.1016/annonc/annonc787
Summary/Conclusion: The PK and ADME profile of IOA-244 in pts with FL appears to match
that in pts with solid malignancies. Additional pts at the 80 mg QD dose are expected
to match the safety observed in pts with solid malignancies. In contrast to other
PI3Kd inhibitors, IOA-244 is highly selective and may therefore target Tregs in pts
with FL without off-target effects induced by inhibition of other PI3K isoenzymes.
P1117: THREE-YEAR FOLLOW-UP OF OUTCOMES WITH KTE-X19 IN PATIENTS WITH RELAPSED/REFRACTORY
MANTLE CELL LYMPHOMA IN ZUMA-2
M. L. Wang1,*, J. Munoz2, A. Goy3, F. L. Locke4, C. A. Jacobson5, B. T. Hill6, J.
M. Timmerman7, H. Holmes8, I. W. Flinn9, D. B. Miklos10, J. M. Pagel11, M. J. Kersten12,
R. Houot13, A. Beitinjaneh14, W. Peng15, X. Fang15, R. R. Shen15, R. Siddiqi15, I.
Kloos15, P. M. Reagan16
1The University of Texas MD Anderson Cancer Center, Houston, TX; 2Banner MD Anderson
Cancer Center, Gilbert, AZ; 3John Theurer Cancer Center, Hackensack University, Hackensack,
NJ; 4Moffitt Cancer Center, Tampa, FL; 5Dana-Farber Cancer Institute, Boston, MA;
6Cleveland Clinic Foundation, Cleveland, OH; 7David Geffen School of Medicine at UCLA,
Los Angeles, CA; 8Texas Oncology, Dallas, TX; 9Sarah Cannon Research Institute and
Tennessee Oncology, Nashville, TN; 10Stanford University School of Medicine, Stanford,
CA; 11Swedish Cancer Institute, Seattle, WA, United States of America; 12Amsterdam
UMC, University of Amsterdam, Amsterdam, Cancer Center Amsterdam, The Netherlands,
on behalf of HOVON/LLPC, Amsterdam, Netherlands; 13CHU Rennes, Université Rennes,
INSERM & EFS, Renne, France; 14University of Miami, Miami, FL; 15Kite, a Gilead Company,
Santa Monica, CA; 16University of Rochester Medical Center, Rochester, NY, United
States of America
Background: Brexucabtagene autoleucel (KTE-X19) is an autologous anti-CD19 chimeric
antigen receptor (CAR) T-cell therapy approved for the treatment of patients (pts)
with relapsed/refractory (R/R) mantle cell lymphoma (MCL). In ZUMA-2, a 93% objective
response rate (ORR; 67% complete response [CR] rate) was reported with KTE-X19 in
pts with R/R MCL (median follow-up: 12.3 mo; 60 efficacy-evaluable pts; Wang et al.
N Engl J Med. 2020).
Aims: To present updated outcomes from ZUMA-2 with 2 years of additional follow-up.
Methods: Adult pts (≥18 years) with R/R MCL underwent leukapheresis and conditioning
chemotherapy followed by a single infusion of KTE-X19. Minimal residual disease (MRD)
was an exploratory endpoint (sensitivity 10-5) evaluated in peripheral blood using
next-generation sequencing. Updated results are reported for all 68 treated pts.
Results: After 35.6 mo median follow-up, the ORR (CR + partial response) was 91% (95%
CI, 81.8-96.7), with a 68% CR rate (95% CI, 55.2-78.5). The median duration of response
(DOR) was 28.2 mo (95% CI, 13.5-47.1), with 25 of 68 treated pts (37%) still in ongoing
response (all CR) at data cutoff. Late relapse >24 mo post-infusion was infrequent
(n=3). The medians for progression-free survival (PFS) and overall survival (OS) were
25.8 mo (95% CI, 9.6-47.6) and 46.6 mo (95% CI, 24.9-not estimable), respectively.
MRD was analyzed in 29 pts total; 24 of 29 were MRD-negative at mo 1, and 15 of 19
with available data were MRD-negative at mo 6. At data cutoff, the medians for DOR,
PFS, and OS in the 15 MRD-negative pts were all not reached, vs 6.1, 7.1, and 27.0
mo, in the 4 MRD-positive pts, respectively. MRD-negative status at mo 1, 3, and 6
was associated with durable response, with 55%, 71%, and 69% of MRD-negative pts at
those timepoints remaining in ongoing CR at data cutoff. Circulating tumor DNA analysis
of MRD at mo 3 and 6 was predictive of relapse (AUC 0.80 and 0.75, respectively).
No new safety signals were observed. Only 3% of treatment-emergent adverse events
(AEs) of interest occurred since the primary report. The most frequent Grade ≥3 AE
was neutropenia (1 [1%] Grade 3; 7 [10%] Grade 4). Two pts had KTE-X19-related Grade
3 serious infections: pneumonia and upper respiratory tract infection (n=1); influenza
(n=1). There were no new cytokine release syndrome AEs and 1 new serious neurologic
AE of Grade 3 encephalopathy (13.0 mo post-infusion) that was considered not related
to study treatment. Three new Grade 5 AEs occurred, none of which were considered
related to study treatment: Salmonella bacteremia (24.9 mo post-infusion), myelodysplastic
syndrome (25.2 mo post-infusion), and acute myeloid leukemia (37.5 mo post-infusion).
Summary/Conclusion: These data represent the longest follow-up of CAR T-cell therapy
in pts with MCL to date and suggest that KTE-X19 induces durable long-term responses
with manageable safety and low late relapse potential in R/R MCL.
P1118: PROGNOSTIC SIGNIFICANCE OF ABSOLUTE MONOCYTE COUNT AND LYMPHOCYTE TO MONOCYTE
RATIO IN MUCOSA-ASSOCIATED LYMPHOID TISSUE (MALT) LYMPHOMA
Y. Li1,*, C. Shang1, Y. Ren1, L. Wang1, J. Li1, W. Xu1
1Hematology, the First Affiliated Hospital of Nanjing Medical University, Nanjing,
China
Background: Extranodal marginal zone B-cell lymphoma of the MALT lymphoma is a unique
type of indolent lymphoma. It usually presents with local disease and exhibit an indolent
clinical course, but some patients have disseminated systemic symptoms and require
systemic treatment. Monocytes, as part of the important immune cells in tumor microenvironment,
are closely linked to the pathogenesis and progression of malignancies. In recent
studies, the absolute monocyte count (AMC) and lymphocyte to monocyte ratio (LMR)
are shown to reflect the host systemic immunity states and have prognostic significance
in different kinds of lymphoma.
Aims: Our study aims to explore the prognostic significance of AMC and LMR in MALT
lymphoma, and to propose a more accurate prognostic index based on MALT-IPI.
Methods: Baseline clinical characteristics collected from the medical records at diagnosis.
PFS and OS were analyzed with the Kaplan-Meier method and compared using the log-rank
test. X-tile analysis was performed to identify cut-off value. Chi-square test was
performed for comparisons between groups. Cox proportional-hazards regression models
were used for univariate and multivariate analysis. Statistical analyses were performed
with MedCalc 19.5.6, SPSS software 26.0, R software version 4.0.4 and Graphpad Prism
9.0.
Results: 316 MALT lymphoma patients were enrolled in this study. A statistically evident
dominance showed that LMR was related to age, LDH level, β2-MG level, B symptom, ECOG
PS and systemic therapy. With a median follow-up time of 39.1 months (1–237 months),
median PFS was 146.4 months and median OS was not reached, estimated PFS rate at 3
years and 5 years were 84.1% and 79.6%, estimated OS rate at 3 years and 5 years were
94.9% and 92.4%, respectively. According to Figure 1, high AMC group (>0.6×109/L)
and low LMR group (<1.8) were associated with poor outcomes. We performed Cox proportional-hazards
regression for PFS and OS, MALT-IPI, ECOG PS and LMR were identified to have independent
prognostic significance for PFS while MALT-IPI, β2-MG and LMR were independently associated
with poor OS. Figure 2A showed that low level of LMR in subgroup was related to a
poor PFS. The prognostic value of low level of LMR for PFS was more significant in
age (p value for interaction =0.020). Across a subgroup analysis of OS (Figure 2B),
the prognostic value of low level of LMR was consistent. Since we have confirmed AMC
showed prognostic significance in MALT lymphoma survival. We combined MALT-IPI and
AMC to generate a new prognostic index named ‘MALT-IPI-M’, which included four parameters:
age≥70 years, Ann Arbor stage III or IV, serum LDH level >UNL, and LMR <1.8. MALT-IPI-M
classified patients into low-risk group (the MALT-IPI-M=0), intermediate-risk group
(the MALT-IPI-M=1) and high-risk group (the MALT-IPI-M≥2), the proportions of different
stratifications in our cohort are 52.5%, 32.9% and 14.6%, respectively. As shown in
Figure 3, we generated Receiver-operator characteristic (ROC) curves to compare the
prognostic prediction capability of MALT-IPI and MALT-IPI-M. The area under the curves
(AUCs) for MALT-IPI-M were 0.682 for PFS and 0.804 for OS, which was larger than AUCs
for MALT-IPI, 0.654 for PFS and 0.788 for OS. This illustrated that MALT-IPI-M has
a better capability of distinguishing MALT patients with different risk.
Image:
Summary/Conclusion: In conclusion, our findings suggest that low level LMR at diagnosis
might be associated with inferior PFS and OS in patients with MALT lymphoma. Incorporating
LMR into MALT-IPI may permit a more accurate risk assessment of disease progression.
P1119: THE ORAL PI3KΔ INHIBITOR LINPERLISIB FOR THE TREATMENT OF RELAPSED OR REFRACTORY
FOLLICULAR LYMPHOMA: A SINGLE-ARM MULTICENTER PHASE 2 CLINICAL TRIAL
T. wang1, X. sun2, L. qiu3, H. su4, J. cao5, Z. li6, Y. song7, L. zhang8, S. yi9,
L. qiu9,*, J. zhou10, H. wu11, W. zhang12, J. li13, K. zhou14, H. zhou15, Y. yang16,
Z. li17, H. cen18, Z. cai19, Z. zhang20, W. fu21, J. jin19, F. li22, W. wu23, X. gu24,
W. zhu25, L. liu26, Z. li27
1Department of Lymphoma and Myeloma, Institute of Hematology & Blood Diseases Hospital,
Chinese Academy of Medical Sciences, Tianjin; 2department of medical oncology, the
second hospital of dalian medical university, dalian; 3Department of Lymphoma, Tianjin
Medical University Cancer Institute and Hospital, Tianjin; 4Department of Lymphoma,
The Fifth Medical Center of PLA General Hospital, Beijing; 5Fudan University Shanghai
Cancer Center, Shanghai; 6Sun Yat-sen University Cancer Center, guangzhou; 7Peking
University School of Oncology, Beijing Cancer Hospital and Institute, Beijing; 8West
China Hospital, Sichuan University, chengdu; 9Institute of Hematology & Blood Diseases
Hospital, Chinese Academy of Medical Sciences, Tianjin; 10Department of Hematology,
Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology;
11Department of Medical Oncology, Hubei Cancer Hospital, Wuhan; 12Department of Hematology,
Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking
Union Medical College, Beijing; 13Hematology, Ruijin Hospital Affiliated to Shanghai
Jiao Tong University School of Medicine, Shanghai; 14Department of Hematology, The
Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou;
15Department of Lymphoma & Hematology, Hunan Cancer Hospital, The Affiliated Cancer
Hospital of Xiangya School of Medicine, Central South University, Changsha; 16Department
of Lymphoma and Head and Neck Cancer, Fujian Provincial Cancer Hospital, The Affiliated
Tumor Hospital of Fujian Medical University, Fuzhou; 17Department of Hematology, The
First Affiliated Hospital of Xiamen University and Institute of Hematology, School
of Medicine, Xiamen University, Xiamen; 18Department of Haematology/Oncology and Paediatric
Oncology, Guangxi Medical University Affiliated Cancer Hospital, Nanning, China, Nanning;
19Department of Hematology, The First Affiliated Hospital, Zhejiang University School
of Medicine, Hangzhou; 20Department of Medical Oncology, Sichuan Cancer Hospital and
Institute, Chengdu; 21Department of Hematology, The Myeloma & Lymphoma Center, Shanghai
Changzheng Hospital, The Second Military Medical University, Shanghai; 22Department
of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang; 23Department
of Internal Medicine and Oncology, Zhongshan Hospital, Xiamen University, Xiamen;
24Department of Hematology, The First Affiliated Hospital of Guangzhou University
of Chinese Medicine; 25Department of Oncology, Zhujiang Hospital of Southern Medical
University, Guangzhou; 26Department of Hematology, The Fourth Hospital of Hebei Medical
Universit, Shijiazhuang; 27Hematological Disorders, Institute of Hematology and Blood
Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College,
Tianjin, China
Background: The orally active phosphatidylinositol 3-kinase delta (PI3Kδ) inhibitor
linperlisib which conveyed acceptable safety and notable efficacy for B-cell lymphoma
medications in a previous phase 1 dose escalation study was investigated in this phase
2 study for the treatment of relapsed and/or refractory follicular lymphoma (FL) patients
who had received at least two prior systemic treatments
Aims: To explore the satefy and efficacy of linperlisib in FL
Methods: Linperlisib was administered at 80 mg qd, po in a 28-day cycle until disease
progression or intolerable toxicity occurred. The primary outcome was the objective
response rate (ORR), while secondary outcomes included the duration of response (DOR),
progression-free survival (PFS), overall survival (OS), disease control rate (DCR)
and the drug safety profile.
Results: For 84 FL patients in the full analysis set (FAS) the ORR was 79.8% (95%
CI: 69.6-87.8, 67 patients), with 13 patients achieving a complete response and 54
a partial response. The median DOR was 12.3 months (range, 9.3-15.9) with 6-month
and 12-month DORs of 80.0% (95% CI: 67.4-88.1) and 55.3% (95% CI: 40.6-67.8). The
median PFS was 13.4 months (95% CI: 11.1-16.7), with 6-month and 12-month PFS rates
of 78.7% (95% CI: 67.5-86.3) and 53.1% (95% CI: 40.3-64.3). The median OS was not
reached, while 6-month and 12-month OS rates were 97.6% (95% CI: 90.6-99.4) and 91.4%
(95% CI: 82.7-95.8), respectively. The most frequent any-grade treatment related AEs
(TRAE) were neutropenia (47%), hypertriglyceridemia (25%), while the most frequent
TRAEs ≥ grade 3 were neutropenia (15%), infectious pneumonia (19.4%) and interstitial
lung disease (6.5%). AEs that were potentially immune-mediated, such as diarrhea,
colitis, pneumonitis, and transaminitis, were observed at significantly lower incidences
of occurrence.
Summary/Conclusion: Linperlisib demonstrated compelling clinical efficacy and a promising
safety profile, which has the potential to surpass the approved PI3K inhibitors for
the treatment of relapsed or refractory FL patients after two prior therapies.
P1120: THE EFFICACY AND SAFETY OF ZANUBRUTINIB AND DEXAMETHASONE IN SYMPTOMATIC WALDENSTROM
MACROGLOBULINNEMIA
A. Liu1, J. Yin1, J. Lu2, Y. Ma3, D. Gao4, L. Hua5, Y. Tian1, Y. Jian1, W. Chen1,*
1Beijing Chaoyang Hospital, affiliated to Capital Medical University; 2Peking University
People’s Hospital, Beijing; 3Second hospital of Shanxi Medical University, Taiyuan;
4The Affiliated Hospital of Inner Mongolia Medical University, Huhehaote; 5Affiliated
Hospital of Hebei University, Shijiazhuang, China
Background: Bruton tyrosine kinase (BTK) inhibition is an effective treatment approach
for patients with Waldenström macroglobulinemia (WM),but the efficacy of single drug
is limited.
Aims: This single arm study (ChiCTR2000038140) evaluated the efficacy and safety of
the combination of zanubrutinib, a novel, highly selective BTK inhibitor, and dexamethasone
in patients with WM.
Methods: Symptomatic patients with WM were enrolled to the regimen of zanubrutinib
and dexamethasone (ZD)*. The primary endpoint was objective response rate (ORR), progression-free
survival (PFS). Key secondary endpoints included the proportion of patients achieving
a complete or very good partial response (CR or VGPR), duration of response (DOR),
Time to response (TOR), disease burden, and safety. The control group** were matched
patients treated by chemotherapy or immunechemotherapy previously in Beijing Chaoyang
Hospital.
Results: A total of 22 Patients with WM were enrolled in this study, median age 67(36-89);
68.2% males, 12 patients were untreated, others were treated patients. IPSS assessment
grade1 23.8%; grade2 19.0%; grade3 57.2%. 90.9% (20/22) patients with MYD88L265P mutation,
27.3% (3/11) patients with CXCR4 mutation.
21 patients received ≥1 dose of study treatment. Median follow-up of 8.2 months, median
DOR and PFS were not reached; 95% of patients were progression-free at 6 months. ORR
was 95% in those (17/18) received ZD regimen more than 2 months. No patient achieved
a CR. 33.3% of patients in ZD group achieved a VGPR, time to VGPR within 3 months
in 57.1% of patients,a statistically significant difference with control group (0%,
P = 0.001). Time to PR in ZD group was 2 months, much faster than control group (11
months) (P = 0.023) by K-M analysis.
The study-safety profile was consistent with previous BTK inhibitor clinical trial
data. 45% of patients had any grade AEs. In which, the most frequent grade <=2 AEs
were hemorrhage(18.2% all grade 1), rash(9.1%), hyperglycemia (13.6%), infection(9.1%),
nausea and vomiting (9.1%), hypogammaglobulinemia(4.5%), neutropenia(9.1%). Grade
3/4 AEs were atrial fibrillation(4.5%), leading to treatment discontinuation. Other
cause of treatment discontinuation is hyperglycemia, bowel obstruction by disease.
Comments
*The regimen of ZD: Zanubrutinib 240mg d1-28, dexamethasone 20mg D1-4,15-18. Patients
more than 75 years old, Zanubrutinib 160mg d1-28, dexamethasone 10mg D1-4,15-18. After
8 cycle, Zanubrutinib use as maintenance.
** The control group: 22 treated patients with WM, median age 69.5(39-84); 68.2% males.
IPSS Grade1 21.4%; Grade2 32.1%; Grade3 42.9%; Unknown 3.6%. 75%(9/12) patients with
MYD88L265P mutation, 33%(1/3) patients with CXCR4 mutation. Treatment included chemotherapy(containing
nitrogen mustard phenylbutyrate, fludarabine, cyclophosphamide), proteasome inhibitor
regimens, rituximab regimens and immunomodulator regimens.
Summary/Conclusion: These results demonstrate that zanubrutinib and dexamethasone
are quickly effective in the treatment of WM, with more deeper response and less toxicity.
P1121: TAKEAIM LYMPHOMA- AN OPEN-LABEL, DOSE ESCALATION AND EXPANSION TRIAL OF EMAVUSERTIB
(CA-4948) IN COMBINATION WITH IBRUTINIB IN PATIENTS WITH RELAPSED OR REFRACTORY HEMATOLOGIC
MALIGNANCIES
E. Joffe1,*, G. Nowakowski2, H. Tun3, A. Rosenthal4, M. Lunning5, R. Ramchandren6,
C.-C. Li7, L. Zhou7, E. Martinez7, R. von Roemeling7, R. Earhart7, M. McMahon7, I.
Isufi8, L. Leslie9
1MSKCC, NY; 2Mayo Clinic-Minnesota, Rochester, Rochester; 3Mayo Clinic-Florida, Jacksonville;
4Department of Hematology, Mayo Clinic- Arizona, Phoenix; 5University of Nebraska,
Omaha; 6University of Tennessee Medical Center, Knoxville; 7Curis, Lexington; 8Yale
New Haven Hospital, New Haven; 9John Theurer Cancer Center, Hackensack, United States
of America
Background: Emavusertib (CA-4948) is a novel oral inhibitor of interleukin-1 receptor-associated
kinase 4 (IRAK4), which is essential for toll-like receptor (TLR) and interleukin-1
receptor (IL-1R) signaling in B cell proliferation. IRAK4 forms a Myddosome complex
with MYD88 adaptor protein and drives overactivation of nuclear factor-kappa B (NF-κB),
causing inflammation and tumor growth. Emavusertib has been reported to be well tolerated
and active as monotherapy in heavily pretreated patients with relapsed/refractory
(R/R) non-Hodgkin lymphoma (NHL). Preclinical studies demonstrated that tumor resistance
and survival via IRAK4 activation could be delayed or reversed. Emavusertib crossed
the blood-brain barrier in a murine PDX model of pCNS lymphoma, resulting in tumor
response and prolonged survival. In combination with Bruton tyrosine kinase (BTK)
inhibitors, emavusertib showed in vivo synergy in B-cell NHL. Here we present an update
on the preliminary efficacy data of emavusertib + ibrutinib in R/R hematologic malignancies.
Aims: Assessment of safety and clinical activity of emavusertib in combination with
ibrutinib at full prescribed dose.
Methods: This is an ongoing open-label trial (NCT03328078) of emavusertib as monotherapy
and in combination with ibrutinib.
Part A1
(completed) dose escalation of emavusertib as monotherapy; the recommended phase 2
dose (RP2D) is 300 mg BID with continuous oral dosing.
Part A2
(dose escalation in combination with ibrutinib), and
Part B
(a basket design of 4 expansion cohorts of emavusertib and ibrutinib: BTK-naïve MZL,
DLBCL, or PCNSL and NHL with adaptive resistance to ibrutinib). The primary endpoints
of
Parts A1
and
A2
include safety, tolerability, and RP2D. The primary endpoints of
Part B
include CR or ORR, with key secondary endpoints of DOR, DCR, PFS and OS following
treatment of emavusertib at dose levels of 200 (DL1) or 300 mg BID (DL2) with ibrutinib
at full prescribed dose.
Results: As of December 7th, 2021, 35 heavily pretreated NHL patients have received
emavusertib monotherapy (median age 66 years, range 50-87), of which six patients
have been on emavusertib for approximately 1 year or longer, suggesting emavusertib
has a long-term acceptable safety and tolerability profile at RP2D (dose level of
300 mg BID). In
Part A2
, 10 patients are treated with emavusertib + ibrutinib (median age 65 years, range
56-82). Median number of prior lines of anti-cancer therapies is 3 (range 1-8). No
DLTs were observed at 200 or 300 mg dose levels to date. The preliminary efficacy
data of seven evaluable patients with combination therapy showed 1 CR (MCL), 2 PR
(MCL and MZL), 3 SD, and 1 PD, 3 of whom had failed prior ibrutinib. The preliminary
data indicate the combination therapy may overcome ibrutinib resistance.
Summary/Conclusion: Emavusertib as a monotherapy and in combination with ibrutinib
is well tolerated with an acceptable long term safety profile and promising efficacy.
Part A2
is transitioning to
Part B
basket cohorts of MZL, ABC-DLBCL, PCNSL and NHL with adaptive resistance to ibrutinib.
P1122: EFFICACY OF ALPELISIB IN PI3K-DRIVEN LANGERHANS CELL HISTIOCYTOSIS
R. Mazor1,*, B. Durham2, O. Abdel-Wahab2, E. Diamond3, G. Itchaki4, M. Ben-Sasson5,6,7,
O. Hershkovitz-Rokah1,8,9, O. Shpilberg1,9,10
1Clinic of Histiocytic Neoplasms, Institute of Hematology, Assuta Medical Center,
Tel Aviv, Israel; 2Human Oncology and Pathogenesis Program; 3Department of Neurology,
Memorial Sloan-Kettering Cancer Center, New York, United States of America; 4Department
of Hematology, Rabin Medical Center, Petah Tikva; 5The Institute for Pain Medicine,
Rambam Medical Center; 6The Rappaport school of medicine, Technion, Haifa; 7Meuhedet
Health Maintenance Organization, Zikhron Ya’akov; 8Department of Molecular Biology,
Faculty of Natural Sciences, Ariel University, Ariel; 9Translational Research Lab,
Assuta Medical Center, Tel Aviv; 10The Adelson School of Medicine, Ariel University,
Ariel, Israel
Background: Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasm
characterized by the accumulation of clonal dendritic cells expressing CD1a and CD207
(Langerin). LCH affects both children and adults and typically ranges from an indolent
unifocal form to a progressive multisystemic disease. In the past decade, in depth
molecular profiling of LCH as well as other histiocytic neoplasms demonstrated that
these diseases are fundamentally dependent on somatic MAPK activating mutations, with
the canonical V600E BRAF mutation emerging in more than 50% of LCH patients. Nonetheless,
MAPK independent driver mutations were previously reported and anecdotal evidence
of successful targeted treatment of various histiocytoses harboring non-canonical
drivers such as RET, ALK and CSF1R were previously described in the literature.
Aims: To report a case of PI3K driven LCH treated with an isoform specific PIK3CA
inhibitor.
Methods: Case summary: a 46 year-old mother of 3 presented with complaints of progressive
headaches, polydipsia and polyuria. Following a pathological water deprivation test
and identification of pituitary stalk thickening on MRI, a diagnosis of central diabetes
insipidus (CDI) was established and desmopressin treatment was initiated. However,
10 months following her CDI diagnosis, the patient began to complain of sub febrile
episodes of fever and worsening dry cough. Imaging studies including PET/CT disclosed
extensive bilateral reticulo-nodular infiltration, nodules with cystic transformation
and ground glass opacity, suspicious of pulmonary LCH. Histological examination of
a wedge biopsy specimen obtained from the lungs confirmed the diagnosis of LCH and
consequent molecular studies identified the M1043V PIK3CA mutation. During her medical
investigation, the patient’s conditioned worsened, with new disease foci emerging
– including cervical lymphadenopathy and a lytic vertebral lesion involving D11. The
patient then received palliative radiotherapy directed at the D11 lytic lesion followed
by escalating systemic therapy with single agent Alpelisib, via the Managed Access
Program at Novartis. Response to treatment was assessed clinically and using PET/CT.
At the molecular level, the effect of Alpelisib was assessed by measuring PTEN expression
levels and cell cycle regulating non-coding RNA molecules by using quantitative real-time
polymerase chain reaction (qRT-PCR).
Results: Treatment with Alpelisib resulted in rapid amelioration of night sweats within
6 days, decrease in back pain within 3 weeks and normalization of all abnormally FDG
avid disease foci within 3 months of treatment. The complete metabolic response observed
persists currently more than a year following treatment initiation. Treatment with
Alpelisib is well tolerated except for a mild increase in HbA1C levels to 6.7%. Moreover,
PTEN, the main regulator of the PI3K pathway was found to be upregulated following
3 and 9 months of treatment, in parallel to downregulation of small non-coding RNAs
molecules that regulates its expression.
Image:
Summary/Conclusion: This is the first study demonstrating that the alpha catalytic
subunit of PI3K is a targetable non-canonical driver of LCH.
P1123: TEMPO: A PHASE 2, RANDOMIZED, OPEN-LABEL, 2-ARM STUDY COMPARING TWO INTERMITTENT
DOSING SCHEDULES OF DUVELISIB IN SUBJECTS WITH INDOLENT NON-HODGKIN LYMPHOMA (INHL)
V. Vorobyev1,*, D. H. Yoon2, M. Kaźmierczak3, S. Grosicki4, C. Tarella5, A. Genua6,
J. S. Kim7, D. Cohan8, M. Daugherty8, I. W. Flinn9, P. L. Zinzani10, L. I. Gordon11
1S. P. Botkin City Clinical Hospital, Moscow, Russia; 2University of Ulsan College
of Medicine, Seoul, South Korea; 3Poznań University of Medical Sciences, Poznań; 4Silesian
Medical University, Katowice, Poland; 5IEO Istituto Europeo di Oncologia IRCCS, Milano;
6Università degli studi di Perugia, Terni, Italy; 7Yonsei University College of Medicine,
Severance Hospital, Seoul, South Korea; 8Secura Bio, Las Vegas; 9Sarah Cannon Research
Institute-Tennessee Oncology, Nashville, United States of America; 10University of
Bologna, Istituto Di Ematologia, Bologna, Italy; 11Northwestern University Feinberg
School of Medicine and the Robert H. Lurie Comprehensive Cancer Center, Chicago, United
States of America
Background: Dosing of duvelisib at 25 mg orally BID has been shown to be active in
iNHL, as the pivotal phase 2 DYNAMO study (NCT01882803) in iNHL met its primary endpoint
of overall response rate (ORR) assessed by an IRC.
Aims: The TEMPO study (NCT04038359; supported by Secura Bio) examined the effects
of prespecified 2-week dose holidays on tumor responses and safety/tolerability in
patients (pts) with iNHL based on data from a phase 3 trial (DUO) in CLL/SLL that
demonstrated no negative impacts of (unscheduled) dose interruptions on response (Flinn
I et al. J Clin Oncol. 2019;37(15):7523).
Methods: In TEMPO, patients were randomized to receive duvelisib 25 mg BID for one
10-week cycle followed by 25 mg BID on Weeks 3 and 4 of each subsequent 4-week cycle
(Arm 1) or duvelisib 25 mg BID on Weeks 1, 2, 5, 6, 9, and 10 of one 10-week cycle,
and then on Weeks 3 and 4 of each subsequent 4-week cycle (Arm 2). Pts continued treatment
until progressive disease (PD), unacceptable toxicity, or study withdrawal. ORR by
investigator assessment, based on 2007 revised International Working Group (IWG) criteria
was the primary endpoint.
Results: As of data cutoff on 5 May 2021, 65 treated pts had at least 4 months of
follow-up on or after study drug treatment. Across both arms, the median age was 63
years (range 34-85), and 47 (72.3%) of patients remained on duvelisib at the time
of this analysis (see Table). The patient population was heavily pretreated (total
prior lines: 36.9% = 1; 26.2% = 2; 10.8% = 3; and 26.2% ≥ 4); 93.8% of pts received
prior rituximab and 43.1% prior bendamustine. As of data cutoff, 27.7% of pts have
discontinued duvelisib, 19.7% due to PD (21.2% in Arm 1; 18.8% in Arm 2). Four patients,
all in Arm 1, had a TEAE that led to treatment discontinuation. Across both arms,
the ORR was 60% (39 of 65 pts), and the complete response (CR) rate was 12.3% (8 of
65 pts). Results were similar in pts who received prior rituximab (59.0%) and those
who received prior bendamustine (57.1%) and were relatively consistent regardless
of the number of prior treatments received (62.5%, 64.7%, 54.2% for pts with 1, 2,
or ≥ 3 prior lines of therapy, respectively).
Overall, the incidence of TEAEs was higher in Arm 1 vs Arm 2, including grade ≥ 3
AEs (ALT elevation 30.3% vs 3.1%; AST elevation 27.3% vs 3.1%; GI disorders 9.1% vs
0; infections 9.1% vs 3.1%; rash 6.1% vs 3.1%; and drug reaction with eosinophilia
3.0% vs 0); the incidence of grade ≥ 3 neutropenia was higher in Arm 2 (9.1% vs 18.8%).
TEAEs leading to discontinuation of study drug occurred only in Arm 1 (15.2%; 5 of
33 pts) and included ALT/AST elevations (2 of 33 pts; 6.1%), skin and subcutaneous
tissue disorders (2 of 33 pts; 6.1%), and pneumonia (1 of 33 pts; 3.0%). There was
1 death on study in Arm 1 due to disease progression. No TEAE-related deaths occurred
in either arm.
Image:
Summary/Conclusion: Based on this interim analysis, continuous followed by intermittent
dosing (Arm 1) and intermittent dosing (Arm 2) are both safe and effective in patients
with iNHL. Efficacy was seen irrespective of prior lines of therapy, including those
heavily pretreated and/or treated with prior rituximab or bendamustine. The ORR and
CR are consistent with those seen in DYNAMO. Because these are early data, the differences
between arms in certain types of AEs or discontinuations due to AEs are difficult
to interpret. The study confirms the overall therapeutic profile of duvelisib and
demonstrates that intermittent dosing may decrease the incidence of AEs without compromising
efficacy.
P1124: MOSUNETUZUMAB RETREATMENT IS EFFECTIVE AND WELL-TOLERATED IN PATIENTS WITH
RELAPSED OR REFRACTORY B-CELL NON-HODGKIN LYMPHOMA
C. Y. Cheah1,*, N. L. Bartlett2, S. Assouline3, S. J. Schuster4, W. Seog Kim5, M.
Shadman6, I. Isufi7, S. Yin8, M. Y. Doral8, J. Sit8, V. Chen8, H. Huang9, M. Zhou10,
M. C. Wei8, L. E. Budde11
1Linear Clinical Research, Sir Charles Gairdner Hospital and The University of Western
Australia, Perth, Australia; 2Washington University School of Medicine, Siteman Cancer
Center, St Louis, MO, United States of America; 3Jewish General Hospital, Montreal,
Canada; 4University of Pennsylvania, Philadelphia, PA, United States of America; 5Samsung
Medical Centre, Seoul, South Korea; 6Fred Hutchinson Cancer Research Center, Seattle,
WA, Seattle, WA; 7Yale School of Medicine, New Haven, CT; 8Genentech, Inc., South
San Francisco, CA, United States of America; 9Hoffmann-La Roche Ltd, Mississauga,
Canada; 10F. Hoffmann-La Roche, Shanghai, China; 11City of Hope, Duarte, CA, United
States of America
Background: The prognosis for heavily pretreated patients (pts) with relapsed/refractory
(R/R) non-Hodgkin lymphoma (NHL) is poor and there is a need for effective treatment
options (Batlevi, et al. 2020; Crump, et al. 2017). Mosunetuzumab (M) is a T-cell
engaging CD20xCD3 bispecific monoclonal antibody that redirects T cells to eliminate
malignant B cells (Sun, et al. 2015; Budde, et al. 2022). Results from a single-arm,
Phase I/II study (NCT02500407) of M in pts with R/R aggressive and indolent NHL showed
durable complete responses (CR) and a manageable safety profile (Budde, et al. 2022).
Specifically, in pts with R/R follicular lymphoma (FL) after ≥2 prior therapies, M
demonstrated a 60% CR rate and an 80% objective response rate with a median duration
of response (DoR) of 22.8 months (Budde, et al. ASH 2021). Initial M treatment had
a fixed duration (8 cycles for pts who achieved CR; 17 cycles for pts with partial
response or stable disease after 8 cycles); however, for pts who achieved a CR but
had progressive disease (PD) after initial treatment completion, M retreatment could
be provided. We report additional data from this ongoing study to describe the M retreatment
experience of pts with R/R NHL.
Aims: To evaluate the efficacy and safety of M monotherapy (mono) retreatment in pts
with R/R NHL from an ongoing Phase I/II study (NCT02500407).
Methods: Pts with R/R NHL who achieved CR after initial treatment with intravenous
(IV) M mono but subsequently developed PD after treatment completion were eligible
for retreatment. Pts were retreated with IV M mono at a previously tested dose and
schedule, which included step-up dosing for cytokine release syndrome (CRS) mitigation.
All pts provided informed consent.
Results: At the clinical cut-off date, March 15, 2021, 12 pts received M retreatment;
6 pts completed retreatment and 6 pts discontinued retreatment (PD, n=5; start of
another anti-lymphoma therapy, n=1). M retreatment step-up dose schedules were as
follows (Cycle [C] 1, Day [D] 1/C1D8/C1D15/C2D1/C3+D1): 1/2/13.5mg (n=1), 1/2/20mg
(n=1), 1/2/27mg (n=1), 1/2/40.5mg (n=1) and 1/2/60/60/30mg (n=8). Median number of
M retreatment cycles received was 8.0 (range, 2–13). Overall, 8 pts with R/R FL, 2
pts with R/R diffuse large B-cell lymphoma (DLBCL), 1 pt with R/R transformed FL (trFL)
and 1 pt with R/R mantle cell lymphoma (MCL) were included in the analysis. During
retreatment, 9 (75.0%) pts had an objective response (6/8 pts with R/R FL; 2/2 pts
with R/R DLBCL; 1/1 pt with trFL; and 0/1 pt with MCL; Table). Six/12 pts achieved
a CR (4/8 pts with R/R FL, 1/2 pts with R/R DLBCL; 1/1 pt with R/R trFL; and 0/1 pt
with MCL). All pts who received M retreatment were included in the safety population
and had ≥1 adverse event (AE). The most common any-grade AE and Grade 3–4 AE was hypophosphatemia
(n=7, 58.3% [each]; none were serious and all events resolved with or without treatment).
Four pts experienced ≥1 serious AE. No Grade 5 events were reported and no pts had
an AE that led to treatment discontinuation. Three pts (25.0%) had low-grade CRS (by
Lee, 2014; Grade 1, n=2; Grade 2, n=1) and there were no Grade 3–4 CRS events.
Image:
Summary/Conclusion: Our results show that IV M mono retreatment was efficacious in
heavily pretreated pts with R/R NHL who initially achieved a CR with M treatment,
but subsequently developed PD. M retreatment had a manageable safety profile, consistent
with the initial treatment.
P1125: CELESTIMO: A PHASE III TRIAL EVALUATING THE EFFICACY AND SAFETY OF MOSUNETUZUMAB
PLUS LENALIDOMIDE VERSUS RITUXIMAB PLUS LENALIDOMIDE IN PATIENTS WITH RELAPSED OR
REFRACTORY FOLLICULAR LYMPHOMA
L. Nastoupil1,*, F. Morschhauser2, C. W. Scholz3, M. Bishton4, S.-S. Yoon5, P. Giri6,
M. C. Wei7, A. Knapp8, C.-C. Li7, A. Bottos8, H. Li9, E. Purev7, N. L. Bartlett10
1Emory University, Atlanta, GA, United States of America; 2University of Lille, CHU
Lille, Lille, France; 3Vivantes Klinikum am Urban, Berlin, Germany; 4Nottingham University
Hospitals NHS Trust, Nottingham, United Kingdom; 5Seoul National University Hospital,
Seoul, South Korea; 6Royal Adelaide Hospital, Adelaide, Australia; 7Genentech, Inc.,
South San Francisco, CA, United States of America; 8F. Hoffmann-La Roche Ltd, Basel,
Switzerland; 9Hoffmann-La Roche Ltd, Mississauga, ON, Canada; 10Washington University
School of Medicine, St. Louis, United States of America
Background: Despite significant progress with first-line immunochemotherapy, most
patients with follicular lymphoma (FL) will eventually relapse with increasing refractoriness
and decreasing duration of response to subsequent therapy lines (Rivas-Delgado et
al. 2019). Mosunetuzumab (M) is a CD20xCD3 bispecific antibody that engages and redirects
T-cells to eliminate malignant B cells (Sun et al. 2015). In an ongoing, pivotal Phase
I/II trial of M monotherapy, patients with relapsed/refractory (R/R) FL who have received
≥2 prior treatment lines achieve deep and durable responses (NCT02500407; Budde et
al. ASH 2021). Preliminary data from a Phase Ib study have suggested favorable safety
and promising activity of M in combination with lenalidomide (Len), a potent immunomodulatory
agent that has shown additive/synergistic activity with an anti-CD20 antibody in R/R
indolent lymphoma (Leonard et al. 2019), in patients with R/R FL who have received
≥1 prior therapy (NCT4246086; Morschhauser et al. ASH 2021). The chemotherapy-free
M-Len combination may represent a promising outpatient therapy option for future management
of patients with R/R FL. The randomized, multicenter Phase III study has been initiated.
Aims: CELESTIMO (NCT04712097) is a randomized, multicenter, open-label Phase III study
evaluating the efficacy and safety of M-Len versus rituximab plus Len (R-Len) in patients
with previously treated R/R FL.
Methods: Patients must have histologically documented CD20+ FL (Grades 1–3a) requiring
systemic therapy and have received ≥1 prior line of systemic therapy. Patients are
randomized (1:1) to receive M-Len (M intravenously [IV] on Days [D] 1, 8 and 15 of
Cycle [C] 1 [21-day cycle] and D1 of C2–12 [28-day cycles], plus Len orally [PO] on
D1–21 of C2–12) or R-Len (R IV on D1, 8, 15 and 22 of C1 then on D1 of C3, 5, 7, 9,
and 11, plus Len PO on D1–21 of C1–12 [all 28-day cycles]), and stratified by disease
progression within 24 months of initial treatment (yes/no), number of prior lines
of therapy (1 versus ≥2), and refractoriness to anti-CD20 therapy (refractory/non-refractory).
All patients must provide informed consent.
Results: The primary endpoint is progression-free survival (PFS) assessed by independent
review committee; secondary endpoints include investigator-assessed PFS, complete
and objective response, overall survival, and safety. Biomarkers predictive of response
to M-Len and R-Len will also be investigated as exploratory endpoints.
Summary/Conclusion: The study started recruitment in 2021 and plans to enroll ~400
patients from approximately 16 countries and 150 sites globally. Further study details
will be presented.
P1126: MOSUNETUZUMAB IS EFFICACIOUS AND WELL TOLERATED IN PATIENTS AGED <65 AND ≥65
YEARS WITH RELAPSED/REFRACTORY FOLLICULAR LYMPHOMA AND ≥2 PRIOR THERAPIES: SUBGROUP
ANALYSIS OF A PIVOTAL PHASE II STUDY
M. Matasar1,*, N. L. Bartlett2, L. H. Sehn3, S. J. Schuster4, S. Assouline5, P. Giri6,
J. Kuruvilla7, M. Canales8, S. Dietrich9, K. Fay10, M. Ku11, L. J. Nastoupil12, M.
C. Wei13, S. Yin13, I. To13, D. Turner13, H. Huang14, J. Min15, E. Penuel13, L. E.
Budde16
1Memorial Sloan Kettering Cancer Center, New York, NY; 2Siteman Cancer Center, Washington
University School of Medicine, St. Louis, MO, United States of America; 3BC Cancer
Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada;
4Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia,
PA, United States of America; 5Jewish General Hospital, Montreal, QC, Canada; 6Royal
Adelaide Hospital, Adelaide, Australia; 7Princess Margaret Cancer Centre, Toronto,
ON, Canada; 8Hospital Universitario La Paz, Madrid, Spain; 9Universitat Heidelberg,
Heidelberg, Germany; 10St Vincent’s Hospital and Royal North Shore Hospital, Sydney;
11St Vincent’s Hospital, University of Melbourne, Melbourne, Australia; 12MD Anderson
Cancer Center, Houston, TX; 13Genentech, Inc., South San Francisco, CA, United States
of America; 14Hoffmann-La Roche Ltd, Mississauga, ON, Canada; 15Roche Products Ltd,
Welwyn Garden City, United Kingdom; 16City of Hope, Duarte, CA, United States of America
Background: Advancing age is associated with a decline in immune function and an increase
in the incidence of comorbidity (Castellino et al. 2017). Vulnerability to treatment-related
toxicities may also increase with age. Mosunetuzumab is a T-cell engaging CD20xCD3
bispecific monoclonal antibody (Ab) that redirects T cells to eliminate malignant
B cells. In a pivotal Phase II study (NCT02500407), mosunetuzumab induced deep and
durable remissions and had a favorable safety profile in patients (pts) with relapsed/refractory
(R/R) follicular lymphoma (FL) and ≥2 prior therapies (Budde et al. ASH 2021).
Aims: In the current study, we evaluated the efficacy and safety of mosunetuzumab
in pts aged <65 and ≥65 years (age stratification per ESMO Clinical Practice Guidelines
for management of newly diagnosed and R/R FL; Dreyling et al. 2021) in the pivotal
Phase II study.
Methods: All pts had Grade (Gr) 1–3a FL and ECOG performance status (PS) 0–1, and
were R/R to ≥2 prior therapies, including an anti-CD20 Ab and an alkylating agent.
Intravenous mosunetuzumab was given in 21-day cycles with Cycle (C) 1 step-up dosing:
C1 Day (D) 1: 1mg; C1D8 2mg; C1D15 and C2D1: 60mg; D1 of C3+: 30mg. Pts with a complete
response (CR) by C8 completed therapy; pts with a partial response or stable disease
continued treatment for up to 17 cycles, unless disease progression or unacceptable
toxicity occurred. The primary endpoint was CR (as best response) rate by PET/CT assessed
by Independent Review Committee using standard response criteria (Cheson et al. 2007).
Cytokine release syndrome (CRS) was graded using ASTCT criteria (Lee et al. 2019).
All pts provided informed consent.
Results: As of August 27, 2021, 90 pts had received mosunetuzumab; 67% were aged <65
years and 33% were aged ≥65 years. Of 30 pts aged ≥65 years, median age was 71 years
(range: 65–90) and 43% had ECOG PS 1 at entry, 53% had FLIPI 3–5, and 70% had Ann
Arbor stage III/IV disease. Median number of prior therapies in the older age group
was 3 (range: 2–8); 77% were refractory to a prior anti-CD20 Ab and 43% were double-refractory
to a prior anti-CD20 Ab and an alkylating agent. Compared with younger pts, those
aged ≥65 years had a numerically higher objective response rate (87% [95% CI: 69–96]
vs 77% [95% CI: 64–87]) and CR rate (70% [95% CI: 51–85] vs 55% [95% CI: 42–68]).
The 18-month event-free rate for duration of response was 54% (95% CI: 31–76) in pts
aged ≥65 years and 59% (95% CI: 43–74) in those aged <65 years. The rate of Gr 3–4
adverse events (AEs) was comparable in the older and younger age groups (73% vs 68%,
respectively). A lower rate of serious AEs of any Gr was observed in pts aged ≥65
years (37% vs 52%). One patient in each age group discontinued treatment due to mosunetuzumab-related
AEs. CRS occurred less frequently in pts aged ≥65 years than in those aged <65 years
(30% vs 52%), while CRS events were predominantly low-Gr in both groups (age ≥65 vs
<65 years: Gr 1, 20% vs 28%; Gr 2, 7% vs 22%); high-Gr CRS was uncommon (one patient
in each group). All CRS events resolved. No cases of aphasia, seizures, encephalopathy
or cerebral edema occurred. Rates of serious AEs of infection (any Gr) were comparable
in pts aged ≥65 and <65 years (17% vs 22%, respectively). The PK disposition of mosunetuzumab
was also comparable in pts aged ≥65 and <65 years.
Summary/Conclusion: Mosunetuzumab is efficacious and well tolerated in older and younger
pts with R/R FL and ≥2 prior therapies. In older pts (aged ≥65 years), numerically
lower rates of CRS and serious AEs were observed.
P1127: WATCHFUL WAITING IS AN ACCEPTABLE TREATMENT OPTION FOR PRIMARY OCULAR ADNEXAL
MUCOSA-ASSOCIATED LYMPHOID TISSUE LYMPHOMA: A RETROSPECTIVE STUDY
K. Mizuhara1,*, T. Kobayashi1, M. Nakao2, R. Takahashi3, H. Kaneko4, K. Shimura4,
K. Hirakawa5, N. Uoshima6, Y. Kamitsuji6, K. Wada7, E. Kawata7, R. Isa1, T. Fujino1,
Y. Matsumura-Kimoto1, T. Tsukamoto1, S. Mizutani1, Y. Shimura1, M. Taniwaki8, J. Kuroda1
1Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University
of Medicine, Kyoto; 2Department of Hematology, Otsu City Hospital, Otsu; 3Department
of Hematology, Omihachiman Community Medical Center, Omihachiman; 4Department of Hematology,
Aiseikai Yamashina Hospital, Kyoto; 5Department of Hematology, Fukuchiyama City Hospital,
Fukuchiyama; 6Department of Hematology, Japanise Red Cross Kyoto Daini Hospital, Kyoto;
7Department of Hematology, Matsushita Memorial Hospital, Moriguchi; 8Center for Molecular
and Therapeutics, Kyoto Prefectural University of Medicine, Kyoto, Japan
Background: Primary ocular adnexal mucosa-associated lymphoid tissue (MALT) lymphoma
(POAML) is the most frequent subtype of ocular adnexal lymphomas. Radiotherapy (RT)
is the standard of care for localized POAML, despite several complications, such as
cataracts, retinitis, dry eye, and optic neuropathy. Watchful waiting (WW) is expected
to be a therapeutic option for POAML, however, its efficacy for POAML remains to be
verified.
Aims: The aim of the study was to clarify the validity of WW for POAML.
Methods: We retrospectively analyzed the background, disease status, treatments, and
clinical outcomes of patients histologically diagnosed with POAML between 2008 and
2019 at seven institutes belonging to the Kyoto Clinical Hematology Study Group. The
primary ocular adnexal disease sites were defined as involvement of the conjunctiva,
orbit, eyelid, or lacrimal gland. Bilateral ocular adnexal lymphoma without other
lymphomatous involvement was defined as stage IE disease.
Results: One hundred and three patients were diagnosed with POAML, of which 11 were
excluded from analysis because of lost follow-up, 11 due to missing data, and 6 due
to short follow-up within 1 year. As a result, 75 patients were analyzed in this study.
The median age was 68 years, and 41 patients (54.7%) were females. The most common
involved site was the conjunctiva (42.7%), followed by the orbit (36.0%), lacrimal
gland (12.0%), and eyelid (8.0%). Sixty-nine patients (92.0%) presented with stage
IE disease, and 61 patients (81.3%) presented with a unilateral ocular region. According
to the International Prognostic Index, most patients (92.0%) were classified as low
risk. The tumor was completely resected at diagnosis in 17 patients (22.7%), while
a residual tumor existed after biopsy in 58 patients (77.0%). WW was the most frequent
first-line modality, selected for 29 patients (38.7%), including 14 patients with
complete tumor resection at diagnosis. Twenty-four patients (32.0%) were treated with
RT and 19 (25.3%) with rituximab monotherapy (R). In the RT group, most patients had
a residual tumor after the biopsy, and the orbit was the most frequent site of lymphoma,
which caused treatment-emergent symptoms that potentially impair visual function.
Complete response rates were 79.2% and 42.1% in the RT and R groups, respectively.
With a median follow-up of 48.8 months, there were no significant differences in the
time to start of new treatment between WW and RT groups (median: both not reached
[NR], p = 0.187) and between WW and R groups (median: NR vs. 69.0 months, p = 0.554).
At 60 months follow-up, the estimated proportions of POAML patients not requiring
new treatment were 69.4%, 85.2%, and 53.8% in the WW, RT, and R groups, respectively.
All patients enrolled in this study were alive at the time of analysis. In the WW
group, 9 patients (31.0%) showed disease progression, seven of whom received treatments
with a median time to next treatment of 15.6 months, but the remaining two patients
continued WW even after disease progression because of no subjective symptoms. In
the RT group, 16 patients (66.7%) developed adverse events, most of which were mild,
while grade 3 cataracts occurred in five patients. We compared the clinical characteristics
to identify predictive factors for disease progression in the WW group, however, all
factors analyzed were not significantly associated with disease progression.
Summary/Conclusion: WW may be an acceptable treatment option for POAML, especially
in asymptomatic patients.
P1128: CLINICAL FEATURES AND OUTCOME OF PATIENTS WITH CASTLEMAN DISEASE: A SPANISH
MULTICENTRIC STUDY OF 134 PATIENTS FROM GELTAMO
J. T. Navarro1,2,3,*, C. Celades2,3, O. García1,2,4,5, E. González-Barca6,7,8, F.
Climent7,9, A. Feu9, A. Jiménez10, A. Gutiérrez de la Peña10, M. Bastos-Oreiro11,12,
T. Aldamiz-Echevarria13, A. Gutiérrez14,15, L. Bento14,15, P. Abrisqueta16,17, C.
M. Alonso18, C. Tejada Chavez18, E. M. Ocio19,20, N. Fernández Escalada19,20, M. B.
Navarro Matilla21, J. M. Mateos Pérez21, A. López-García22, C. Castillo-Girón23, S.
F. Pinzón24, E. Pérez Ceballos25, J. Á. Hernández Rivas26,27, R. del Campo García15,28,
E. Pardal de la Mano29, R. García-Sanz30,31,32,33, J. Rovira34, J. M. Sancho1,2,3,
G. Tapia35,36
1Hematology, Institut Català d’Oncologia-Germans Trias i Pujol Hospital; 2Lymphoid
Neoplasms, Josep Carreras Leukaemia Research Institute; 3Medicine, Universitat Autònoma
de Barcelona; 4Institut Germans Trias i Pujol; 5Universitat Autònoma de Barcelona,
Badalona; 6Hematology, Institut Català d’Oncologia, Hospitalet de Llobregat; 7IDIBELL;
8Universitat de Barcelona, Hospitalet; 9Pathology, Hospital Universitari de Bellvitge,
Hospitalet de Llobregat; 10Hematology, Hospital 12 de Octubre; 11Hematology, Hospital
Gregorio Marañón; 12Instituto de Investigación Sanitaria Gregorio Marañón; 13Infectious
Diseases-Clinical Microbiology, Hospital Gregorio Marañón, Madrid; 14Hematology, Son
Espases University Hospital; 15IdISBa, Palma de Mallorca; 16Hematology, Hospital Universitari
Vall d’Hebron; 17Vall d’Hebron Institute of Oncology (VHIO), Barcelona; 18Hematology
and Hemotherapy, Hospital Arnau de Vilanova, Valencia; 19Hematology, Hospital Universitario
Marqués de Valdecilla (IDIVAL); 20Universidad de Cantabria, Santander; 21Hematology,
Hospital Universitario Puerta de Hierro Majadahonda; 22Hematology, Hospital Universitario
Fundación Jiménez Diaz, Madrid; 23Amyloidosis and Myeloma, Hospital Clinic; 24Hematology,
Hospital del Mar, Barcelona; 25Hematology, Hospital Universitario Morales Meseguer,
Murcia; 26Hematology and Hemotherapy, Hospital Universitario Infanta Leonor; 27Universidad
Complutense de Madrid, Madrid; 28Hematology, Hospital Universitari Son Llàtzer, Palma
de Mallorca; 29Hematology and Hemotherapy, Hospital Virgen del Puerto, Plasencia;
30Hematology, University Hospital of Salamanca & Cancer Research Center (CiC-IBMCC,
CSIC/USAL); 31Center for Biomedical Research in Network of Cancer (CIBERONC); 32Institute
of Biomedical Research of Salamanca (IBSAL); 33Universidad de Salamanca, Salamanca;
34Hematology, Hospital Joan XXIII-ICO Tarragona, Tarragona; 35Pathology, Hospital
Germans Trias i Pujol, Badalona; 36Medicine, Universitat Autònoma de Barcelona, Barcelona,
Spain
Background: Castleman disease (CD) is a heterogeneous rare disorder, characterized
by lymph node enlargement with a common histopathological spectrum. This disease comprises
the subtypes unicentric (UCD), HHV-8 multicentric CD (HHV-8 MCD), POEMS-asociated
MCD (POEMS MCD) and idiopathic multicentric CD (iMCD).2 Whereas UCD is a localized
reversible disease, the other three are systemic diseases with multiple lymphadenopathies.
The TAFRO (thrombocytopenia, anasarca, fever, reticulin myelofybrosis, organomegaly)
syndrome is an aggressive form of iMCD. Each CD subtype has different clinical features,
prognosis and treatment.
Aims: This study aims to study a large series of patients diagnosed with CD to describe
the clinical and biological characteristics, as well as the outcome, of the different
CD subtypes.
Methods: Multicentric retrospective study which includes patients diagnosed with different
subtypes of CD in 19 Spanish hospitals from 2006 to 2020. Clinical and biological
data were retrieved from the clinical records. The project has been approved by the
Ethical Committee of Germans Trias I Pujol Hospital (PI-20-103). Statistical analyses
were performed using SPSS v24.0 (IBM, Somer, NY).
Results: One hundred and thirty-four patients with available data were included; 47
with UCD and 87 with any of the 3 subtypes of MCD. The median follow-up was 3.4 years.
The main clinical and biological characteristics of the 4 CD subtypes are shown in
Table 1 and differences in OS in figure 1. Most patients with HHV-8 MCD were HIV-infected
(73%); median CD4 lymphocyte count 0.229x109/L (range: 0.016, 1.2) and 46% of them
had concomitant or prior Kaposi’s sarcoma. Three iMCD patients had TAFRO. All patients
with UCD were treated with surgery. Most patients with HHV-8 MCD were treated at front-line
with rituximab (69%) or polychemotherapy plus rituximab (21%). Patients with iMCD
were treated with anti-IL-6 (25%), polychemotherapy (25%), rituximab (25%) and steroids
(25%). Treatment of POEMS MCD was also diverse; 3 patients received lenalidomide plus
dexamethasone, 2 polychemotherapy, 1 anti-IL-6 and 1 auto SCT (3 patients unrecorded).
Eleven patients (8.2%) had concomitant or evolved to lymphoma (2 UCD, 2 iMCD and 7
HHV-8 MCD). Thirty-two patients are dead (20 HHV8-MCD, 7 iMCD, 2 UCD, 3 POEMS), 6
of them from CD (4 HHV8-MCD, 2 iMCD).
Table 1.
Main clinical and biological characteristics of the CD subtypes
Variable
UCD (n=47)
iMCD (n=24)
HHV8-MCD (n=52)
POEMS (n=11)
TOTAL (n=134)
p value
Male gender, %
36
46
90
36
59
<0.001
Age, median (range)
40 (5, 84)
47 (10, 82)
47 (25, 88)
64 (30, 78)
47 (5, 88)
0.154
ECOG<2, n (%)
36/38 (95)
13/19 (68)
24/39 (62)
5/8 (63)
78/104 (75)
0.005
Bulky mass, n (%)
3/41 (7)
0/19
1/41 (2)
0/9
4/110 (4)
0.422
B symptoms, n (%)
4/41 (10)
6/17 (35)
31/41 (76)
3/9 (33)
44/108 (41)
<0.001
Hemoglobin g/L, median (range)
135 (74, 990)
132 (40, 355)
106 (43, 332)
144 (83, 172)
126 (40, 990)
<0.001
Leukocytes x10
9
/L, median (range)
7.1 (3.8, 21.1)
7.2 (2, 13.2)
6.3 (0.5, 23.7)
7.9 (2.4, 23)
6.7 (0.5, 23.7)
0.075
Platelets x10
9
/L, median (range)
247(67, 483)
241 (13.5, 501)
190.5 (3.5, 461)
467 (1.4, 560)
232 (1.4, 560)
0.05
High LDH, n (%)
2/35 (6)
7/16 (44)
6/41 (15)
0/9
15/101 (15)
0.002
High Ferritin, n (%)
1/24 (4)
2/9 (22)
19/29 (66)
0/6
22/68 (32)
<0.001
High Beta-2 microglobulin, n (%)
1/15 (7)
4/11 (36)
20/26 (77)
4/5 (80)
29/57 (51)
<0.001
Low albumin, n (%)
3/35 (9)
6/14 (43)
19/39 (49)
2/8 (25)
30/96 (31)
0.002
Image:
Summary/Conclusion: Castleman Disease subtypes have different characteristics and
outcomes. First-line treatment is heterogeneous in MCD subtypes pointing a need for
national guidelines.
P1129: ACALABRUTINIB IN PATIENTS WITH RELAPSED/REFRACTORY (R/R) MARGINAL ZONE LYMPHOMA
(MZL): RESULTS OF A PHASE 2, MULTICENTER, OPEN-LABEL TRIAL
P. Strati1,*, M. Coleman2, D. Stevens3, S. Ma4, C. Patti5, M. Levy6, I. S. Lossos7,
P. Ramakrishnan Geethakumari8, S. Lam9, R. Calvo10, K. Higgins11, L. Budde12
1The University of Texas MD Anderson Cancer Center, Houston, TX; 2Clinical Research
Alliance/Weill Cornell Medicine, New York City, NY; 3Norton Cancer Institute, Louisville,
KY; 4Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg
School of Medicine, Chicago, IL, United States of America; 5A.O.O.R. Villa Sofia-Cervello,
U. O. Ematologia I, Palermo, Italy; 6Baylor University Medical Center, Dallas, TX;
7Sylvester Comprehensive Cancer Center, University of Miami−Miller School of Medicine,
Miami, FL; 8Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern
Medical Cente, Dallas, TX, United States of America; 9London Health Sciences Centre,
London, Ontario, Canada; 10AstraZeneca, Gaithersburg, MD; 11AstraZeneca, South San
Francisco, CA; 12City Of Hope National Medical Center, Duarte, CA, United States of
America
Background: MZL is a rare indolent B-cell malignancy considered incurable at recurrent
stage. Bruton tyrosine kinase (BTK) inhibitors have produced durable responses in
patients (pts) with R/R MZL. Acalabrutinib (acala) is a potent next-generation BTK
inhibitor with high selectivity for BTK.
Aims: We report data for acala monotherapy from the R/R MZL cohort (phase 2) of a
phase 1b/2 clinical trial (NCT02180711).
Methods: Pts with histologically confirmed MZL, ECOG performance status ≤2, and ≥1
prior therapy (including ≥1 CD20-directed regimen) received oral acala 100 mg twice
daily until disease progression or unacceptable toxicity ± R. The primary objective
was overall response rate (ORR; Lugano criteria as assessed by the investigator).
Secondary objectives were duration of response (DOR), progression-free survival (PFS),
overall survival (OS), and safety. Data were analyzed descriptively (no formal hypothesis
testing).
Results: Forty-two pts received acala (median age 69 y [range 42–84]; median 2 prior
systemic regimens [range 1–4]). MZL subtypes were extranodal (43%), nodal (31%), and
splenic (26%). At data cutoff (Oct 15, 2021), median follow-up duration was 10.7 mo
(range 0.4–42.8). Sixteen (38%) pts discontinued acala, most commonly due to disease
progression (26%). Among pts evaluable for response (n=37; 3 pts had not reached the
first assessment timepoint and 2 pts exited the study without response assessment),
ORR was 54% (95% CI 37%–71%) with 6 complete (16%) and 14 partial (38%) responses;
17 (46%) pts had stable disease. ORRs in extranodal, nodal, and splenic subtypes were
65%, 44%, and 45%, respectively. Median time to initial response was 3.0 mo; median
DOR was 19.3 mo (95% CI 8.4–not estimable). Median PFS was 27.4 mo with a 12-mo PFS
rate of 66%. Four pts died (disease progression, n=2; transformation to diffuse large
B-cell lymphoma after stopping treatment, n=1; adverse event [AE], n=1 [septic shock
unrelated to treatment]); median OS was not reached. Treatment was well tolerated
with most AEs being grade 1 or 2. Sixteen pts (38%) had grade ≥3 AEs, most commonly
(in ≥2 pts) anemia, dyspnea, neutrophil count decrease (n=3 each), fatigue, thrombocytopenia,
and neutropenia (n=2 each). AEs led to treatment discontinuation in 2 pts (grade 3
hypotension and grade 1 myalgia). Among AEs of clinical interest, hypertension was
reported in 2 pts (both grade 2); no cases of atrial fibrillation/flutter or major
hemorrhage were reported.
Summary/Conclusion: These early results indicate that acala is efficacious and well
tolerated in pts with R/R MZL. The AE profile is consistent with the known safety
profile of acala.
P1130: ACALABRUTINIB IN TREATMENT-NAIVE OR RELAPSED/REFRACTORY WALDENSTRÖM MACROGLOBULINEMIA:
5-YEAR FOLLOW-UP OF A PHASE 2, SINGLE-ARM STUDY
R. Owen1,*, H. McCarthy2, S. Rule3,4, S. D’Sa5, S. Thomas6, O. Tournilhac7, F. Forconi7,
M. José Kersten8, P. Luigi Zinzani9, S. Iyengar10, J. Kothari11, M. Minnema12, E.
Kastritis13, B. Cheson14, H. Walter15, D. Greewald16, R. Calvo17, C.-C. Wun18, R.
Furman19
1St. James’s University Hospital, Leeds; 2Royal Bournemouth Hospital, Bournemouth;
3Plymouth University Medical School, Plymouth, United Kingdom; 4AstraZeneca, Mississauga,
Ontario, Canada; 5University College London Hospitals NHS Trust, London, United Kingdom;
6MD Anderson Cancer Center, Houston, TX, United States of America; 7University of
Southampton Hospital Trust, Southampton, United Kingdom; 8Amsterdam University Medical
Centers, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, Netherlands;
9Institute of Hematology University of Bologna, Bologna, Italy; 10Royal Marsden Hospital,
London; 11Churchill Hospital, Oxford, United Kingdom; 12University Medical Center
Utrecht, University Utrecht, Utrecht, The Netherlands; on behalf of the Lunenburg
Lymphoma Phase I/II Consortium – HOVON/LLPC, Utrecht, Netherlands; 13National and
Kapodistrian University of Athens, Athens, Greece; 14Georgetown University Hospital,
Lombardi Comprehensive Cancer Center, Washington, DC, United States of America; 15Ernest
and Helen Scott Haematological Research Institute and Leicester Cancer Research Centre,
University of Leicester, Leicester, United Kingdom; 16Cancer Center of Santa Barbara,
Santa Barbara; 17AstraZeneca, Gaithersburg, MD; 18AstraZeneca, South San Francisco;
19Weill Cornell Medical College, New York Presbyterian Hospital, New York, NY, United
States of America
Background: Bruton tyrosine kinase (BTK) inhibitors are effective in the treatment
of Waldenström macroglobulinemia (WM). Acalabrutinib is a highly selective, potent,
covalent BTK inhibitor that demonstrated less toxicity and improved tolerability in
a head-to-head trial versus ibrutinib in chronic lymphocytic leukemia.
Aims: The aim of this trial was to determine the efficacy and safety of acalabrutinib
in patients with treatment-naive (TN) or relapsed/refractory (R/R) WM. Interim results
at 25 months (mo) of median follow-up have been previously reported. At a 5-year median
follow-up, we report the final results of this trial.
Methods: In this single-arm, multicenter, phase 2 trial, after informed consent was
obtained, patients with TN or R/R WM received 100 mg oral acalabrutinib twice daily
(or 200 mg acalabrutinib once daily, later switched to 100 mg twice daily). The primary
endpoint was investigator-assessed overall response rate (ORR). Key secondary endpoints
included duration of response (DOR), progression-free survival (PFS), overall survival
(OS), and safety. Efficacy parameters were based on the modified 3rd International
Workshop on Waldenström Macroglobulinemia (IWWM) criteria (Kimby 2006). Mutation data
were available for a minority of patients and therefore are not reported.
Results: Of 106 patients treated with acalabrutinib, 14 were TN and 92 were R/R; median
age was 69 years (range 39–90); 94% had ECOG PS ≤1; median serum IgM was 3615 mg/dL
(range 291–9740). Patients with R/R disease had a median of 2 prior therapies (range
1–7). ORR was 93% (95% CI: 66, 100) in the TN cohort and 95% (88, 98) in the R/R cohort;
major response rates (≥partial response) were 79% (49, 95) and 82% (72, 89), respectively.
Median DOR was not estimable (NE) (12, NE) in the TN cohort and 65 mo (55, NE) in
the R/R cohort; the estimated 66-mo DOR was 90% (TN; 47, 99) and 45% (R/R; 27, 61).
At a 63.7-mo median follow-up, median PFS rate was NE (TN; 19, NE) and 68 mo (R/R;
53, NE); estimated 66-mo PFS rate was 84% (TN; 48, 96) and 52% (R/R; 39, 63). Median
OS rate was NE for the TN and R/R cohorts; estimated 66-mo OS rate was 91% (TN; 51,
99) and 71% (R/R; 60, 80). Of the patients who remained on treatment, 7 (50%) were
in the TN cohort and 43 (47%) were in the R/R cohort. The main reasons for discontinuing
acalabrutinib were adverse events (AEs) (4 [29%] patients in the TN cohort, 15 [16%]
patients in the R/R cohort) and progressive disease (PD) (1 [7%] patient in the TN
cohort, 20 [22%] patients in the R/R cohort). The most common AEs and selected events
of clinical interest are listed in the Table. There were 12 grade 5 AEs (11 and 1
patients in the R/R and TN cohorts, respectively); 1 (intracranial hematoma [R/R])
was considered treatment-related.
Image:
Summary/Conclusion: Acalabrutinib is a highly effective treatment that achieved durable
responses with a favorable safety profile in patients with TN or R/R WM.
P1131: POST HOC ANALYSIS OF PATIENTS WITH HIGHLY PROLIFERATIVE VARIANTS OF MANTLE
CELL LYMPHOMA TREATED WITH ACALABRUTINIB
S. Le Gouill1,*, M. Długosz-Danecka2, S. Rule3,4, P. Luigi Zinzani5, A. Goy6, S. D.
Smith7, J. K. Doorduijn8, C. Panizo9, B. Shah10, A. Davies11, R. Eek12, E. Jacobsen13,
A. P. Kater14, T. Robak15, P. Jain16, R. Calvo17, H. Yang18, M. Wang16
1Institut Curie, Paris, France; 2Maria Sklodowska-Curie National Research Institute
of Oncology, Krakow, Poland; 3Plymouth University Medical School, Plymouth, United
Kingdom; 4AstraZeneca, Mississauga, Ontario, Canada; 5IRCCS Azienda Ospedaliero-Universitaria
di Bologna Istituto di Ematologia “Seràgnoli,” Dipartimento di Medicina Specialistica,
Diagnostica e Sperimentale Università di Bologna, Bologna, Italy; 6John Theurer Cancer
Center, Hackensack University Medical Center, Hackensack, NJ; 7University of Washington,
Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America; 8Erasmus
MC Cancer Institute, HOVON Lunenburg Lymphoma Phase I/II Consortium, Rotterdam, Netherlands;
9Clínica Universidad de Navarra, Pamplona, Spain; 10Moffitt Cancer Center, Tampa,
FL, United States of America; 11Cancer Research UK Experimental Cancer Medicines Centre,
University of Southampton Faculty of Medicine, Southampton, United Kingdom; 12Border
Medical Oncology, Albury, NSW, Australia; 13Dana Farber Cancer Institute, Harvard
Medical School, Boston, MA, United States of America; 14Amsterdam University Medical
Center, Amsterdam, on behalf of HOVON, Amsterdam, Netherlands; 15Copernicus Memorial
Hospital, Medical University of Lodz, Lodz, Poland; 16MD Anderson Cancer Center, University
of Texas, Houston, TX; 17AstraZeneca, Gaithersburg, MD; 18AstraZeneca, South San Francisco,
CA, United States of America
Background: Blastoid and pleomorphic variants of mantle cell lymphoma (MCL) are considered
high risk and have poor prognoses. Patients with these variants rarely achieve durable
remission or prolonged clinical outcomes with currently available chemotherapies and
targeted therapies. Final results from ACE-LY-004, an open-label, multicenter, single-arm
phase 2 study of the Bruton tyrosine kinase (BTK) inhibitor acalabrutinib in adult
patients with relapsed/refractory (R/R) MCL confirmed acalabrutinib was highly active
in patients with R/R MCL, with a median overall survival of 59.2 months, a median
progression-free survival (PFS) of 22.0 months, and a median duration of response
(DOR) of 28.6 months after a median 38.1 months of follow-up. In a subgroup analysis,
we found that patients with 1 prior line of therapy had unexpectedly shorter DOR than
those who had ≥2 prior lines of therapy.
Aims: To investigate a possible reason for shorter DOR in patients with R/R MCL treated
with acalabrutinib who received only 1 prior line of therapy, we conducted a post
hoc analysis on a subgroup of patients with blastoid/pleomorphic disease.
Methods: ACE-LY-004 enrolled adult patients with confirmed R/R MCL and an Eastern
Cooperative Oncology Group performance status (ECOG PS) of ≤2. Informed consent was
obtained. Acalabrutinib 100 mg was administered orally twice daily until occurrence
of progressive disease or unacceptable toxicity.
Results: A total of 124 patients were enrolled; median age was 68 years and 80% of
patients were male. As of December 4, 2020, median follow-up was 38.1 months and median
treatment exposure was 17.5 months (range 0.1–65.3). Fifty-nine of the 124 patients
(48%) had 1 prior therapy. The remaining 65 patients (52%) had 2 prior therapies (n=37)
or ≥3 prior therapies (n=28). Median DOR was longer in patients with ≥2 prior lines
of therapy versus those with 1 prior therapy (33.8 and 25.6 months, respectively).
Similarly, median PFS was longer in patients who received acalabrutinib in later lines
of therapy than in those who received acalabrutinib as second-line therapy (24.8 and
19.2 months, respectively). Among the 59 patients with 1 prior therapy, 16 (27%) had
blastoid or pleomorphic disease and 43 (73%) did not, compared with 10 patients (15%)
with blastoid/pleomorphic disease and 55 (85%) who did not among the 65 patients with
≥2 prior therapies. Overall response rate (ORR) in those with 1 prior therapy was
88% (n=14; 95% CI 61.7–98.4) in those with blastoid/pleomorphic disease (complete
response [CR] in 50%, n=8) versus 81% (n=35; 95% CI 66.6–91.6) in those without blastoid/pleomorphic
disease (CR in 47%, n=20). Median DOR for these patients was 20.3 (95% CI 3.5–46.9)
and 31.8 (95% CI 14.9–not estimable) months, respectively.
Summary/Conclusion: In this post hoc analysis of the ACE-LY-004 study of acalabrutinib
in adult patients with confirmed R/R MCL, a shorter DOR was observed among patients
who received acalabrutinib after 1 prior therapy compared with those who received
acalabrutinib after ≥2 prior therapies, likely explained by a high proportion of patients
with blastoid/pleomorphic variants in the group of patients receiving acalabrutinib
after 1 prior therapy. While patients with blastoid/pleomorphic variant had comparable
ORR, DOR was shorter but still substantial at 20.3 months, indicating that some patients
with highly proliferative disease and poorer prognosis also achieve clinical benefit
from acalabrutinib.
P1132: PET INTERIM RESULTS PREDICT PROGRESSION-FREE-SURVIVAL IN FOLLICULAR LYMPHOMA
PATIENTS WITH ADVANCED STAGE
M. Poza1,*, I. Zamanillo1, R. Íñiguez1, P. Sarandeses2, A. Rodriguez1, T. Baumann1,
S. Barrio3, C. Grande1, A. Martin3, L. Rufian1, J. Martinez-Lopez1, A. Jiménez-Ubieto1
1Hematology; 2Department of Nuclear Medicine, Hospital 12 Octubre; 3Altum Sequencing
Co,., Madrid, Spain
Background: Follicular lymphoma (FL) is the second most common NHL. It is generally
associated with an indolent course, but a subgroup of patients has worse outcome,
lower progression-free-survival (PFS) and higher risk of histological transformation
(HT). Since Lugano classification, the Deauville criteria using the 5-point scale
(D5PS) is validated as the interpretation method for response assessment PET scans
in HL and NHL. Nevertheless, the prognostic value of D5PS in the interim evaluation
in FL (PETINT) is not well stablished. Additionally, the quantitative value of SUVmax
parameter in the response assessment is almost not studied.
Aims: The aim of this study is to evaluate the prognostic value of PETINT evaluation
and quantitative mid-induction SUVmax parameter in a cohort of FL patients followed
and treated in a single, academic centre with chemo-immunotherapy.
Methods: 179 consecutive FL patients were newly diagnosed between 2011 and 2020. 40
patients that did not require systemic treatment and 68 that did not have a PETINT
evaluation were excluded. As well, patients with HT (9) and FL grade 3B (4) were excluded.
From the 58 remaining patients, clinical, laboratory and PET examinations data was
recorded. All patients were Ann-Arbor stage III-IV and were treated with chemo-immunotherapy.
PET/CT examinations were acquired at baseline, mid-induction (after 4 cycles of treatment)
and end of treatment (EOT). PET positive was considered when Deauville score (DS)
was 4 or 5. Statistical analysis was performed with SPSS® using Cox regression, Kaplan-Meier
survival curves and log-rank test.
Results: The median age at diagnosis was 64 years (26-95) and 29 (50%) patients were
female. 33/58 (57%) patients had a high risk FLIPI and 18/55 (33%) had a high risk
PRIMA-PI. 37 (64%) patients were treated with R-CHOP and 21 with R-bendamustine. 44
(76%) patients received maintenance with rituximab.
After a median follow-up of 43 months, 20 (34%) patients had progression. On PETINT,
19 (32%) patients were PET(+), and 10 of them remained PETEOT(+). Only one patient
with PETINT(-), became PETEOT(+). PETINT(+) patients had a median PFS of 31 months
compared with a median PFS of 88 months in PETINT(-) (p < 0.001), Figure 1. 38.2%
PETINT(+) presented progression of disease in less than 24 months (POD24) vs 5.2%
on those who were in PETINT(-), with a p<0.001. PETINT(-) presented a negative predictive
value of 95%. No differences in OS was observed.
3/19 (16%) patients with PETINT(+) presented HT vs 1/39 (2%) with PETINT(-). Transformation-free
survival was lower in patients with PETINT(+), with a trend towards significance (p=
0.057).
Similar PFS results were obtained independently of prognostic index, treatment or
maintenance received.
Analysing the prognostic value of quantitative SUVmax, patients with SUVmax< 3,8 (28/58;
48%) presented better outcomes, with larger PFS (p < 0.001) and less HT (p= 0.004).
Figure 2.
Additionally, PET/CT findings at EOT were predictive of relapse, with a median PFS
of 30 months in PETEOF (+) vs 88 months in PETEOT(-) (p = 0.005). PETEOT (+) presented
a higher risk of HT (p= 0.004). Figure 3.
Image:
Summary/Conclusion: This study suggests that response assessment by PET at mid-induction
is predictive of relapse in advanced stage FL patients treated with 1st line chemo-immunotherapy.
With previous conflicting results, further studies are needed to confirm this preliminary
data. Probably, new laboratory tests such as MRD monitored by liquid biopsy could
improve the detection of this subgroup of high risk patients and help to develop a
response-adapted precision therapy.
P1133: SUB-CUTANEOUS RITUXIMAB INDUCTION FOLLOWED BY SHORT RITUXIMAB MAINTENANCE IMPROVES
PFS IN PATIENTS WITH LOW-TUMOR BURDEN FOLLICULAR LYMPHOMA. FINAL RESULTS OF FLIRT
PHASE III TRIAL, A LYSA STUDY.
G. Cartron, MD, PhD, E. Bachy, MD, PhD, H. Tilly, MD3, N. Daguindau, MD4, G.-M. Pica,
MD5, F. Bijou, MD6, C. Mounier, MD7, A. M. Clavert, MD8, G. L. Damaj, MD, PhD, B.
Slama, MD10, O. Casasnovas, MD11, R. Houot, MD, PhD, K. Bouabdallah, MD13, D. Sibon,
MD, PhD, O. Fitoussi, MD15, N. Morineau, MD16, C. Herbaux, MD, PhD, T. Gastinne, MD18,
L.-M. Fornecker, MD19, C. Haioun, MD, PhD, V. Launay, MD21, C. Araujo, MD22, O. Benbrahim,
MD23, L. Sanhes, MD24, R. Gressin, MD25, H. Gonzalez, MD26, F. Morschhauser, MD, PhD,
L. Xerri, MD, PhD, K. Tarte, PhD12, D. Pranger, MD29
1Haematology, CHU de Montpellier, MONTPELLIER Cedex 5; 2CHU Lyon Sud, Pierre Benite;
3Centre Henri Becquerel, Rouen; 4Centre Hospitalier Annecy-Genevois - Site d’Annecy,
Pringy Cedex; 5CH de Chambéry, Chambéry; 6Institut Bergonié, Bordeaux; 7Institut de
Cancérologie Lucien Neuwirth, St-Priest-en-Jarez; 8CHU Angers, Angers; 9CHU Côte de
Nacre, Caen; 10CH d’Avignon - Hôpital Henri Duffaut, Avignon; 11CHU Dijon - Hôpital
d’Enfants, Dijon; 12CHU Rennes, Rennes; 13CHU Bordeaux, Bordeaux; 14Hôpital Necker,
Paris; 15Polyclinique Bordeaux Nord Aquitaine, Bordeaux; 16CH Départemental Vendée,
La Roche sur Yon; 17CHU Montpellier, Montpellier; 18CHU de Nantes - Hôtel Dieu, Nantes;
19Hôpitaux Universitaires de Strasbourg - Hôpital de Hautepierre, Strasbourg; 20Hôpital
Henri Mondor, Créteil; 21CH de Saint-Brieuc - Hôpital Yves Le Foll, Saint- Brieuc;
22Centre Hospitalier de la Côte Basque - Hôpital de Bayonne, Bayonne; 23CHR de la
Source, Orléans; 24Hôpital Saint Jean, Perpignan; 25CHU de Grenoble - Hôpital Albert
Michallon, Grenoble; 26CH René Dubos, Pontoise; 27CHRU de Lille - Hôpital Claude Hurriez,
Lille; 28Departement de bio-pathologie, Institut Paoli Calmette, Marseille, France;
29GHdC, Charleroi, Belgium
Background: In low–tumor burden follicular lymphoma (FL), maintenance rituximab (MR)
has been shown to improve progression-free survival (PFS) when compared with observation.
RESORT study (Kahl et al. 2014) clearly showed that Rituximab (R) retreatment strategy
provided similar time to treatment failure that maintenance strategy with less rituximab
use.
Aims: It is not known whether short MR using sub-cutaneous (sc) route could improve
PFS while reducing R infusions.
Methods: Patients with the diagnosis of low-tumor burden FL (GELF criteria) within
the last 4 months before signing informed consent were randomly assigned to either
Iv-Arm: 4-weekly iv infusions of R 375 mg/m2 or Sc-Arm: one iv infusion of R (D1,
375 mg/m2) followed by 3 sc infusions of R (1400 mg, on days 8, 15 and 22) followed
by Rsc maintenance on months (M) 3, 5, 7 and 9. The primary endpoint was PFS and secondary
endpoints included safety, response rates (M3, M12), duration of response (DOR), time
to next anti-lymphoma treatment (TTNTL) and overall survival (OS).
Results: A total of 202 patients were included, 102 in Iv-Arm and 100 in Sc-Arm and
constitute the intent to treat population. The median uses of R were 4 infusions (range:
1-4, Iv-Arm) and 8 infusions (range: 2-8, Sc-Arm). With a median follow-up of 50.2
months (95% CI: 48.3-54.5), 4‐year PFS was 41.2% (95% CI: 30.6%; 51.6%) in Iv-Arm
and 58.1% (95% CI: 47.5%; 67.4%) in Sc-Arm. Median PFS was then 36.1 months (95% IC:
23.9-52.6) in Iv-Arm and 73.8 months (95% CI: 39.4-NA) in Sc-Arm (Fig 1.) (HR: 0.58;
95% CI: 0.39-0.87; P = 0.0076). Patients with at least one AE grade ≥ 3 were 8 (7.8%)
and 12 (12.4%) in Iv-Arm and Sc-Arm, respectively. According to Cheson criteria, ORR
at M3 were: 83% and 80% including 38% and 29% of CR/CRu, in Iv-Arm and Sc-Arm, respectively.
According to Lugano criteria, 36.3% (Iv-Arm; 95% CI: 27.0%. 46.4%) and 59.0% (Sc-Arm;
95% CI: 48.7%; 68.7%) were in CMR at M12. The median DOR was 32.7 months (95% IC:
20.6-49.7) and 70.8 months (36.4-NR) (HR: 0.56; 95% IC: 0.37-0.84) in Iv-Arm and Sc-Arm,
respectively. 4-year TTNLT was 54% (95% CI: 42.9%; 63.8%) in Iv-Arm and 61.8% (95%
CI: 50.8.6%; 71.0%) in Sc-Arm (HR: 0.81, 95% IC: 0.53-1.24). 4-year TTNLT chemotherapy
was 60.8 % (95% CI: 49.6%; 70.3%) in Iv-Arm and 71.4% (95% CI: 60.7%; 79.8%) in Sc-arm
(HR: 0.69, 95% IC: 0.42-1.12) (Fig 2.). OS was not different according to treatment
arm, 4-year OS was 95.0% (95% CI: 88.5%; 97.9%) in Iv-Arm and 96.7% (95% CI: 89.9%;
98.9%) in Sc-Arm.
Summary/Conclusion: This phase III study met its primary endpoint and demonstrated
that Rsc induction followed by a short MRsc improves PFS of patients with low-tumor
burden. MRsc did not however improved TTNLT. R in low-tumor burden FL allowed to avoid
cytotoxic use in most patients 6 years after treatment initiation.
P1134: OUTCOMES FOR PATIENTS WITH MANTLE CELL LYMPHOMA POST-CBTK INHIBITOR THERAPY
IN THE UNITED STATES AND JAPAN: A STUDY OF TWO REAL-WORLD DATABASES
S. Rai1,*, L. Hess2, Y. Chen2, P. Abada2, H. Konig2, R. Walgren2, Y. Tanizawa3, Z.
Cai3, M. Tajimi3
1Kindai University Faculty of Medicine, Osakasayama, Japan; 2Eli Lilly and Company,
Indianapolis, United States of America; 3Eli Lilly Japan K.K, Kobe, Japan
Background: Patients with mantle cell lymphoma (MCL) have limited treatment options
following covalent Bruton’s tyrosine kinase inhibitor (cBTKi) therapy, with no standard
regimens defined.
Aims: The aim of this study was to investigate real-world treatment patterns and the
outcomes for patients with MCL following cBTKi treatment utilizing two separate datasets—one
from the US and one from Japan.
Methods: De-identified patient-level electronic medical record data from two real-world
databases (ConcertAI in the US and Medical Data Vision in Japan) were utilized for
this study. Eligible patients were ≥18 years old and were diagnosed with MCL between
January 2011 and October 2020 in the US or between December 2010 and July 2020 in
Japan. Data were available through 2020 to allow a minimum of 1 year of follow-up
for all patients. Patients were required to have completed treatment with at least
one cBTKi during the first through third lines of therapy. Time-to-event analyses
utilized the Kaplan-Meier method.
Results: 946 US patients (median age 72 years, male 75.1%) and 196 Japanese patients
(median age 78 years, male 73.5%) met the eligibility criteria. 352 (37.2%) patients
in the US and 103 (52.6%) in Japan received subsequent post-cBTKi therapy. The remaining
387 (40.9%) patients in the US and 93 (47.5%) patients in Japan did not receive further
treatment. In the US, immediate post-cBTKi regimens included rituximab (with or without
other chemotherapy agents, n=152, 43.2%) or additional cBTKi-based 146 (41.5%) or
B-cell lymphoma 2 inhibitor–based (n=30, 8.5%) therapy. In Japan, immediate post-cBTKi
regimens were nearly exclusively chemotherapy based (n=92, 89.3%), and only 11 (10.7%)
patients were retreated with cBTKi-containing regimens. Median time from the end of
cBTKi therapy to discontinuation of the immediate post-cBTKi therapy or death was
3.9 months (95% confidence interval [CI]: 3.3-4.6) in the US and 2.4 months (n=196,
95% CI: 1.7-3.0) in Japan. Among those who received post-cBTKi therapy, the duration
of therapy was 2.6 months (95% CI: 2.1-3.3) in the US and 1.8 months (n=103, 95% CI:
1.1-2.3) in Japan. Median overall survival (OS) from discontinuation of cBTKi therapy
was 10.3 months (95% CI: 8.0-13.0 in US patients and 7.1 months (n=196, 95% CI: 4.6-12.7)
in Japanese patients.
Summary/Conclusion: Patients with MCL previously treated with cBTKi experience very
poor outcomes, as measured by their duration of subsequent therapy and OS. Data were
generally consistent between the US and Japan. The development of new safe and effective
therapies after cBTKi is needed to address the growing unmet medical need in this
treatment setting.
P1135: SUBCUTANEOUS EPCORITAMAB IN COMBINATION WITH RITXUIMAB + LENALIDOMIDE IN RELAPSED
OR REFRACTORY FOLLICULAR LYMPHOMA: PHASE 1/2 TRIAL UPDATE
D. Belada1,*, S. Leppä2, B. E. Wahlin3, M. Nijland4, J. H. Christensen5, S. de Vos6,
H. Holte7, K. M. Linton8, A. Abbas9, L. Wang9, M. Dinh10, B. Elliott9, L. Falchi11
14th Department of Internal Medicine – Hematology, University Hospital and Faculty
of Medicine, Hradec Králové, Czechia; 2Helsinki University Hospital Comprehensive
Cancer Center and University of Helsinki, Helsinki, Finland; 3Karolinska Institutet,
Stockholm, Sweden; 4University Medical Center Groningen and University of Groningen,
Groningen, Netherlands; 5Odense University Hospital, Odense, Denmark; 6Ronald Reagan
University of California Los Angeles Medical Center, Los Angeles, United States of
America; 7Oslo University Hospital and KG Jebsen Center for B-cell Malignancies, Oslo,
Norway; 8The Christie NHS Foundation Trust and Manchester Cancer Research Centre,
Manchester, United Kingdom; 9Genmab, Princeton; 10AbbVie, North Chicago; 11Lymphoma
Service, Memorial Sloan Kettering Cancer Center, New York, United States of America
Background: Patients (pts) with relapsed or refractory (R/R) follicular lymphoma (FL)
experience less frequent and shorter responses with each line of treatment. Although
the rituximab + lenalidomide (R2) regimen is effective and has an acceptable safety
profile in R/R FL, FL remains incurable; therefore, better treatment options are needed
to help pts achieve durable responses. Epcoritamab is a subcutaneously administered
bispecific antibody that binds to CD3 on T cells and CD20 on malignant B cells. In
the dose-escalation portion of the first-in-human phase 1/2 trial (EPCORE NHL-1),
epcoritamab monotherapy (0.76–48 mg) resulted in an overall response rate (ORR) of
90% and a complete response rate of 50% in pts with R/R FL. Because epcoritamab and
R2 have distinct mechanisms of action and generally non-overlapping toxicity profiles,
combining these treatments may safely enhance antitumor response.
Aims: To present updated results for epcoritamab with R2 in pts with R/R FL (EPCORE
NHL-2 arm 2; NCT04663347).
Methods: Adult pts with R/R CD20+ FL received subcutaneous epcoritamab + R2 for 12
cycles (28 d/cycle). In the dose-escalation part of the trial, epcoritamab was administered
at 24 or 48 mg; in the expansion part, the dose was 48 mg. Pts received their assigned
epcoritamab dose as follows: every wk, cycles 1–3; every 2 wk, cycles 4–9; and every
4 wk, cycles ≥10 up to 2 y. Corticosteroid prophylaxis and step-up epcoritamab dosing
were required during cycle 1 to mitigate CRS. PET-CT was used to assess response.
All pts provided informed consent before enrollment.
Results: As of the data cutoff date of December 1, 2021, 30 pts had received epcoritamab
(24 mg, n=3; 48 mg, n=27) + R2. Median age was 68 y. Overall, 70% of pts had stage
IV disease, 67% had FLIPI scores 3–5, 30% had primary refractory disease, and 40%
had disease progression within 24 mo after starting first-line treatment (20% within
24 mo after starting immunochemotherapy). The median number of prior lines of therapy
was 1 (range, 1–5). With a median follow-up of 5.1 mo (range, 0.8–12.3), 25 pts (83%)
remained on study treatment; 5 pts had discontinued due to disease progression (n=2),
AEs (n=2), or withdrawal of consent (n=1). Frequent treatment-emergent AEs (TEAEs;
any grade [G]) included infections (57%), injection-site reactions (50%), constipation
(37%), fatigue (37%), and neutropenia (37%). CRS was reported in 15 pts (50%; G1 30%,
G2 13%, G3 7%). Most CRS events occurred in cycle 1, and all CRS events resolved with
standard management (median time to resolution, 3 d; range, 1–8). Tocilizumab was
used in 3 pts with CRS, and 1 pt discontinued study treatment due to CRS. ICANS (G2)
was reported in 1 pt and resolved. There were no fatal TEAEs. Response profiles for
individual pts are shown in Figure 1. Among the efficacy-evaluable pts, the ORR was
100% (27/27), with 93% (25/27) achieving a complete metabolic response (CMR) and 7%
(2/27) achieving a partial metabolic response. As of the data cutoff date, the longest
duration of response was 7.0+ mo and ongoing.
Image:
Summary/Conclusion: Subcutaneous epcoritamab + R2 demonstrated promising efficacy
in pts with R/R FL, with a high CMR rate. The safety profile remained consistent with
prior data. CRS events occurred mostly in cycle 1 and were generally low grade. Updated
data with 30 additional pts will be presented at the meeting.
P1136: INTERIM IMAGING DOES NOT PREDICT CLINICAL EVOLUTION IN FOLLICULAR LYMPHOMA
IN A REAL-LIFE CLINICAL SETTING
V. Ramos De Ascanio1,*, L. Sánchez Paz1, M. S. Infante1, J. Churruca Sarasqueta1,
M. Á. Foncillas García1, E. Landete Hernández1, K. D. C. Marín Mori1, C. Muñoz Novas1,
J. Á. Hernández Rivas1, I. González Gascón y Marín1
1Hematology, Infanta Leonor University Hospital, Madrid, Spain
Background: Interim scan evaluation is a common practice during the treatment of some
types of lymphomas with (immuno)-chemotherapy (ICT). In follicular lymphoma (FL) its
role remains controversial and few studies have looked at this topic specifically.
However, it is performed in clinical practice, clinical trials and recommended by
some international guidelines (ESMO).
Aims: In this study, we analyzed the clinical and prognostic value of interim imaging
(iPET/iCT) during the first line-treatment of patients diagnosed with FL.
Methods: In order to execute this, we carried out a retrospective analysis of all
FL patients that received a first line treatment (ICT) at our institution and underwent
an iPET/iCT between 2008 and 2021. Patients with Grade 3b or histologic transformation
were excluded from the analysis. All data was extracted from electronical medical
records. The Lugano response criteria were used to assess disease response. Statistical
analysis was performed using SPSS version 25.0 and survival was estimated by Kaplan-Meier
curves and log rank tests.
Results: Baseline characteristics of patients included in this study are described
in Table 1. Median follow-up was 71 months (6-153). All patients underwent iPET/iCT
(iCT=52 (81.3%); iPET=12 (18.8%)). Most iPET/iCT showed a partial response (PR) (n=48,
75%), and a minority a complete response (CR) (n=14, 21.9%). Two patients (3.1%) did
not respond to induction. The 14 patients with CR maintained the response at the end
of induction; 34 of 48 PR patients (70.8%), achieved a CR on final imaging; whereas
14 patients (29.2%) maintained a PR. Out of the 64 patients, 17 (26.6%) had disease
progression during the observation period. In 6 patients (9.4%) interim imaging conditioned
a treatment modification (premature discontinuation of therapy, increase/decrease
in the number of ICT cycles or ICT dose attenuation). No significant differences were
found in progression-free survival (PFS) (p=0.3) between the 14 patients with CR and
the 48 patients who showed PR or less in the interim imaging. Nevertheless, patients
in PR who converted to CR at the end of therapy had a significantly higher PFS than
patients who did not achieve CR at the end of induction (p<0.01). Consequently, patients
who reached a CR at the end of therapy achieved a significantly higher PFS compared
to those who did not (p<0.01).
Image:
Summary/Conclusion: Obtaining an early CR on interim imaging did not predict PFS in
FL patients treated with IQT. In addition, clinical decisions based on iPET/iCT were
only made in a minority of the patients, with the detection of early refractoriness
being residual. Conversely, end of treatment imaging assessment was a useful tool
capable of predicting PFS.
P1137: ACCURACY AND PROGNOSTIC IMPACT OF FDG PET/CT AND BIOPSY IN BONE MARROW ASSESSMENT
OF FOLLICULAR LYMPHOMA AT DIAGNOSIS: A NATION-WIDE STUDY.
I. Ródenas Quiñonero1,*, T. Chen-Liang1, T. Martín-Santos2, A. Salar3, M. Fernández-González2,
C. Celades4, J. T. Navarro5, A. B. Martinez6, R. Andreu7, A. Balaguer7, A. Martín8,
M. Baile8, J. López-Jiménez9, J. Marquet9, A. I. Teruel10, M. J. Terol10, C. Benet11,
L. Frutos12, J. L. Navarro12, J. Uña13, M. Suarez14, M. Cortes15, J. Contreras16,
C. Ruiz17, P. Tamayo18, J. Mucientes19, P. Sopena20, L. Reguilón-Gallego1, J. J. Sánchez-Blanco1,
E. Pérez-Ceballos1, A. Jerez1, F. J. Ortuño1
1Hematology, H. Morales Meseguer, Murcia; 2Hematology, H. Universitario de Canarias,
Tenerife; 3Hematology, H. del Mar, Barcelona; 4Hematology, Josep Carreras Leukaemia
Research Institute (IJC); 5Hematology, ICO-H. Germans Trias i Pujol, Badalona; 6Hematology,
H. Santa Lucía, Cartagena; 7Hematology, H. La Fe, Valencia; 8Hematology, H. Clínico
Universitario, Salamanca, Salamanca; 9Hematology, H. Ramon y Cajal, Madrid; 10Hematology,
H. Clínico, Valencia; 11Hematology, H. Arnau de ViIlanova, Valencia; 12Nuclear Medicine,
H. Virgen de la Arrixaca, Murcia; 13Nuclear Medicine, H. Universitario N.S. de la
Candelaria, Tenerife; 14Nuclear Medicine, H. del Mar; 15Nuclear Medicine, H. Universitari
de Bellvitge-IDIBELL, Barcelona; 16Nuclear Medicine, H. Santa Lucía, Cartagena; 17Nuclear
Medicine, H. La Fe, Valencia; 18Nuclear Medicine, H. Clínico Universitario de Salamanca/IBSAL,
Salamanca; 19Nuclear Medicine, H. Puerta de Hierro, Madrid; 20Nuclear Medicine, H.
9 de Octubre, Valencia, Spain
Background: In the workout of Follicular Lymphoma (FL), bone marrow biopsy (BMB) assessment
is a key component of FLIPI and FLIPI2, the most widely used outcome scores. During
the previous decade, several studies explored the role of FDG-PET/CT for detecting
nodal and extranodal disease, with only one large study comparing both techniques.
Aims: The aim of this study was to evaluate the diagnostic accuracy and the prognostic
impact of both procedures in a retrospective cohort of 299 FL patients.
Methods: Two hundred and ninety-nine, ≥18 y.o. patients diagnosed with FL between
June 2005 and December 2018 were included. We used the Kaplan-Meier and the Cox method
to analyze overall survival (OS) and progression free survival (PFS). To avoid collinearity
in the multivariate regressions, we deconstructed the FLIPI2 composite in its foundational
factors, considering bone marrow involvement (BMI) by mean of different measures in
four models: BMB+, PET/CT+, “BMB+ and PET/CT+” and “BMB+ or PET/CT+”. In Cox regressions,
examination of log (-log) survival plots ant partial residuals was performed to assess
that the underlying assumption of proportional hazards was met.
Results: The median follow-up was 57.3 months (range 3.6-185.8 months). The PET/CT
was positive in 58 patients and negative in 238. Among those positive, 37 also had
a BMB+, whereas 151 patients had a BMB+ with a PET/CT-. The BMB was positive in 124
patients and negative in 172. Among those negative, 21 had a PET/CT+. Considering
PET/CT, the sensitivity was 63.7% (95% confidence interval(CI);50.5-77.0), specificity
was 63.4% (95%CI;57.1-69.7), negative predictive value (NPV) was 87.7% (95%CI;82.6-92.9),
accuracy 63.5% (95%CI:57.8-69.1). Focusing BMB, the sensitivity was 29.8% (95%CI;21.3-38.3),
specificity was 87.7% (95%CI;82.6-92.9), NPV was 63.4% (95%CI;57.1-63.7) and accuracy
was 63.5% (95%CI:57.8-69.1).
Either beta2-microglobulin (B2M) over the upper normal limit (ULN), a diameter of
the largest involved node (LoDLIN) over 6cm and a hemoglobin lower than 120g/L, were
significantly associated with a shorter PFS in the univariate analysis. Also in univariate,
a positive BMB and the combined “PET/CT or BMB positive”, were significantly associated
with a shorter PFS. Neither BMI positive or the combined “PET/CT or BMB positive”,
could add an independent prognostic value to the two factors than remained significant
in the multivariate model: B2M higher than ULN and a LoDLIN over 6cm.
Regarding OS, an elevated B2M, a LoDLIN over 6cm, a hemoglobin lower than 120g/L and
an age older than 60y.o., were significantly associated with a shorter OS in the univariate
analysis. A positive result by BMB, a positive PET/CT and the combined “PET/CT or
BMB positive” result, were significantly associated with a shorter OS in univariate
regression. The combined “PET/CT or BMB positive” and an age older than 60y.o. remained
significant for a shorter OS in the multivariate model (p=0.030, HR 2.318, 95%CI 1.083-4.961
and p= 0.020, HR 2.473, 95%CI 1.151-5.314, respectively).
Image:
Summary/Conclusion: In our FL series, the combined “PET/CT or BMB positive” was the
only BMI variable that showed an independent prognostic value in FL. Therefore, our
data support that both procedures should be carried out in the upfront work of all
suitable FL patients.
P1138: COPANLISIB + RITUXIMAB VS RITUXIMAB + PLACEBO IN PATIENTS WITH RELAPSED INDOLENT
NON-HODGKIN LYMPHOMA (NHL): UPDATED SAFETY AND EFFICACY FROM THE PHASE III CHRONOS-3
TRIAL
P. L. Zinzani1,2,*, M. Özcan3, K. Sapunarova4, W. Jurczak5, A. Hamed6, K. Bouabdallah7,
G. Saydam8, K. Geissler9, Á. Szomor10, M. Lazaroiu11, A. Salar12, A. Tempescul13,
M. Nalcaci14, L. Gercheva15, M. Egyed16, P. Panayiotidis17, L. Mongay Soler18, A.
Cao18, C. Phelps18, B. H Childs18, M. J Matasar19
1IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seragnoli”;
2Dipartimento di Medicina Specialistica, Diagnostica e Sperintale, Universita di Bologna,
Bologna, Italy; 3Ankara University School of Medicine, Ankara, Turkey; 4Medical University,
Plovdiv, Bulgaria; 5Maria Skłodowska Curie National Research Institute of Oncology,
Krakow, Poland; 6Petz Aladár Megyei Oktató Kórház, Gyor, Hungary; 7Hematology and
Cellular Therapy Department, University Hospital of Bordeaux, Bordeaux, France; 8Ege
Üniversitesi Tip Fakültesi, Izmir, Turkey; 9Sigmund Freud University, Vienna, Austria;
10Pécsi Tudományegyetem Klinikai Központ, Pécs, Hungary; 11S.C. Policlinica de Diagnostic
Rapid S.A., Brasov, Romania; 12Hospital del Mar, Barcelona, Spain; 13Hôpital Morvan
- Brest, Brest, France; 14Istanbul Universitesi İstanbul Tip Fakultesi, Istanbul,
Turkey; 15MHAT Sveta Marina EAD, Varna, Bulgaria; 16Somogy Megyei Kaposi Mor Oktato
Korhaz, Kaposvar, Hungary; 17First Department of Propaedeutic Internal Medicine, National
and Kapodistrian University of Athens, School of Medicine, Athens, Greece; 18Bayer
HealthCare Pharmaceuticals, Inc., Whippany, NJ; 19Department of Medicine, Memorial
Sloan Kettering Cancer Center, New York, NY, United States of America
Background: The Phase III CHRONOS-3 trial of copanlisib plus rituximab (C+R) vs placebo
plus rituximab (P+R) in patients (pts) with indolent NHL (iNHL) showed a significant
improvement in progression-free survival (PFS; hazard ratio [HR] 0.52; Matasar et
al. Lancet Oncol 2021). At the time of data cut-off (August 31, 2020), 23% of pts
were ongoing on treatment in the C+R arm and 43% of pts had discontinued treatment
and entered active follow-up; corresponding numbers in the P+R arm were 19% and 17%.
Aims: Here we report an updated 1-year follow-up analysis of efficacy and safety for
all pts from CHRONOS-3 based on a data cut-off of August 6, 2021.
Methods: Eligible pts with iNHL who relapsed after the last R-containing regimen and
were progression-free and treatment-free for ≥12 months (mo) after the last R-containing
regimen, or for >6 mo if unwilling/unfit to receive chemotherapy, were randomized
2:1 to receive C+R or P+R. All pts provided informed consent. Treatment continued
until progression or unacceptable toxicity and was administered i.v. on a 28-day cycle,
with C 60 mg/P given on days 1, 8, and 15 and R 375 mg/m2 given on days 1, 8, 15,
and 22 of cycle 1 and on day 1 of cycles 3, 5, 7, and 9. Primary endpoint was centrally
assessed PFS and secondary efficacy endpoints included overall survival (OS), objective
response rate (ORR), duration of response (DoR), complete response rate (CRR), and
treatment-emergent adverse events (TEAEs).
Results: 307 pts were randomized to C+R and 151 to P+R. As of the August 2021 cut-off,
46 pts (15.0%) receiving C+R and 16 (10.6%) receiving P+R were ongoing on treatment.
The most common reason for discontinuation was adverse events (AEs) unrelated to clinical
progression (35.8%) for C+R and radiologic progression (49.7%) for P+R. Similar to
the initial disclosure, C+R reduced the risk of progression or death vs P+R by 47%,
with median PFS of 22.3 mo (95% confidence interval [CI] 19.4, 30.7) for C+R and 13.8
mo (10.8, 17.5) for P+R (HR 0.53 [95% CI 0.41, 0.70]; p=0.000001). PFS benefit was
preserved across all cancer subtypes (HR [95% CI]): follicular lymphoma (n=275; 0.57
[0.41, 0.80]); marginal zone lymphoma (n=95; 0.53 [0.28, 0.99]); small lymphocytic
lymphoma (n=50; 0.22 [0.10, 0.49]); lymphoplasmacytic/Waldenström macroglobulinemia
(n=38; 0.42 [0.15, 1.16]). Updated ORRs were 80.8% vs 49.7% and CRRs were 34.9% vs
14.6% for C+R vs P+R, respectively. Median OS was not estimable and no significant
benefit was seen for C+R over P+R, although the risk of death was reduced (HR 0.86
[95% CI 0.55, 1.32]) vs the August 2020 disclosure (HR 1.07 [0.63, 1.82]). Median
DoR was 25.7 mo (17.6, 31.5) for C+R vs 17.3 mo (12.6, 25.3) for P+R; for pts with
complete responses, median DoR was 42.3 mo (29.2, not estimable) vs 24.7 mo (10.2,
29.7) (HR 0.46 [0.24, 0.87]). Safety profiles for C+R and P+R were mostly unchanged
from the initial disclosure, and the most common treatment-related AEs for pts receiving
C+R were hyperglycemia (69.4%), hypertension (49.5%), and diarrhea (33.9%). For C+R,
6 additional pts (2.0%) discontinued C due to an AE, bringing the total to 33.2%.
The incidence of grade 3/4 treatment-related TEAEs was generally unchanged with the
longer follow-up (2 new grade 4 for C+R; 1 new grade 4 for P+R), with no new grade
5 events.
Summary/Conclusion: 1 year after the primary disclosure of CHRONOS-3, C+R continued
to demonstrate acceptable safety and superior efficacy with durable complete responses
vs P+R in pts with relapsed iNHL. These data support the long-term use of C+R in pts
with relapsed iNHL.
P1139: A POPULATION-BASED STUDY OF ISOLATED ADULT PULMONARY LANGERHANS CELL HISTIOCYTOSIS
IN THE UNITED STATES: 2010-2017
G. Ruan1,*, J. Abeykoon1, A. Hazim2, A. Ravindran3, K. Rech3, J. Young4, C. Cox4,
J. Ryu5, M. Koster6, O. Tobin7, M. Shah1, N. Bennani1, R. Vassallo5, G. Goyal1,8,
R. Go1
1Hematology; 2Internal Medicine; 3Laboratory Medicine and Pathology; 4Radiology; 5Pulmonology;
6Rheumatology; 7Neurology, Mayo Clinic Rochester, Rochester; 8Hematology, University
of Alabama, Birmingham, United States of America
Background: Isolated adult pulmonary Langerhans cell histiocytosis (pLCH) is a rare
histiocytic neoplasm that involves the lungs as single system disease. Retrospective
studies have suggested that patients with isolated adult pLCH have better overall
survival compared to multisystem LCH. While population-based studies have been done
for LCH with multisystem disease, there has been no epidemiologic studies done to
date on isolated adult pLCH.
Aims: To characterize the incidence and outcomes of isolated adult pLCH in the United
States.
Methods: We queried the Surveillance, Epidemiology, and End Results Program (SEER)
18 Registries (2010-2017) using ICD-0-3 code 9751/3 with the primary site of involvement
being the lung. Patients were included if they were ≥ 18 years and had localized pulmonary
involvement. Patients with extrapulmonary disease or unknown staging were excluded.
The primary outcome was overall survival (OS), which was analyzed using the Kaplan-Meier
method. Data on incidence rates (IR) and relative survival (RS) were calculated using
the SEER*Stat software. IR (cases/1,000,000) was age-adjusted to the U.S. 2000 standard
population. RS was defined as the ratio of the proportion of observed survivors in
a cohort of isolated adult pLCH to the proportion of expected survivors in a comparable
set of individuals that do not have isolated adult pLCH adjusting for the general
survival of the US population for race, sex, age, and time when the diagnosis was
established.
Results: In SEER, there were a total 147 patients with isolated adult pLCH. Between
the years 2010-2017, the median IR of isolated adult pLCH was 0.3 (range 0.1-0.5)
and did not change significantly. The median IR from 2010-2013 was 0.2 (range 0.1-0.2)
and 2014-2017 was 0.5 (range 0.3-0.5). The median age at diagnosis was 53 years (range
23-79) and 54 (37%) were male. 115 (78%) were white, 21 (14%) were black, and 11 were
labeled as other. 116 (79%) were diagnosed on histopathologic evaluation while the
remainder were diagnosed without microscopic confirmation. No patients were documented
to have secondary malignancies.
With a median follow up of 35 months, the median OS was not reached (95% CI: not reached-not
reached) and the 3-year OS was 89%. The 3-year OS between males and females was 93%
and 87% (p=0.16) respectively. The 3-year OS between white and non-white was 90% and
87% (p=0.98) respectively. In the general U.S. population, the expected survival for
1, 2, 3 years were 99.2%, 98.5%, 97.8% respectively. In contrast, the RS for patients
with isolated adult pLCH at the same time points were 98.3%, 94.8%, 90.9% (Figure).
A total of 14 patients died. 3 patients died from isolated adult pLCH while 11 patients
died of other causes, including chronic obstructive pulmonary disease (3), cardiac
disease (3), chronic liver disease (1), and unknown (4).
Image:
Summary/Conclusion: The incidence of isolated adult pLCH has not changed over the
years from 2010-2017. Patients with isolated adult pLCH have lower relative survival
compared to the general population. Among patients who died, the majority died from
cause other than pLCH.
P1140: TIME TO TUMOR, SYMPTOMATIC AND LABORATORY RESPONSES FOLLOWING SILTUXIMAB TREATMENT
IN IDIOPATHIC MULTICENTRIC CASTLEMAN DISEASE
F. van Rhee1,*, A. Rosenthal2, K. Kanhai3, R. Martin3, K. Nishimura2, A. Hoering2,
D. Fajgenbaum4
1University of Arkansas for Medical Sciences, Little Rock; 2Cancer Research And Biostatistics,
Seattle, United States of America; 3EUSA Pharma, Hemel Hempstead, United Kingdom;
4University of Pennsylvania, Philadelphia, United States of America
Background: Idiopathic multicentric Castleman disease (iMCD) is a rare, heterogeneous
disorder involving multicentric lymphadenopathy, systemic inflammation, and cytokine-driven
organ dysfunction. Siltuximab—a monoclonal antibody against interleukin-6—is the only
FDA- and EMA-approved treatment for iMCD. A phase 2, randomized, double-blind, placebo-controlled
trial of siltuximab for the treatment of iMCD (NCT01024036) showed that 18/53 patients
(34%) responded to treatment, compared with 0% in the placebo group, but it is not
known how long patients should be maintained on siltuximab before deciding whether
the treatment is beneficial.
Aims: To determine the time to and sequence of normalization of laboratory, clinical,
and lymph node responses in patients who responded to siltuximab.
Methods: The phase 2 trial enrolled adults with symptomatic iMCD who were human immunodeficiency
virus seronegative and human herpesvirus 8 negative. In this post hoc analysis, we
aimed to determine the sequence of and time to normalization of laboratory, clinical,
and lymph node responses in patients who responded to siltuximab. The Kaplan–Meier
and cumulative incidence with competing risks methods with corresponding log-rank
tests were produced to estimate and compare the progression-free survival (PFS), overall
survival, time to treatment failure, time to lymph node response, time to durable
symptomatic response, and time to normalization of laboratory values.
Results: Seventy-nine patients were enrolled in the trial (n=53 siltuximab, n=26 placebo;
all patients received best supportive care). The median duration of follow-up was
422 days (range 55‒1051). Siltuximab treatment improved PFS compared with placebo
(p=0.0001). The median PFS was 14.5 months (95% CI 13.6–upper bound not reached) for
patients on placebo, whereas it was not reached for patients on siltuximab. The 2-year
estimates for overall survival were 93% (95% CI 85–100%) for the siltuximab arm and
77% (95% CI 55–98%) for the placebo arm (p=0.11). Time to treatment failure in the
placebo arm was 4.8 months and was not reached with siltuximab (p=0.005). In the 18
siltuximab-treated patients who achieved the primary endpoint of durable tumor (radiologic)
and symptomatic response, most first achieved rapid normalization of various laboratory
parameters and symptomology (median of 0.8 months to normalization based on a 34-point
MCD symptom score) (Table 1). Lymph node responses occurred later (median of 4.1 months
to normalization).
Image:
Summary/Conclusion: Significant improvements in biochemical parameters such as platelet
count, albumin, and symptomatic responses occur within 3 months in the majority of
responders, showing them as early indicators of response. Lymph nodes were slower
to show an improvement in response to treatment, and so involution of these should
not be used to drive early therapeutic decisions in iMCD patients following siltuximab.
The present study shows the recovery sequence of different parameters over time following
initiation of treatment with siltuximab.
P1141: IBRUTINIB AND RITUXIMAB AS FIRST LINE THERAPY FOR MANTLE CELL LYMPHOMA: A MULTICENTRE,
REAL-WORLD UK STUDY
A. Tivey1,*, R. Shotton1, T. A. Eyre2, D. Lewis3, N. Crosbie3, E. Nga4, R. Guerrero
Camacho5, W. Swe6, H. Marr7, C. Rees8, S. Moule8, T. Sutton9, D. Wrench10, N. Thomas11,
M. Wilson11, J. Bailey12, M. Prahladan13, A. Hodson13, M. Koppana13, S. Smith14, S.
Jones14, F. Miall15, J. Norman16, E. Davies16, C. Hildyard17, L. Lowry18, S. Paneesha19,
I. Qureshi19, A. Beech20, C. Bedford21, A. Everden21, D. Tucker21, J. Wright22, J.
Goddard22, T. Nicholson23, J. Wilson24, A. Lord25, B. Jackson25, M. Flont26, A. Gibb27,
K. Linton27
1Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester; 2Oxford
University Hospitals NHS Foundation Trust, Oxford; 3University Hospitals Plymouth
NHS Foundation Trust, Plymouth; 4Airedale NHS Foundation Trust, Airedale; 5Blackpool
Teaching Hospitals NHS Foundation Trust, Blackpool; 6Calderdale and Huddersfield NHS
Foundation Trust, Huddersfield; 7The Freeman Hospital, Newcastle; 8Frimley Health
NHS Foundation Trust, Frimley; 9Gateshead Health NHS Foundation Trust, Gateshead;
10Guy’s and St Thomas’ NHS Foundation Trust, London, London; 11Harrogate and District
NHS Foundation Trus, Harrogate; 12Hull University Teaching Hospitals NHS Trust, Hull;
13East Suffolk and North Essex NHS Foundation Trust, Ipswich; 14Sherwood Forest Hospitals
NHS Foundation Trust, Sutton in Ashfield; 15University Hospitals of Leicester NHS
Foundation Trust, Leicester; 16Manchester University NHS Foundation Trust, Manchester;
17Milton Keynes University Hospital, Milton Keynes; 18Somerset NHS Foundation Trust,
Taunton; 19University Hospitals Birmingham NHS Foundation Trust, Birmingham; 20Nottingam
University Hospitals, Nottingham; 21Royal Cornwall Hospitals NHS Trust, Truro; 22Sheffield
Teaching Hospitals NHS Foundation Trust, Sheffield; 23St Helens & Knowsley Teaching
Hospitals NHS Trust, Whiston; 24University Hospitals Sussex NHS Foundation Trust,
Chichester; 25Torbay and South Devon NHS Foundation Trust, Torbay; 26York and Scarborough
Teaching Hospitals NHS Foundation Trust, York; 27Department of Medical Oncology, The
Christie NHS Foundation Trust, Manchester, Manchester, United Kingdom
Background: Ibrutinib (IBR) is an oral covalent Bruton tyrosine kinase inhibitor (BTKi),
licensed for treatment of relapsed or refractory mantle cell lymphoma (MCL). Under
NHS interim Covid-19 agreements in England, IBR with or without rituximab (R) was
approved for the frontline treatment for MCL patients (pts) as a safer alternative
to conventional immunochemotherapy. Although recent phase 2 studies have reported
high response rates in low-risk patients for this combination in the frontline setting,
randomised phase 3 and real-world data are currently lacking.
Aims: To describe the real-world response rates (overall response rate (ORR), complete
response (CR) rate) and toxicity profile of IBR +/- R in adult patients with previously
untreated MCL.
Methods: Following institutional approval, adults commencing IBR +/- R for untreated
MCL under interim Covid-19 arrangements were prospectively identified by contributing
centres. Hospital records were interrogated for demographic, pathology, response,
toxicity and survival data. ORR/CR were assessed per local investigator according
to the Lugano criteria using CT and/or PET-CT.
Results: Data were available for 66 pts (72.7% male, median age 71 years, range 41-89).
Baseline demographic and clinical features are summarised in Table 1. 23/66 pts (34.8%)
had high-risk disease (defined as presence of TP53 mutation/deletion, blastoid or
pleomorphic variant MCL, or Ki67%/MiB-1 ≥30%). IBR starting dose was 560mg in 56/62
pts (90%) and was given with R in 22/64 pts (34%). At a median follow up of 8.7 months
(m) (range 0-18.6), pts had received a median of 7 cycles of IBR. 19/60 pts (32%)
required a dose reduction or delay in IBR treatment. New atrial fibrillation and grade
≥3 any-cause toxicity occurred in 3/59 pts (5.8%) and 8/57 (14.0%) respectively.
For the whole population and high-risk pts only, ORR was 74.4% and 64.7% respectively
(p=0.2379), with a median time to response of 3.8m, coinciding with the first response
assessment scan. Seven pts (16.7%), of whom 2 had high-risk disease, attained CR at
a median of 6.0m. ORR for pts receiving vs not receiving R were 84.2% and 66.7% respectively
(p=0.1904). IBR was discontinued in 20/61 pts (32.8%) at a median time to discontinuation
of 4.1m, due to progressive disease (PD, 19.7%), toxicity (4.9%), death (3.3%; 1 pt
each of Covid-19 and E. coli infection), pt choice (3.3%) and other unspecified reasons
(1.6%). 15/66 pts (22.7%) overall and 7/23 (30.4%) with high-risk disease progressed
on IBR at a median time to PD of 4.0m. No pts underwent autologous stem cell transplantation
consolidation during the study period. 12/57 pts (21.1%) received second line treatment
(R-chemotherapy n=7, Nordic MCL protocol n=2, VR-CAP n=2, pirtobrutinib n=1). Response
to second line treatment was CR in 4/11 pts, PD in 7/11. Of the 2 Nordic-treated patients,
1 had CR after cycle 2 and 1 PD. Fourteen pts (21.2%) died during the follow up period,
due to MCL (n=11), Covid-19 (n=2) and congestive cardiac failure (n=1). Overall survival
was lower for patients with high-risk disease (HR 0.55, p=0.038).
Image:
Summary/Conclusion: In this real-world UK cohort of pts receiving first-line IBR +/-R
for MCL, including older and high-risk pts, we report high ORR rates in a similar
range to the phase II Geltamo IMCL-2015 study of combination IBR-R in an exclusively
low-risk population. Documented CR rates were lower, possibly reflecting a low usage
of rituximab in the Covid-19 pandemic as well as CT assessment of response. Treatment
was generally well tolerated, with low rates of toxicity-related treatment discontinuation.
The study is ongoing.
P1144: RADIOIMMUNOTHERAPY (RIT) VERSUS AUTOLOGOUS HEMATOPOIETIC STEM-CELL TRANSPLANTATION
(ASCT) IN RELAPSED/REFRACTORY (R/R) FOLLICULAR LYMPHOMA: A FONDAZIONE ITALIANA LINFOMI
(FIL) PHASE III TRIAL.
M. Ladetto1,*, R. Tavarozzi1, A. Evangelista2, M. Zanni3, A. Tucci4, A. Anastasia4,
B. Botto5, C. Boccomini6, S. Bolis7, S. Volpetti8, V. R. Zilioli9, B. Puccini10, A.
Arcari11, V. Pavone12, G. Gaidano1, P. Corradini13, M. Tani14, S. Ferrero15, F. Cavallo15,
G. Milone16, C. Ghiggi17, A. Pinto18, D. Pastore19, A. J. Ferreri20, G. Latte21, C.
Patti22, F. Re23, L. Arcaini24, F. Benedetti25, S. V. Usai26, S. Luminari27, D. Mannina28,
A. Pulsoni29, C. Stelitano30, E. Pennese31, G. Pietrantuono32, F. Gherlinzoni33, F.
Pomponi34, A. Olivieri35, T. Perrone36, D. Rota Scalabrini37, C. Califano38, B. Falini39,
G. Ciccone2, U. Vitolo37
1Department of Translational Medicine, University of Eastern Piedmont, Novara; 2Unit
of Clinical Epidemiology, A.O.U. Città della Salute e della Scienza, Torino; 3SCDU
of Hematology, AO SS Antonio e Biagio e Cesare Arrigo, Alessandria; 4Hematology, ASST
Spedali Civili, Brescia; 5Hematology, A.O.U. Città della Salute e della Scienza; 6Hematology,
A. O. U. Città della Salute e della Scienza, Torino; 7Hematology Department, ASST
San Gerardo University Hospital, Monza; 8Institute of Hematology, University and Hospital
of Udine, Udine; 9ASST Grande, Ospedale Metropolitano Niguarda, Milano; 10Hematology
Department, AOU Careggi, Firenze; 11Hematology, Ospedale Guglielmo da Saliceto, Piacenza;
12Hematology, Card. Panico Hospital S.Pio X, Tricase; 13Division of Hematology and
Stem Cell Transplantation, University of Milano Istituto Nazionale Tumori, Milano;
14Hematology, Santa Maria delle Croci Hospital, Ravenna; 15Department of Molecular
Biotechnologies and Health Sciences, Division of Hematology, University of Turin,
Torino; 16Hematology and BMT Unit, Azienda Policlinico Vittorio Emanuele, Catania;
17Clinic of Hematology, IRCCS Ospedale San Martino, Genova; 18Hematology-Oncology
and Stem-Cell Transplantation Unit, National Cancer Institute, Fondazione ‘G. Pascale,
Napoli; 19Ospedale Antonio Perrino, Brindisi; 20Onco-Hematology Department, Fondazione
Centro San Raffaele, Milano; 21Unità di Ematologia e Trapianto di Midollo Osseo, San
Francesco hospital, Nuoro; 22Divisione di Oncoematologia, Azienda Villa Sofia-Cervello,
Palermo; 23Hematology, A.O.U. di Parma, Parma; 24Department of Onco-Hematology, Fondazione
IRCCS Policlinico San Matteo, University of Pavia, Pavia; 25Department of Medicine,
Section of Hematology and Bone Marrow Transplant Unit, University of Verona, Verona;
26Ospedale Businco, Cagliari; 27Hematology, IRCCS Reggio Emilia, Reggio Emilia; 28Hematology,
Azienda Ospedaliera Papardo, Messina; 29Hematology, Department of Translational and
Precision Medicine, Sapienza University of Rome, Roma; 30Hematology, Azienda Ospedaliera
Bianchi-Melacrino-Morelli, Reggio Calabria; 31Lymphoma Unit, Department of Hematology,
Ospedale Spirito Santo, Pescara; 32IRCCS Centro Oncologico Della Basilicata, Rionero
In Vulture; 33Division of Hematology, Ca’ Foncello Hospital, Treviso; 34ULSS8 Berica,
Vicenza; 35Hematology, AOU Ospedali Riuniti, Ancona; 36Unit of Hematology with Transplantation,
Dept. of Emergency and Organ Transplantation, University of Bari, Bari; 37Candiolo
Cancer Institute, FPO-IRCCS, Candiolo; 38U.O.C. Ematologia, P.O. “Andrea Tortora”,
Pagani; 39Department of Medicine, Division of Hematology and Clinical Immunology,
University of Perugia, Perugia, Italy
Background: Optimal consolidation for young R/R FL in the rituximab age remains uncertain
and the benefit of ASCT is not clearly established.
Aims: The FIL FLAZ12 trial (NCT01827605) is a prospective, multicenter, randomized,
phase 3 trial, comparing RIT versus ASCT, as consolidation after chemoimmunotherapy,
both followed by R maintenance in R/R FL.
Methods:
Pts aged 18-65 yrs, with R/R FL after 1 or 2 lines of chemoimmunotherapy, without
significant comorbidities were enrolled. Patients received 3 courses of therapy chosen
by the investigator among RCHOP, R-DHAP, R-FM, R-ICE, R-IEV or R-B. Pts achieving
at least PR (according to Cheson et al. 2007) were randomized 1:1 to either RIT or
ASCT before CD34+ collection. Conditioning for ASCT was BEAM or TEAM. RIT was given
as previously described (Morschhauser et al., 2008). After consolidation, pts received
R maintenance every 3 months for eight courses. Primary endpoint was PFS. Considering
ASCT toxicity, it was hypothesized to be a superior choice, if capable of increasing
3-years PFS from 40% to 60% (two-side log-rank test with alpha of 5% and a power of
85%). Clinical secondary endpoints were ORR, CRR, OS, EFS and TTF.
Results:
Between Aug 2012, and Sep 2019, 164 pts were screened and 159 enrolled by 38 FIL Centers
(enrolled population). Unfortunately, the study was prematurely closed due to low
accrual. The data were analyzed on an ITT basis on May 2, 2021 with a median follow-up
(mFU) from enrollment of 43 months and 75 PFS events. The two arms were clinically
well balanced, with median age of 57 yrs (IQR 49- 62), 55% male, 57% stage IV, 20%
bulky disease. Tumor re-biopsy was performed in 79% pts. POD-24, retrospectively assessed
was observed in 32% of pts. Two pts (1%) did not start treatment (non-confirmed histology
and withdrawal). Sixteen (10%) pts discontinued before randomization (7 SD, 3 PD,
3 AE, 1 withdrawal, 2 poor compliance) and 141 (89%) were randomized to either RIT
(71) or ASCT (70) (randomized population). Of these 19 (13%) (RIT 8, ASCT 11) did
not receive the planned consolidation due to 7 PD, 4 AE, 1 medical decision, 2 poor
mobilization, 2 withdrawals, 1 poor compliance, 2 protocol breaches, while 63 (89%)
received RIT and 59 (84%) ASCT. After RIT, 61% of pts achieved CR and 23% PR, while
after ASCT these were 70% and 9%.
Estimated PFS at 3 yrs was 60% (95% CI: 46%-71%) in the RIT arm vs. 59% (95% CI:45%-70%)
in the ASCT arm, p = 0.8613 (HR 0.96, 95%CI: 0.57,1.59). (Figure 1) 3yrs-OS was again
superimposable in the two arms: 83% (95%CI: 69%-91%) in the RIT vs 85% (95% CI: 72%-91%)
in the ASCT, p = 0.8310 (HR 1.10, 95%CI: 0.45,2.72). Grade ≥ 3 hematological toxicity
was 46% in the RIT vs 94% in the ASCT arm (p < 0.001). For ASCT vs RIT grade ≥3 neutropenia
occurred in 94% vs 41% of pts (p < 0.001). During follow-up, 4 pts died in remission:
1 AML (RIT), 2 SARS- COV2 infections (RIT) and 1 pneumonia (ASCT). Second cancers
occurred in 3 pts after RIT and 7 after ASCT (p = 0.480). Multivariable analysis for
PFS indicated POD-24, male sex, LDH and refractory disease as adverse parameters.
Subgroup analysis for PFS including gender, age, LDH, POD-24 and extranodal disease
show no subgroup favoring RIT nor ASCT.
Image:
Summary/Conclusion: Even if prematurely interrupted, our study demonstrated no meaningful
difference in efficacy between ASCT and RIT, but ASCT was more toxic and more demanding
for pts and health service. Both strategies induced a similar and favorable long-term
outcome suggesting that consolidation programs milder than ASCT require further investigation
in R/R FL.
P1145: OUTCOMES AND TREATMENT PATTERNS AFTER FIRST RELAPSE IN PATIENTS WITH WALDENSTRÖM
MACROGLOBULINEMIA
R. Tawfiq1,*, J. Abeykoon2, S. Zanwar2, J. Paludo2, P. Kapoor2
1Department of Medicine; 2Division of Hematology, Mayo Clinic, Rochester, United States
of America
Background: Waldenström macroglobulinemia (WM) is a rare, B cell lymphoma, with a
relapsing-remitting course. Despite expansion of therapeutic options, WM remains incurable.
Data on outcomes of patients (pts) after first relapse or primary refractory disease
are sparse. Frequently used salvage therapies include chemoimmunotherapy (CIT), proteasome
inhibitor (PI) based regimens and Bruton tyrosine kinase inhibitors (BTKi).
Aims: Analyze the outcomes of pts with WM after initial relapse and/or refractory
(RR) disease and the impact of the type of therapy used in the second line setting.
Methods: ecords of pts with WM seen at Mayo Clinic between 2000 and 2021 were reviewed.
Pts with RRWM requiring second line therapy were included in the primary analysis.
Response rates were assessed per the modified IWWM-6 criteria. All time to event analyses,
barring progression free survival 2 (PFS2), were calculated from the start of the
second line treatment, using the Kaplan Meier method. Pts were grouped based on sequence
of therapy and response to frontline treatment for additional subgroup analysis.
Results: Records of 220 pts with WM were reviewed; 92 had RRWM and were treated with
a second line therapy. For the entire cohort, frontline regimens included dexamethasone,
rituximab, cyclophosphamide (DRC) (n=93, 42%), bendamustine rituximab (BR) (n=71,
32%), bortezomib, dexamethasone, rituximab (BDR) (n=33, 15%), BTKi (n=20, 9%), and
other (n=3, 1%). Median follow-up of RRWM cohort from second line therapy was 5.1
years (95% CI: 4.5-6.3). Pts with RRWM received the following therapies at first relapse:
BR (n=24, 26%), BTKi (n=24, 26%), PI (n=17, 19%), DRC (n=6, 7%), autologous transplant
(n=4, 4%), and other (n=17, 18%). Following second-line treatment, the overall response
rate and major response rate (MRR) were 72% and 67% (CR=5%, VGPR=15%, and PR=47%),
respectively. The median PFS was 2.7 years (CI: 1.7-4). 2-year overall survival (OS)
rate was 87%. The median PFS2 from frontline therapy was 7.03 years (CI: 5.5-9.8).
Baseline characteristics were similar for subgroups in all the following subgroup
analyses. With first-line therapy, 54% of pts (n=50) achieved MRR vs. 46% (n=42) who
did not. The former group had similar progression-free survival (PFS) rates as well
as overall survival (OS) rates and response rates (MRR and ORR) to the second-line
treatment compared to their counterparts who did not achieve MRR with the frontline
regimen (Table).
Thiry one pts received BTKi as either first or second line and 61 pts received no
BTKi in either line. As expected, the BTKi group was enriched for pts with MYD88
L265P mutation (n=29, 94%) vs 49% (n=30) in the no BTKi group, p<0.001. The outcomes
between the 2 groups were comparable (Table). The group that received frontline BTKi
followed by CIT/PI based Rx as second line (n=7) was compared to that receiving frontline
CIT/PI based Rx followed by BTKi (n=21). The outcomes were similar (Table).
Image:
Summary/Conclusion: Currently available second line therapies are effective in RRWM.
Achieving MRR with frontline regimen does not significantly affect overall outcomes
with the second-line therapy. Although follow up is short, using a BTKi as the first-
or second-line therapy appears to show comparable outcomes after second line treatment
as compared to not treating with a BTKi in the upfront setting or in second-line setting.
Similarly, using frontline BTKi followed by CIT as second line shares similar outcomes
to treating with CIT initially followed by a BTKi. These findings require confirmation
in prospective studies.
P1147: SERUM ALBUMIN AND NEUTROPHIL-TO LYMPHOCYTE RATIO TWO INDEPENDENT FACTOR PREDICTING
SURVIVAL IN FOLLICULAR LYMPHOMA. MULTI-INSTITUTIONAL 763 COHORT LA
M. A. Torres Viera1,*
1LYMPHOMAS, Hematologia Oncologia 360 Clinica Sta Sofia, Caracas, Venezuela
Background: Introduction: The neutrophil-lymphocyte ratio (NLR) is a measure of systemic
inflammation that appears prognostic in different cancers. Although the exact mechanism
remains to be elucidated, reduced lymphocyte intratumoral infiltration coupled with
the formation of neutrophil extracellular traps (or NETosis) have been postulated
as endogenous mechanisms for tissue damage and inflammation. Along this line, serum
albumin has also been studied as a biomarker of inflammation and has been associated
to prognosis in certain cancers. We have previously reported on the prognostic value
of the NLR and serum albumin in diffuse large B-cell lymphoma (Villela, ASH meeting,
2019; Castro, ASH meeting, 2019) and peripheral T-cell lymphoma, not otherwise specified
(Idrobo, ASH meeting, 2019), but nothing on follicular lymphoma (FL) yet.
Aims: Therefore, we aim to investigate the role of different biomarkers on the prognosis
of patients with FL diagnosed and managed in Latin America
Methods: We analyzed patients with FL diagnosed between 2010 and 2020 from 30 centers
in 10 Latin American countries. The study outcomes were overall survival (OS) and
progression-free survival (PFS) in relation to different biomarkers. Kaplan-Meier
and log-rank test were used for survival analysis. Univariate and multivariate Cox
regression analysis were used to estimate hazard ratios (HR) with a 95% confidence
interval (CI) and adjusted to the Follicular Lymphoma International Prognostic Index
(FLIPI) score. Outcomes with a p-value <0.05 were considered statistically significant
Results: We identified 939 FL patients; 741 were included for the final analysis (median
age 58 y, female 52%). There was no significant correlation between the NLR and other
clinical factors such as: age, clinical stage, histological FL grading, and chemotherapy
regimen used. A cutoff of 2.15 for NLR was defined as the maximum point for sensitivity
and specificity based on ROC analysis. Table 1 and 2 summarizes the results from the
univariate and multivariate analysis for 2 years OS and PFS, respectively. Both, serum
albumin <3.5 g/dL and a NLR >2.15 were independently associated with worse OS (adjusted,
aHR 2.48 [1.26-4.91], p=0.009; and 2.55 [1.21-5.37], p=0.014) and PFS (aHR 1.62 [1.03-2.55],
p=0.038; 2.22 [1.45-3.40], p<0.001), respectively. The lymphocyte:monocyte ratio (LMR)
was not found to be prognostic for OS or PFS, although with a trend for worse PFS
with a LMR ≤2.5. With a median follow of 43 months, (95% CI: 40-47), the survival
rates in patients with FL and albumin <3.5 were OS of 83% (vs. 95%) and PFS of 70%
(vs. 83%); whereas in patients with NLR >2.15 the survival rates were OS of 91% (vs.
96%) and PFS of 75% (vs. 88%) (Figures 1 and 2; Table 3).
Summary/Conclusion: In this study, serum albumin and NLR emerge as reliable predictors
for survival for FL patients in Latin America. Although these markers have been associated
to an increased inflammatory state in cancer patients; other factors such as poor
nutritional status, and advanced disease stage due to delayed access to specialized
cancer care in our region may have contributed to the observed outcome. Further studies
are needed to better understand the role of these biomarkers on lymphoma care and
to validate our findings. Lastly, we are currently working on evaluating these biomarkers
on existing prognostic models and to improve prognostication for FL patients in Latin
America
P1148: REAL WORLD DATA (RWD) AMONG FOLLICULAR LYMPHOMA (FL) PATIENTS IN GERMANY WITH
AT LEAST TWO PRIOR LINES OF SYSTEMIC THERAPY AND COMPARISON WITH CLINICAL DATA FOR
MOSUNETUZUMAB
N. Marschner1,*, J. Hanselmann2, S. McGough3, S. Braun4, S. Pfizler-Dempfle5, R. Sandner6,
J. Mohm7, M. Jaber8, D. Hamm9, N. Hamm10, M. Jaenicke11, A. Shewade3
1Outpatient Centre for Interdisciplinary Oncology and Haematology; 2Biostatistics,
iOMEDICO, Freiburg, Germany; 3Roche Global Product Development Data Sciences, Genentech,
Inc., South San Francisco, CA, United States of America; 4Biometrics and Epidemiology.
Roche Pharma AG, Grenzach-Wyhlen; 5Oncology and Haematology Outpatient Centre, Kaiserslautern;
6MVZ for Haematology and Oncology, Passau; 7Oncology and Haematology Outpatient Centre,
Dresden, Germany; 8Roche Global Access, F. Hoffmann-La Roche Ltd, Basel, Switzerland;
9Medical Affairs Excellence, Roche Pharma AG, Grenzach-Wyhlen; 10Medical Department,
iOMEDICO; 11Clinical Epidemiology, iOMEDICO, Freiburg, Germany
Background: FL is characterized by its indolent nature, with patients (pts) experiencing
multiple relapses and successively poorer outcomes with each relapse (Link et al.
2019). Treatment with immunochemotherapy regimens has significantly improved outcomes
in pts with newly diagnosed FL, but limited treatment options are available in Europe,
with no standard of care for pts with multiple relapses, such as those who have received
at least two prior lines of systemic therapy (3L+ FL). Mosunetuzumab (M) is a T-cell
engaging CD20xCD3 bispecific antibody that has shown high rates of complete response
and favorable safety in pts with 3L+ FL in a single-arm Phase II trial (NCT02500407;
Budde et al. ASH 2021). RWD can provide relevant context to interpret data from the
single-arm trial (SAT) of M as a treatment option in 3L+ FL.
Aims: This study summarizes RWD in pts with 3L+ FL utilizing the Tumor Registry Lymphatic
Neoplasms extension (TLNext) and plans to perform a matching-adjusted indirect comparison
(MAIC) between the M SAT and TLNext.
Methods: TLN was a multicenter, longitudinal, observational, prospective cohort study
collecting data from medical charts of pts with lymphoid B-cell neoplasms receiving
care at office-based hematology practices in Germany (NCT00889798; Knauf et al. 2019).
Patients were enrolled between 2009–14 and followed for up to 5 years. TLNext collected
additional data up to November 2021 from consented pts originally enrolled in TLN
as well as additional newly-consented pts with 3L+ FL. Baseline characteristics, treatment
patterns, and outcomes were summarized for pts with 3L+ FL in TLNext. A MAIC is planned
between the M SAT and a TLNext cohort selected by applying key eligibility criteria
from the M SAT. An index line of therapy (LoT) will be selected for all pts with more
than one recorded LoT. Imbalance adjustment will be performed by re-weighting the
TLNext cohort using weights determined from distributions of prognostic clinical factors
observed in M SAT.
Results: TLNext included 69 pts who had received 112 documented LoTs in 3L+ FL. Median
follow-up was 40 months from the start of the 3rd LoT. 65 pts had received an anti-CD20
therapy and an alkylator therapy prior to index therapy and had a known relapse or
refractory status (Table). Latest recorded LoT was selected as the index therapy for
every patient. Compared to M SAT, the TLNext cohort was slightly older and had more
female pts. A higher proportion of pts in TLNext had received only 2 prior LoT. Overall,
M SAT had a higher proportion of pts with factors known to be associated with poorer
outcomes in 3L+ FL, e.g. refractory to prior anti-CD20 therapy, double-refractory
to prior anti-CD20 therapy and alkylator therapy. The most common regimens in the
TLNext cohort were bendamustine-like regimens (34%), PI3K-like regimens (15%), and
anti-CD20 monotherapy (14%). Comparative MAIC analyses are ongoing.
Image:
Summary/Conclusion: TLNext represents a real-world 3L+ FL cohort from Germany that
provides relevant context for the M SAT. TLNext outcomes data will be re-weighted
using the MAIC approach to generate a descriptive benchmark for outcomes observed
in M SAT. While comparisons between clinical trials and RWD carry several limitations
pertaining to measurement, reporting of data, and choice of analytical approach to
conduct the comparison, RWD from TLNext may be useful to describe a benchmark for
clinical practice in 3L+ FL in Germany and globally prior to the availability of randomized
evidence on newer treatment options like mosunetuzumab.
P1149: THE EVOLVING STATE OF PLAY IN 1000 PATIENTS WITH WALDENSTRÖM’S MACROGLOBULINAEMIA
IN THE UNITED KINGDOM (UK): A REAL-WORLD DATA ANALYSIS FROM THE WMUK RORY MORRISON
REGISTRY PROJECT
E. Uppal1,*, A. Otamas1, J. Khwaja1, H. McCarthy2, J. Kothari3, A. Rismani1, D. El-Sharkawi4,
C. Kyriakou1, S. D’Sa1
1Department of Haematology, University College London Hospitals NHS Foundation Trust,
London; 2Department of Haematology, Royal Bournemouth Hospital, Bournemouth; 3Department
of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford; 4Department
of Haematology, The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
Background: Waldenström’s Macroglobulinaemia (WM) is a rare B-NHL, with an incidence
of 0.55 per 100,000 per year in the UK (1). As novel therapies move from the bench
to the clinic, real world data (RWD) is important to understand trends and supplement
data from clinical trials. The Rory Morrison Registry (RMR) collects sequential data
on diagnosis, treatment landscape and outlook for patients from 25 UK centres.
Aims: To characterise therapy trends in the UK by an analysis of disease and treatment
characteristics, and survival status.
Methods: The RMR was searched for all patients with a diagnosis of WM. Information
regarding demographics, disease characteristics, bone marrow (BM) results, treatment
and survival status were collected retrospectively; the earliest diagnosis was in
1978 and the data lock was on 25/2/22.
Results: 1007 patients with a diagnosis of WM using consensus criteria (2) at 25 UK
Centres (94.5% academic, 5.5% district centres) were identified. The following data
pertain to diagnosis; median age was 64 years (21-93); M:F ratio 1.6:1, 90% of white
ethnicity. Diagnostic sub-entities included peripheral neuropathy (116), cryoglobulinaemia
(68), amyloidosis (22), Bing-Neel syndrome (18) and Schnitzler’s syndrome (9); 53%
(501/945) were symptomatic at diagnosis. The median IPSSWM for 496 patients was 2.
MYD88 L265P by RT-PCR was found in 87.1% (304/349) and CXCR4 mutations in 28.9% (26/90).
Of the total, 690 (68.5%) patients received treatment with a median time from diagnosis
to first line (L1) treatment of 3 months (0-312). Indications for L1 were recorded
as: 274/690 lymphoma related, 248/690 paraprotein-related, 65/690 B-Symptoms, 101/690
other indications. Regarding later lines, 402 (39.9%) patients received a 2nd treatment
(L2); median time from end L1 to beginning of L2 was 15.1 months (0-163). A 3rd line
(L3) was administered in 226 (22.4%) patients; median time from end of L2 to start
of L3 was 9 months (0-158); 125 (12.4%) patients received ≥4 treatments.
Between 1984 and 2021 the three most popular treatments received at L1 were: Dexamethasone-Rituximab-Cyclophosphamide
(DRC) 20.1% (144/690), R-Bendamustine (BR) 15.2% (105/690), Fludarabine based (±Cyclophosphamide,
Rituximab) 9.0% (62/690) [Figure 1].
The three most popular treatments at L2 were: Ibrutinib 24.9% (100/402), BR 9.5% (38/402),
DRC 5.0% (20/402). Ibrutinib is the leading 2nd, 3rd, 4th line treatment option from
2018 across all centres, due to its availability on the national Cancer Drugs Fund
from Nov 2017. The most received treatments from 2020 at: Ibrutinib 31.1% (60/193),
R-Bendamustine 12.4% (24/193), DRC 10.4% (20/193). 38 patients underwent autologous
and 7 allogeneic stem cell transplants between 2002 to 2020 at a median of L3 (range
1-7).
At the time of data lock, 184 patients were labelled deceased. Cause of death was
available for 78 patients: 15 due to WM, 7 patients high grade transformation, 3 amyloid,
3 during ASCT, 2 AML/MDS.
Image:
Summary/Conclusion: This RWD analysis of >1000 UK patients shows that inter/national
consensus guidelines are resulting in more streamlined treatment approaches in WM.
As targeted therapies have become available, there is a paradigm shift towards their
use; however there is still much to learn about the optimal selection and sequencing
of therapies in WM patients whose diseases are heterogenous by nature. Patient-related
outcome studies are underway to complement ongoing follow-up of clinically meaningful
remission duration and tolerance.
P1150: REAL WORLD DATA ON BORTEZOMIB-BASED THERAPY IN WALDENSTRÖM’S MACROGLOBULINAEMIA:
EFFECTIVE EVEN IN MULTIPLY TREATED PATIENTS INCLUDING PRIOR BTK-INHIBITORS
E. Uppal1,*, J. Khwaja1, A. Rismani1, C. Kyriakou1, S. D’Sa1
1Department of Haematology, University College London Hospitals NHS Foundation Trust,
London, United Kingdom
Background: Waldenström macroglobulinemia (WM) is an indolent lymphoma with a prolonged
disease course which typically follows a remitting and relapsing trajectory, eventually
leading to treatment resistance. Several treatment options exist including Bruton
tyrosine kinase inhibitors (BTKi), rituximab-containing regimens, and bortezomib-containing
regimens. Treatment selection is based on patient performance status, disease characteristics,
drug tolerability and availability.
Aims: To assess the effectiveness and tolerability of bortezomib-based regimens in
WM.
Methods: Data for patients who had Bortezomib-containing regimens between 2010 and
2021 from 6 centres in the United Kingdom were retrospectively reviewed. Data was
acquired from the WMUK Rory Morrison Registry. Research ethics approval was obtained.
Results: Thirty-four patients were identified: 32/34 had Bortezomib-containing regimens
once and 2/34 had >1 Bortezomib-containing regimens on separate occasions, giving
a total of 38 subcutaneous Bortezomib-containing regimens administered at bi-weekly
and weekly schedules. Median age was 62 years (37-87), with a median of 2 prior lines
of therapy (0-7), at a median duration of 49.6 months from date of WM diagnosis (0.7-422).
5 patients received a prior BTKi, with the Bortezomib regimen prescribed following
a median of 3 prior lines of therapy (2-5) in this group. Patients who were treated
at first line had elected for non-chemotherapy regimens. Median performance status
was 1 (0-2) in 23 evaluable patients. The median M-protein at initiation was 34.5g/l
(8-60) with bone marrow infiltration 70%, and haemoglobin 94g/l (88-107). A median
of 5 cycles (1-8) were delivered and 65% (13/20) received a dose of 1.6mg/m2 and 35%
(7/20) received 1.3mg/m2.
Grade (G) 1 to 2 neuropathy occurred in 19% (5/26) of evaluable patients but did not
result in treatment cessation in any case. Six of 25 (24%) needed a dose reduction,
the majority due to G1-2 neuropathy (67%; 4/6). Gastrointestinal disturbance occurred
in 12% (3/26) patients, 1 required admission with G4 diarrhoea and remaining cases
were G1.
Of 34 evaluable cases, major response rate (≥ PR) was 74% (5 CR, 6 VGPR, 14 PR). 62%
(8/13) of patients receiving 1.6mg/m2 achieved a major response and 86% (6/7) of those
who received 1.3mg/m2. Three of 5 patients who had prior BTKi achieved PR, 1 MR, 1
SD.
Two patients had treatment discontinued due refractory disease. The overall median
time to best response was 81 days from end of treatment. Six of 19 (26%) evaluable
patients achieved best response during therapy. Two patients died during treatment
due to infection (COVID; respiratory sepsis), not attributable to disease relapse.
Eighteen patients (60%; 18/30) had treatment after bortezomib regimens at a median
of 5.3 months (0-75) and are alive. Median follow up was 30 months (1-111). Twenty-one
evaluable patients (72%; 21/29) were alive at the end of follow up.
Image:
Summary/Conclusion: This retrospective real-world analysis shows that bortezomib-containing
regimens have utility in WM with effective major response rates even in those with
multiple prior lines of therapy and heavy marrow infiltration including BTKi failures.
Lower bortezomib doses are effective, and GI and neurotoxicity are manageable with
dose reductions but no treatment discontinuations in this real-world cohort indicating
an acceptable safety profile.
P1151: METABOLIC TUMOR VOLUME IMPROVES OUTCOME PREDICTION IN UNTREATED MANTLE CELL
LYMPHOMA
V. K. Vergote1,*, G. Verhoef1, A. Janssens1, F. S. Woei-a-jin2, A. Laenen3, T. Tousseyn4,
D. Dierickx1, C. M. Deroose5
1Hematology; 2General Medical Oncology, UZLeuven; 3Biostatistics and Statistical Bioinformatics
Center, KULeuven; 4Pathology; 5Nuclear Medicine, UZLeuven, 3000, Leuven, Belgium
Background: Mantle cell lymphoma (MCL) is a rare, incurable subtype of B-cell non-Hodgkin’s
lymphoma, with heterogenous clinical behavior ranging from indolent to very aggressive
disease course with rapid relapses and short survival.
Aims: The aim of this retrospective study is to analyze the prognostic value of baseline
[18F]FDG-PET/CT metabolic parameters in untreated MCL.
Methods: We retrospectively analyzed baseline [18F]FDG-PET/CT of patients in our institution
(University Hospitals Leuven, Belgium) with histopathological confirmed newly diagnosed
MCL between January 1st 2004 and 31st December 2020. Maximum standardized uptake value
(SUVmax), mean standardized uptake value (SUVmean), peak standardized uptake value
(SUVpeak), metabolic tumor volume (MTV) and total lesion glycolysis (TLG) were analyzed.
Lesion dissemination (Dmax) and lesion dissemination standardized to body surface
area (SDmax) were calculated. Univariate and multivariate analysis were performed
using Cox proportional hazard models. Kaplan Meier survival curves were constructed.
Results: We included 83 patients with a median age of 66 years. The median MTV was
141.5 ml and the median Dmax was 0.6 m. A median follow-up of 47 months was achieved.
In total 42 patients died. Higher MTV was an adverse factor for overall survival (OS)
(p=0.0137; hazard ratio (HR): 1.48; 95% confidence interval (CI): 1.08-2.03), progression-free
survival (PFS) (p=0.0106; HR: 1.41; 95% CI: 1.08-1.84) and disease-specific survival
(DSS) (p=0.0366; HR: 1.56; 95% CI: 1.03-2.36). Furthermore, higher age at diagnosis,
MCL International Prognostic Index-score (MIPI) and Eastern Cooperative Oncology Group
Performance status (ECOG PS) were adverse factors for OS, PFS and DSS. We found no
significant correlation between Dmax, SDmax and any of the outcomes considered. In
multivariate analysis accounting for clinical characteristics, MTV was significantly
associated with DSS (p=0.0072; HR: 1.53; 95% CI: 1.12-2.08), but not with OS and PFS.
OS in the patients with MTV above the median (high, ≥141 ml) was significantly worse
compared to the group with low MTV (<141 ml) (median OS 6.1 years versus 7.6 years;
p=0.0077). Furthermore, there was a significant difference in PFS (median 2.9 versus
5.0 years; p=0.0055) and DSS (median 7.3 years versus not reached; p=0.0219) between
the groups with high and low MTV. MTVspleen was an adverse factor for OS (p=0.0103;
HR: 1.36; 95% CI: 1.08-1.72) and PFS (p=0.0019; HR: 1.40; 95% CI: 1.13-1.72) in multivariate
analysis. Ann Arbor stage was not withheld as a significant factor for outcome prediction
in univariate nor multivariate analysis.
Summary/Conclusion: MTV is an important prognostic tool and can further improve patient
risk stratification at staging of untreated MCL. We found no correlation between lesion
dissemination (Dmax, SDmax) on [18F]FDG-PET/CT and outcome.
P1152: RESULTS FROM A PHASE I PHARMACOKINETIC (PK) AND SAFETY STUDY OF TRPH-222, A
NOVEL CD22-TARGETING ANTIBODY-DRUG CONJUGATE, IN PATIENTS WITH RELAPSED/REFRACTORY
B-CELL NON-HODGKIN LYMPHOMA (R/R NHL)
F. J. Hernandez-Ilizaliturri1, J. Kuruvilla2, B. A. Christian3, I. W. Flinn4, S. E.
Assouline5, M. L. Ulrickson6, D. J. Landsburg7, M. Stuart8, H. Lowman8, N. Levin8,
D. Maetzel9, N. N. Viller9, A. MacLaren8,*
1Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, United States
of America; 2The Princess Margaret Hospital, Toronto, ON, Canada; 3The Ohio State
University, Columbus, OH; 4Sarah Cannon Research Institute/Tennessee Oncology, Nashville,
TN, United States of America; 5Division of Hematology, Sir Mortimer B. Davis-Jewish
General Hospital, Department of Oncology, McGill University, Montreal, QC, Canada;
6Banner MD Anderson Cancer Center, Gilbert, AZ; 7Abramson Cancer Center, University
of Pennsylvania, Philadelphia, PA; 8Triphase Accelerator, La Jolla, CA, United States
of America; 9Triphase Accelerator, Toronto, ON, Canada
Background: TRPH-222 is a novel antibody-drug conjugate (ADC) comprised of a humanized
anti-CD22 monoclonal antibody and maytansine, a potent microtubule inhibitor, joined
by a novel third-generation linker-conjugation technology (SMARTag®). This approach
enables site-specific conjugation of the maytansine payload while tightly controlling
drug:antibody ratio (DAR), resulting in a highly stable anti-CD22 ADC with a non-cleavable
linker designed to widen the therapeutic window.
Aims: The primary objectives of this first in human study are to determine the safety,
tolerability and pharmacokinetics (PK) of TRPH-222 monotherapy in patients with R/R
NHL.
Methods: TRPH-222-100 is an open-label, multicenter study comprised of dose-escalation
and dose-expansion stages. TRPH-222 was administered IV once every 3 weeks. 22 patients
were enrolled in dose-escalating cohorts of TRPH-222 (0.6 mg/kg to 10 mg/kg) from
DLBCL, FL, TFL, MCL and MZL histologies, and 10 patients in a dose-expansion cohort
(7.5 mg/kg) focusing on DLBCL and FL histologies.
Results: As of January 7, 2022, 32 NHL patients have been enrolled: 15 indolent (14
FL and 1 MZL) and 17 aggressive histologies (15 DLBCL, 1 TFL and 1 MCL). Patients
had a median age of 64.5 years, a median of 4 prior lines of therapy including 7 patients
receiving prior CAR-T treatment.
Three DLTs occurred in 2 patients during the study and comprised Grade 3 and 4 transaminase
elevations; one each at 4.2 and 10 mg/kg and one Grade 3 thrombocytopenia at 4.2 mg/kg.
Treatment-related serious adverse events (AEs) occurred in 2 patients (6.3%), caused
by thrombocytopenia and pyrexia (both at 7.5 mg/kg TRPH-222). AEs were more prevalent
at doses ≥7.5 mg/kg and less prevalent at lower doses. There was a trend to higher
grade AEs in patients with aggressive histologies, compared to indolent ones. The
most frequent (≥ 5%) treatment-emergent related AEs (Grade ≥3) included thrombocytopenia
(34%), neutropenia (22%), ALT/AST elevation (6%), dry eye (6%) and blurred vision
(6%). Cytopenias were non-febrile, infrequent, asymptomatic and resolved without significant
intervention. Ocular findings were consistent with known epithelial keratopathy of
ADCs and were generally low grade and resolved to ≤ Grade 1 with dose interruptions
and/or reductions. Overall, TRPH-222 demonstrated a favourable safety profile with
most AEs being predominantly low grade, tolerable, easily managed and reversible.
Preliminary efficacy results suggest evidence of anti-tumor activity, most notably
in patients with R/R FL. Of the 13 response-evaluable FL patients, 4 complete responses
(CR) and 2 partial responses (PR) were observed, with an overall response rate (ORR)
of 46% and a complete response rate (CRR) of 31%. Four patients with metabolic CRs
maintained these CRs for long periods off treatment; 3 patients remain in CR with
responses maintained for up to 25 months. Responses were generally early, durable
and CRs were maintained off-therapy. Beyond FL, CRs were also observed in 1 DLBCL
patient and in 1 MCL patient.
Summary/Conclusion: TRPH-222 was found to be well tolerated at higher dose levels
than evaluated for other ADCs. TRPH-222 monotherapy resulted in robust and durable
CRs in FL across dose levels where patients were able to discontinue TRPH-222 while
remaining in remissions. Collectively, these characteristics of TRPH-222 are favorable
for further development in the indolent lymphoma setting either as monotherapy or
in combination with other anti-tumor agents in B-cell lymphoma patients.
P1153: LONG-TERM SURVIVAL OUTCOMES OF PATIENTS WITH PRIMARY OCULAR ADNEXAL MALT LYMPHOMA:
A LARGE SINGLE-CENTER COHORT STUDY
Y. Liang1, R.-Y. Fu1, X. Li1, Y.-S. Piao2, J.-M. Ma3, L. Wang1,*
1Hematology; 2Pathology; 3Eye center, Beijing Tongren Hospital, Capital Medical University,
Beijing, China
Background: Primary ocular adnexal extranodal marginal zone mucosa-associated lymphoid
tissue lymphoma (OAML) is a rare subtype of non-Hodgkin’s lymphoma, and no consensus
has been defined concerning the optimal treatment strategies.
Aims: This study aims to investigate the associations of disease characteristics and
different treatments with long-term outcomes of patients with localized OAML.
Methods: A large retrospective cohort study was conducted in a single-center of China,
and 166 patients with newly diagnosed primary localized OAML between April 2008 and
August 2021 were enrolled in our analysis. Detailed data of disease characteristics
at diagnosis and treatments were collected for all patients, and treatment response
was evaluated at one month after therapy and every six months thereafter. We compared
treatment response and progression-free survival (PFS) among patients with different
characteristics and treatments.
Results: Of the 166 patients, 52 received complete resection of neoplasm, whereas
114 had residual lesion after surgery. Among the 114 patients, 61 underwent watchful
waiting and 53 received further treatment including localized radiotherapy, chemotherapy
(including immunochemotherapy), or combined radiotherapy and chemotherapy. Median
follow-up of these patients was 49 months (range: 2-156). A total of 31 patients had
disease progression or relapse during follow-up, including four patients with such
event more than five years after initial treatment. The 5-year PFS was 73.9%, 70.6%
and 85.9%, whereas the 10-year PFS was 69.3%, 59.2% and 79.3%, among patients with
complete resection of neoplasm, patients in the watchful waiting group and patients
with further treatment, respectively. Patients with further treatment had longer PFS,
compared with patients in the watchful waiting group (P=0.011), while no difference
in PFS was noted between patients with further treatment and patients with complete
resection of neoplasm (P=0.127). Bilateral involvement at diagnosis was associated
with significantly inferior PFS (P=0.029), whereas age, IPI score, or TNM staging
was not associated with PFS. No serious adverse reaction was reported among patients
with further treatment.
Image:
Summary/Conclusion: Bilateral involvement was associated with poor prognosis. Among
patients with residual lesions after surgery, further treatment was associated with
improved survival. Patients with OAML might experience disease progression or relapse
more than five years after initial treatment.
P1154: STUDY ZILO-301: A PHASE 3, RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED, MULTICENTER
STUDY OF ZILOVERTAMAB PLUS IBRUTINIB VS IBRUTINIB IN PATIENTS WITH RELAPSED OR REFRACTORY
MANTLE CELL LYMPHOMA
S. Yazji1,*, S. Hamburger1, Y. Wang1, S. Yavrom1, A. Pietrofeso1, R. Bliss2, J. B.
Breitmeyer1, M. Dreyling3, M. Wang4
1Oncternal Therapeutics, Inc., San Diego, CA; 2Veristat, LLC, Southborough, MA, United
States of America; 3University of Munich, Munich, Bavaria, Germany; 4Department of
Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston,
TX, United States of America
Background: Zilovertamab (ZILO) is a humanized monoclonal antibody that inhibits the
tumor promoting activity of ROR1 and has demonstrated additive/synergistic activity
with anti-cancer agents including ibrutinib (Ibr). In the Phase 1/2 Study CIRM-0001,
ZILO in combination with Ibr was well tolerated and demonstrated promising efficacy
in heavily pretreated, relapsed and refractory (R/R) patients (pts) with mantle cell
lymphoma (MCL). As of 1 Oct 2021, 26 evaluable pts were enrolled, the objective response
rate (ORR) was 80.8 % of which 34.6% achieved complete response (CR), with a median
progression free survival (mPFS) of 35.9 months (mos). The rationale for the novel
enrichment design (Figure 1) of Study ZILO-301 is (1) there is an unmet medical need
for pts with R/R MCL that only achieve a partial response (PR) or have stable disease
(SD) on Ibr, (2) there is an early opportunity for marketing authorization based upon
an accepted surrogate endpoint (namely ORR), and (3) a confirmatory study for regular
approval is built into the study design based on PFS. The timeframe from initial accelerated
marketing authorization to regular approval is shorter than historical approaches
for this patient population. Additionally, in Study ZILO-302, the safety and efficacy
of ZILO rescue therapy can be evaluated in pts that progress early on Ibr in Study
ZILO-301.
Aims: To evaluate the efficacy and safety of ZILO plus Ibr vs Ibr in R/R MCL pts.
Methods: Study ZILO-301 is a Phase 3 randomized, double-blind, placebo-controlled,
multicenter study under which pts with R/R MCL will initially receive single agent
Ibr (560 mg daily). After 4 mos, pts who only have PR or SD will be randomized (1:1)
to receive an infusion of ZILO or matching placebo (Pbo) and continue to receive Ibr.
Study ZILO-301 key inclusion criteria: Aged ≥18 years, confirmed diagnosis of MCL,
received ≥ 1 prior treatment for MCL (other than Bruton’s tyrosine kinase inhibitor),
have measurable disease, and signed informed consent. Key exclusion criteria: History
or current CNS involvement, significant cardiovascular disease, prior stem cell transplant
requiring immunosuppressive therapy, or graft versus host disease. Efficacy assessments
will be evaluated by a Blinded Independent Central Review Committee utilizing Lugano
Classification (Cheson 2014).
Results: Three analysis timepoints are planned. Analysis 1 is a futility assessment
focused on ORR. Analysis 2 is an analysis for potential early marketing authorization
(using accelerated assessment pathways by FDA and EMA), based upon ORR, duration of
response (DOR), and safety. This analysis is expected to occur approximately 2 years
after the first pt is enrolled in Study ZILO-301. Regardless of the Analysis 2 result,
the study will proceed without modification. Analysis 3 is the end of study analysis
for the primary endpoint of PFS and secondary endpoints of CR rate, duration of CR,
time to CR on therapy, OS, proportion of pts experiencing Grade ≥ 3 neutrophil or
platelet decrease, and overall safety. Regular marketing approvals would be sought
based on Analysis 3. Figure 1 provides a schematic and subject flow for Study ZILO-301.
Study ZILO-302 data may also support additional labeling for the combination of ZILO
plus Ibr.
Image:
Summary/Conclusion: Study ZILO-301, a phase 3, multinational, double-blind, placebo-controlled
study is open for enrollment in June 2022.
P1155: BREXUCABTAGENE AUTOLEUCEL FOR RELAPSED/REFRACTORY MANTLE CELL LYMPHOMA IN ROUTINE
PRACTICE: UPDATED REPORT FROM THE US LYMPHOMA CAR T CONSORTIUM
Y. Wang1,*, P. Jain2, F. Locke3, M. Maurer1, M. Frank4, J. Munoz5, S. Dahiya6, A.
Beitinjaneh7, M. Jacobs8, J. Mcguirk9, J. Vose10, A. Goy11, C. Andreadis12, B. Hill13,
K. Dorritie14, O. Oluwole15, A. Deol16, B. Shah3, J. Paludo1, T. Wang7, R. Banerjee12,
S. Neelapu2, D. Miklos4, A. Rapoport6, L. Lekakis7, A. Ghobadi8, Y. Lin1, M. Wang2,
M. Jain3
1Mayo Clinic, Rochester; 2The University of Texas MD Anderson Cancer Center, Houston;
3Moffitt Cancer Center, Tampa; 4Stanford University Medical Center, Stanford; 5Mayo
Clinic, Phoenix; 6University of Maryland School of Medicine, Greenebaum Comprehensive
Cancer Center, Baltimore; 7University of Miami Miller School of Medicine, Sylvester
Comprehensive Cancer Center, Miami; 8Washington University School of Medicine, Siteman
Cancer Center, St Louis; 9University of Kansas Medical Center, Kansas City; 10University
of Nebraska Medical Center, Buffett Cancer Center, Omaha; 11John Theurer Cancer Center,
Hackensack Meridian Health, Hackensack; 12University of California San Francisco,
San Francisco; 13Cleveland Clinic, Cleveland; 14UPMC Hillman Cancer Center, Pittsburg;
15Vanderbilt-Ingram Cancer Center, Nashville; 16Wayne State University, Karmanos Cancer
Institute, Detroit, United States of America
Background: The ZUMA-2 trial and real-world studies showed high efficacy of brexucabtagene
autoleucel (brexu-cel) in relapsed/refractory mantle cell lymphoma (MCL). We previously
reported an objective response rate (ORR) of 89% in 95 patients receiving standard-of-care
brexu-cel in the US Lymphoma CAR T Consortium (Wang et al, ASH 2021).
Aims: To update the safety and efficacy results of the Consortium study with more
patients and longer follow-up.
Methods: Patients who underwent leukapheresis for standard-of-care brexu-cel manufacture
at one of the 16 Consortium centers between 8/1/2020 and 12/31/2021 were included.
Baseline characteristics, bridging therapy, cytokine release syndrome (CRS), immune
effector cell-associated neurotoxicity syndrome (ICANS), and treatment outcome data
were collected. Eligibility for ZUMA-2 was retrospectively determined based on characteristics
at leukapheresis. Time-to-event data were analyzed using the Kaplan-Meier method.
Results: At the data cut-off date of 1/31/2022, 189 patients underwent leukapheresis;
167 (88%) completed infusion and 22 (12%) did not (manufacture failure n=7, death
n=6, disease progression n=5, organ dysfunction n=2, complete response [CR] to bridging
therapy n=1, patient decline n=1).
The median age of the 167 infused patients was 67 years (range 34-89) and 76% were
male. 16% had high risk simplified MIPI, 57% had Ki-67 ≥50%, 41% had blastoid or pleomorphic
variant, 49% had TP53 alteration, 31% had complex karyotype, 14% had bulky disease
(≥10 cm), and 10% had CNS involvement. The median prior lines of therapy was 3 (range
1-10), and 86% were BTKi-exposed (89% of which were refractory). 130 (78%) patients
would not have met ZUMA-2 eligibility criteria, and the most common reasons were prior
therapies (eg, anthracycline- or bendamustine-naïve [16%], BTKi-naïve [14%], >5 lines
of prior therapy [11%]), renal dysfunction (21%), cardiac comorbidities (14%), cytopenias
(11%), ECOG PS ≥2 (11%), and CNS involvement (10%).
113 (68%) patients received bridging therapy, which included immunochemotherapy (n=45),
BTKi (n=45), venetoclax (n=24), radiotherapy (n=23), corticosteroid (n=18), and lenalidomide
(n=6). The median time from leukapheresis to lymphodepletion chemotherapy was 28 days
(range 17-140).
The rate of CRS was 90% (8% grade ≥3, 1 grade 5), and the rate of ICANS was 61% (32%
grade ≥3, 0 grade 5). Medications used to manage CRS and/or ICANS included tocilizumab
(76%), corticosteroid (68%), anakinra (16%), and siltuximab (3%). 20% of patients
required ICU admission, with a median stay of 3 days (range 1-12); 18% required vasopressors,
3% required mechanical ventilation, and 3% required dialysis.
Among 159 evaluable patients, the day 30 ORR was 89%, with 70% CR and 19% partial
response (PR). With continued follow-up, the best ORR was 89%, with 80% CR and 9%
PR. The best ORR/CR rate were 89%/77% for high Ki-67% (≥50%), 88%/79% for blastoid
or pleomorphic variant, 88%/73% for complex karyotype, 90%/72% for TP53 altered, 81%/75%
for CNS involved, 89%/79% for BTKi-exposed, 91%/83% for BTKi-naïve, and 89%/78% for
ZUMA-2 ineligible.
With a median follow-up of 5.6 months (range 0.2-15.3), the 6-month estimates for
duration of response, progression-free survival, and overall survival were 67% (95%
CI 57-75), 63% (95% CI 54-71) and 85% (95% CI 77-90), respectively.
Summary/Conclusion: This updated analysis demonstrated favorable safety and efficacy
results of brexu-cel in R/R MCL that are consistent with the initial report. Despite
78% of patients being ineligible for ZUMA-2, the responses, CRS, ICANS and survival
outcomes were comparable to ZUMA-2 data.
P1156: MAGNIFY PHASE 3B STUDY OF LENALIDOMIDE + RITUXIMAB (R2) FOLLOWED BY MAINTENANCE
IN RELAPSED/REFRACTORY INDOLENT NON-HODGKIN LYMPHOMA: COMPLETE INDUCTION PHASE ANALYSIS
F. Lansigan1,*, D. J. Andorsky2, M. Coleman3, A. Yacoub4, J. M. Melear5, S. R. Fanning6,
K. S. Kolibaba7, C. Reynolds8, G. S. Nowakowski9, M. Gharibo10, J. R. Ahn10, J. Li10,
M. J. Rummel11, J. P. Sharman12
1Dartmouth–Hitchcock Medical Center, Lebanon, NH; 2Rocky Mountain Cancer Centers,
US Oncology Research, Boulder, CO; 3Clinical Research Alliance Inc, Weill Cornell
Medicine, New York, NY; 4University of Kansas Cancer Center, Westwood, KS; 5Texas
Oncology — Austin, US Oncology Research, Austin, TX; 6Prisma Health, US Oncology Research,
Greenville, SC; 7US Oncology Research, Vancouver, WA; 8IHA Hematology Oncology Consultants
— Ann Arbor, Ypsilanti, MI; 9Mayo Clinic, Rochester, MN; 10Bristol Myers Squibb, Princeton,
NJ, United States of America; 11Justus-Liebig-Universität, Giessen, Germany; 12Willamette
Valley Cancer Institute and Research Center, US Oncology Research, Eugene, OR, United
States of America
Background: Patients with relapsed indolent NHL (iNHL) have limited standard treatment
options. Lenalidomide combined with rituximab (R2) has shown complimentary clinical
activity and is a tolerable regimen in both untreated and relapsed or refractory (R/R)
patients with iNHL (RELEVANCE: N Engl J Med 2018;379:934 and AUGMENT: J Clin Oncol.
2019;37:1188).
Aims: These analyses examine the MAGNIFY interim primary endpoint of overall response
rate (ORR; 1999 IWG) for induction R2 in efficacy-evaluable patients receiving ≥ 1
treatment and who have available baseline and post-baseline assessments.
Methods: MAGNIFY is a multicenter, phase 3b trial in patients with R/R follicular
lymphoma (FL) grades 1–3b, transformed FL (tFL), marginal zone lymphoma (MZL), or
mantle cell lymphoma (MCL; NCT01996865) exploring optimal lenalidomide duration. In
the induction phase, lenalidomide 20 mg PO on days 1–21 of a 28-day cycle + rituximab
IV at 375 mg/m2/week cycle 1 and then every 8 weeks starting with cycle 3 (R2) are
administered for 12 cycles. Patients with stable disease, partial response, or complete
response/complete response unconfirmed (CR/CRu) were randomized 1:1 to R2 vs rituximab
maintenance for 18 months. Data presented here are the complete analysis from the
induction phase in efficacy-evaluable patients with FL grades 1–3a or MZL (FL grade
3b, tFL, and MCL not included).
Results: As of March 5, 2021, 394 patients (318 [81%] FL gr1–3a; 76 [19%] MZL) were
enrolled. The median follow-up was 40.6 mo (range, 0.6–79.6). Median age was 66 y
(range, 35–91), 328 (83%) had stage III/IV disease, with a median of 2 prior therapies
(94% prior rituximab-containing). ORR was 71% (n = 279) with 42% (n = 164) CR/CRu
(Table). All patients have completed R2 induction (n = 232, 59%) or discontinued study
treatment (n = 162, 41%). 141 patients (36%) prematurely discontinued both lenalidomide
and rituximab, primarily due to adverse events (AEs) (n = 54, 14%) or progressive
disease (n = 42, 11%). The majority of patients who have completed induction have
been randomized and entered maintenance (n = 217). Median duration of response in
the induction period was not reached (95% CI, 43.9 mo–NR), and median progression-free
survival in the induction safety population (n = 393) was 50.5 mo (95% CI, 39.5–NR).
Efficacy results are reported in the table by histology subgroups (FL vs MZL), and
rituximab-refractory, double-refractory, and early relapse statuses. Most common all-grade
treatment emergent AEs (TEAEs) were 47% fatigue, 43% neutropenia, 37% diarrhea, 30%
nausea, and 30% constipation. Grade 3/4 AEs occurring in ≥ 5% of patients included
37% neutropenia (10 patients [3%] had febrile neutropenia), 8% leukopenia, 6% thrombocytopenia,
5% anemia, and 5% fatigue. TEAEs led to discontinuation of lenalidomide in 19% of
patients and rituximab in 12% of patients; reduction or interruption of lenalidomide
in 64% of patients; and to interruption of rituximab in 30% of patients (dose reduction
for rituximab was not allowed). Neutropenia was the most common TEAE leading to lenalidomide
discontinuation in 6% and reduction/interruption in 32%, and rituximab discontinuation
in 3%. Infusion-related reaction was the most common TEAE leading to rituximab interruption
in 8%.
Image:
Summary/Conclusion: These data represent complete analysis of all patients in the
induction phase of MAGNIFY which continue to support that R2 is active with a tolerable
safety profile in patients with R/R FL grade 1–3a and MZL, including rituximab-refractory,
double-refractory, and early relapse patients.
P1157: REAL-WORLD TREATMENT PATTERNS AND COMPARATIVE EFFECTIVENESS OF BRUTON TYROSINE
KINASE INHIBITORS IN PATIENTS WITH MANTLE CELL LYMPHOMA
B. Shah1,*, K. Yang2, A. Klink3, T. Liu2, T. Zimmerman2, A. Gajra3, B. Tang2
1H. Lee Moffitt Cancer Center & Research Institute, Tampa; 2Beigene USA, San Mateo;
3Cardinal Health, Dublin, United States of America
Background: Bruton tyrosine kinase inhibitor (BTKi) therapies, approved for relapsed
or refractory (R/R) mantle cell lymphoma (MCL), have not been comprehensively evaluated
in real world populations.
Aims: This study aimed to assess patient characteristics, treatment patterns and associated
outcomes in real world BTKi-treated MCL patients.
Methods: The retrospective multicenter chart review was conducted in the Cardinal
Health Oncology Provider Extended Network. EMR data were extracted for eligible patients
diagnosed with MCL who initiated any of the approved BTKi (ibrutinib, acalabrutinib,
zanubrutinib) from 2018 to 2021; patients enrolled in trials were excluded. Index
date was defined as the use of any of the BTKis. Patients were required to have 12-month
pre-index for medical history, and from index to last follow-up or death. Descriptive
analyses were conducted to assess demographic/clinical characteristics, MCL baseline
features, BTKi treatment patterns, adverse events (AE), and response rates by BTKi.
Multivariable logistic regression was performed to assess factors associated with
response and AE.
Results: The study cohort consisted of 300 MCL patients (59% male; 69% white); most
(64%) patients were covered by Medicare, 34% had commercial insurance. BTKis were
given mainly as monotherapy (93%) and in R/R setting (86%). Patients in zanubrutinib
group were significantly older (n = 100, median age = 71, range = 50-91) than patients
in ibrutinib (n = 100, median age = 69, range = 39-87) and acalabrutinib (n = 100,
median age = 70, range = 51-86) groups. Significantly fewer patients in the zanubrutinib
group had baseline Ann Arbor stage I-II (4%) than ibrutinib (10%) or acalabrutinib
(13%), while more zanubrutinib patients had presence of B symptoms (67%) than ibrutinib
(44%) or acalabrutinib (57%). Patients in the zanubrutinib group also had significantly
less with ECOG of 3+ (4%) compared to ibrutinib (8%) or acalabrutinib (6%). At BTKi
initiation, significantly more patients in zanubrutinib group (18%) had history of
atrial fibrillation than ibrutinib (1%) or acalabrutinib (5%). BTKis were given mainly
as second-line (86%) and as monotherapy (93%). Most patients were started at an on-label
BTKi dose. In zanubrutinib patients, the dose of 160 mg BID was more commonly administered
(64%) than 320 mg QD (31%). Multivariable regression reported a significant association
of age, gender, extranodal/splenic involvement, and timing of BTKi initiation with
response and AE (Table).
Image:
Summary/Conclusion: This study provides the first real world evidence on comparative
effectiveness of ibrutinib, acalabrutinib, and zanubrutinib in MCL patients. While
patients treated with zanubrutinib were older and had more complex MCL baseline features
at initiation, multivariable regression suggested a trend favoring zanubrutinib over
ibrutinib or acalabrutinib for both response and AE. Frontline initiation of BTKi
therapy was also associated with improved tolerability. Future real world studies
are needed to discern long-term outcomes.
P1158: REAL-WORLD TREATMENT PATTERNS AND ECONOMIC BURDEN OF PATIENTS WITH MARGINAL
ZONE LYMPHOMA
K. Yang1, T. Liu1, B. Tang1, B. Shah2,*
1Beigene USA, San Mateo; 2H. Lee Moffitt Cancer Center & Research Institute, Tampa,
United States of America
Background: Marginal zone lymphoma (MZL) is an indolent non-Hodgkin lymphoma that
is treatable, yet incurable with a remitting and relapsing disease course. Given its
disease rarity and underlying heterogeneity, MZL remains understudied with limited
real-world evidence on how current treatment pattern conform to clinical guidelines,
and the economic outcomes associated with current treatments.
Aims: This study aimed to assess real-world treatment patterns, costs, and healthcare
resource utilization (HRU) in US MZL patients.
Methods: A retrospective, observational study was conducted using the IBM MarketScan®
commercial and Medicare supplemental claims dataset (2017-2020). Newly diagnosed adult
MZL patients (≥18 years) continuously enrolled 6 month pre- and 3-month post-index
date, defined as the first diagnosis date, were included. Treatment regimens were
identified by line of therapy and mutually exclusively categorized as rituximab monotherapy
(R-mono), bendamustine + rituximab (BR), rituximab, cyclophosphamide, doxorubicin,
vincristine, and prednisone (R-CHOP), ibrutinib, or other. Descriptive analyses were
conducted to assess patient sociodemographic and clinical characteristics, and treatment
utilization patterns including the frequency, duration, and discontinuation of each
treatment regimen. Costs and HRU assessed included inpatient, outpatient, and pharmacy
visits per-patient-per-month (PPPM). Treatment regimens, costs, and hospitalizations
were examined overall, and by line of therapy. Multivariable logistic regression was
conducted to examine predictors of costs and HRU.
Results: Among the 2491 newly-diagnosed MZL patients (median age = 63 years; 52% male),
59% were commercially insured (median age = 57 years) and 41% in Medicare (median
age = 76 years). The most common comorbidities were hypertension (44%), diabetes (17%),
atrial fibrillation (AF; 16%), and gastroesophageal reflux disease (15%). Mean time
from diagnosis to treatment initiation was 223 days. A total of 1,781 (72%) patients
received first-line (1L), 518 (29%) patients received second-line (2L) and 239 (13%)
patients received third-line (3L) therapies. R-mono was the most common regimen across
both commercial and Medicare patients and all treatment lines (Table). R-CHOP and
BR were the second most used regimen in 1L with decreased use in 2L+. Ibrutinib was
used more in 2L+ setting but had the lowest 1L PPPM cost (median $2958.9) than other
regimens. Overall MZL patients had PPPM 4.6 outpatient visits, 0.5 hospitalization,
and mean length of stay of 2.6 days. Total PPPM healthcare cost was $19,895.8. Multivariable
regression showed that baseline comorbidities (AF, renal disease, neutropenia) and
treatment discontinuation were significant predictors of higher costs and HRU.
Image:
Summary/Conclusion: This real-world data suggested that the overall US MZL real-world
treatment pattern across lines of therapy follows the regimen recommendations by the
National Comprehensive Cancer Network clinical practice guidelines and that MZL patients
incur high economic burden. Future studies are needed to evaluate long-term outcomes
and the impact of heterogenous MZL subtypes.
P1159: A MULTICENTER, INTERNATIONAL COLLABORATIVE STUDY EVALUATING FRONTLINE THERAPY
WITH BENDAMUSTINE RITUXIMAB FOR WALDENSTRÖM MACROGLOBULINEMIA
S. Zanwar1,*, J. Abeykoon1, J. Castillo2, E. Durot3, E. Kastritis4, E. Uppal5, P.
Morel6, R. Tawfiq7, L. Montes8, J. Paludo1, S. Sarosiek2, S. Kumar1, O. Ogunbiyi9,
P. Cornillet-Lefebvre10, R. Kyle1, A. Delmer3, M. Gertz1, M. Dimopoulos11, S. Ansell1,
S. Treon2, S. D’Sa12, P. Kapoor1
1Hematology, Mayo Clinic, Rochester, Rochester; 2Hematology, Dana Farber Cancer Institute,
Boston, United States of America; 3Hematology, University Hospital of Reims and UFR
Médecine, Reims, France; 4Department of Clinical Therapeutics, National and Kapodistrian
University of Athens, Athens, Greece; 5Hematology, University College London Hospitals
(UCLH) NHS Foundation Trust, London, United Kingdom; 6Hematology, Hôpital Schaffner,
Lens, France; 7Internal Medicine, Mayo Clinic, Rochester, Rochester, United States
of America; 8Service d’Hématologie Biologique, CHU Amiens, Amiens, France; 9Hematology,
Imperial College of London, London, United Kingdom; 10Laboratory of Hematology, University
Hospital of Reims and UFR Médecine, Reims, France; 11Department of Clinical Therapeutics,
National and Kapodistrian University of Athens, Athens, Greece; 12Hematology, University
College London Hospitals, London, United Kingdom
Background: Bendamustine rituximab (BR) is a frequently used chemoimmunotherapy for
indolent lymphomas, including Waldenström macroglobulinemia (WM), a rare B-cell malignancy.
The promising results of a small subset analysis (n= 41) of the Study group indolent
Lymphomas (StiL) trial, demonstrating a median progression-free survival (PFS) of
69.5 months in the frontline setting served as the basis for widespread adoption of
BR. Whether the more recently identified somatic mutations within the MYD88 and CXCR4
genes impact the outcomes of patients (pts) treated with BR remains less clear.
Aims: To study a large cohort of BR-treated pts with active WM through an international,
multicenter collaborative effort.
Methods: Records of pts with newly diagnosed active WM who received BR between January
2012 and July 2021, in the US and Europe, were reviewed. The MYD88
L265P and CXCR4 mutation status were captured, if available. All time-to-event analyses
were performed from the frontline therapy initiation, using the Kaplan-Meier method.
Results: Among 248 pts who were treated with BR, 208 pts received BR induction without
rituximab maintenance, and were included in the primary analysis. The median age at
treatment initiation was 65 (range 40-86) years; 64 % were males. The baseline characteristics
are outlined in Table 1. The median follow-up was 4 (95% CI: 3.6-4.6) years.
The estimated median PFS was 5.9 years [95% CI: 5.3-not reached (NR)]. The estimated
5-year overall survival (OS) rate was 90%. Among 174 pts evaluable for response, the
overall response rate (ORR), major response rate (MRR) and very good partial response
(VGPR) rates were 95%, 93% and 31%, respectively, per the modified IWWM-6 criteria
based on serum IgM level alone. Pts with progression of disease (POD) within 24 months
of BR therapy (11%) had an inferior subsequent survival compared to those without
POD within 24 months, the reference group [5-year subsequent survival rate, 71 % versus
(vs) 86%, p=0.02].
Among 131 (63%) pts with a known MYD88
L265P status, 88% (n=116) had MYD88
L265P genotype. The 4-year PFS was 71% for both pts with MYD88
L265P and MYD88
WT genotypes (p=0.44), Figure 1A. The VGPR rates were also comparable between the
two groups (41% for MYD88
L265P and 50% for MYD88
WT genotypes, p=0.55). Among 42 (20%) pts with a known CXCR4 mutation status, 28%
harbored a CXCR4 mutation. The ≥VGPR rate for pts with CXCR4
MUT genotype was numerically lower; 33% vs 57% for those with CXCR4
WT genotype, p=0.3. A trend towards shorter PFS among pts with CXCR4
MUT genotype [estimated median PFS for 3.9 years (95% CI: 0.8-NR) vs 5.5 years (95%
CI 5.3-NR) for pts with CXCR4
WT genotype, p=0.05, Figure 1B] was observed. The PFS rates for pts without a known
MYD88 or CXCR4 mutational status was comparable to their counterparts with known genotypes,
respectively.
Among 40 (16%) pts who had received rituximab maintenance following BR, the median
age at initiation of therapy was 67 years (range 44-88). After 1:1 matching for age,
the 4-year PFS for the rituximab maintenance group was 89% vs 73% for pts who did
not receive rituximab maintenance (p=0.09); OS was comparable (5-year OS rate, 85%
for both groups, p=0.99).
Image:
Summary/Conclusion: Fixed-duration BR is a highly effective regimen for pts with previously
untreated symptomatic WM, irrespective of the MYD88
L265P mutation status, although early POD, within 2 years of initiation of BR, is
associated with inferior subsequent survival. Our preliminary analysis, suggesting
that the presence of CXCR4 mutation confers resistance to BR, warrants confirmation
in prospective studies.
P1160: RESULTS OF A PHASE 2 EXPANDED ACCESS STUDY OF ZANUBRUTINIB IN PATIENTS WITH
WALDENSTRÖM MACROGLOBULINEMIA
J. J. Castillo1,*, E. C. Kingsley2, M. Narang3, H. A. Yimer4, C. A. Dasanu5, J. M.
Melear6, M. Coleman7, C. M. Farber8, M. Gupta9, J. Shulman10, E. H. Mantovani11, X.
Zhang11, A. Cohen11, J. Huang11
1Dana-Farber Cancer Institute, Boston, MA; 2Comprehensive Cancer Centers of Nevada,
Las Vegas, NV; 3US Oncology Research, Maryland Hematology Oncology, Columbia, MD;
4Texas Oncology, US Oncology Research, Tyler, TX; 5Lucy Curci Cancer Center, Eisenhower
Health, Rancho Mirage, CA; 6US Oncology Research, Texas Oncology, Austin Midtown,
Austin, TX; 7Clinical Research Alliance, New York, NY; 8Atlantic Hematology Oncology,
Morristown Medical Center, Morristown, NJ; 9Ridley-Tree Cancer Center at Sansum Clinic,
Santa Barbara, CA; 10Icahn School of Medicine at Mount Sinai, New York, NY; 11BeiGene
(Beijing) Co., Ltd., Beijing, China and BeiGene USA, Inc., San Mateo, CA, United States
of America
Background: Bruton tyrosine kinase (BTK) inhibition is an emerging standard of care
for Waldenström macroglobulinemia (WM). Zanubrutinib (ZANU; BGB-3111) is a next-generation
BTK inhibitor designed to maximize BTK occupancy and minimize off-target inhibition
of TEC- and EGFR-family kinases. ZANU was recently approved by the United States (US)
Food and Drug Administration, Health Canada, and the European Union at a dose of 320 mg
once daily (QD) or 160 mg twice daily (BID) for the treatment of adult patients (pts)
with WM. BGB-3111-216 (NCT04052854) is a single-arm, expanded access study of ZANU
for treatment-naïve (TN) pts who are unsuitable for standard chemoimmunotherapy or
pts with relapsed refractory (R/R) WM.
Aims: To provide real-world experience with ZANU in pts with WM.
Methods: Eligible pts with TN or R/R WM were assigned to receive ZANU at a dose of
320 mg QD or 160 mg BID. The primary endpoint was the number of pts enrolled/treated
and the number of enrolling sites. Secondary endpoints of safety and efficacy included
treatment emergent adverse events (TEAEs) of special interest, disease response (overall
response rate [ORR] and very good partial response or better [VGPR+]), progression-free
survival (PFS), and overall survival (OS). Response was evaluated by investigator-assessment
according to the 6th International Workshop on WM (Br J Haematol. 2013;160(2):171-6)
every 6 months at minimum. The study was closed by the sponsor in July 2021 and active
pts were transitioned to commercial ZANU via a patient assistance program.
Results: Fifty pts with WM (17 TN; 33 R/R), were enrolled from December 2019 to June
2021 across 10 academic and community medical centers in the US. At study entry, median
age was 72 years, 54% had intermediate-risk disease, 40% had high-risk disease, and
median number of prior therapies for R/R pts was 2. Median treatment exposure was
9.2 months (range 1.4 to 20.0). Thirty-eight (76%) pts had ≥1 TEAE, and 36 (72%) experienced
≥1 TEAE of special interest (Table). Grade ≥3 TEAEs of special interest included hypertension
(8%), infection (8%), atrial fibrillation or flutter (2%), neutropenia (2%), and second
primary malignancy (2%). No new safety signals were observed. In the 41 pts with ≥1
response evaluation, 39.0% achieved a best overall response (BOR) of VGPR (16; 95%
CI, 24.2-55.5). ORR was 85.4% (35; 95% CI, 70.8-94.4), and major response rate was
73.2% (30; 95% CI, 57.1-85.8). Of the 4 pts who achieved a BOR of progressive disease,
3 had IgM values that met partial response criteria before the first 6 months response
assessment. PFS and OS were immature due to short follow-up, and the median was not
met.
Image:
Summary/Conclusion: The results of this real-world expanded access study were consistent
with the established ZANU profile in WM and other B-cell malignancies when administered
as monotherapy at a daily dose of 320 mg orally (either as 160 mg BID or 320 mg QD)
in pts with intermediate or high-risk R/R or TN WM.
P1161: ASPEN: LONG-TERM FOLLOW-UP RESULTS OF A PHASE 3 RANDOMIZED TRIAL OF ZANUBRUTINIB
(ZANU) VS IBRUTINIB (IBR) IN PATIENTS (PTS) WITH WALDENSTRÖM MACROGLOBULINEMIA (WM)
M. Dimopoulos1,*, S. Opat2, S. D’Sa3, W. Jurczak4, H.-P. Lee5, G. Cull6, R. G. Owen7,
P. Marlton8, B. E. Wahlin9, R. Garcia-Sanz10, H. McCarthy11, S. Mulligan12, A. Tedeschi13,
J. J. Castillo14, J. Czyz15, C. Fernandez De Larrea Rodriguez16, D. Belada17, E. Libby18,
J. Matous19, M. Motta20, T. Siddiqi21, M. Tani22, M. Trneny23, M. Minnema24, C. Buske25,
V. Leblond26, S. P. Treon14, J. Trotman27, W. Y. Chan28, J. Schneider28, H. Allewelt28,
A. Cohen28, J. Huang28, C. S. Tam29
1National and Kapodistrian University of Athens, Athens, Greece; 2Monash Health and
Monash University, Clayton, Victoria, Australia; 3Centre for Waldenström’s Macroglobulinemia
and Associated Disorders, University College London Hospital Foundation Trust, London,
United Kingdom; 4Maria Sklodowska-Curie National Institute of Oncology, Krakow, Poland;
5Flinders Medical Centre, Adelaide, SA; 6Sir Charles Gairdner Hospital, University
of Western Australia, Perth, WA, Australia; 7St. James University Hospital, Leeds,
United Kingdom; 8Princess Alexandra Hospital, University of Queensland, Brisbane,
Queensland, Australia; 9Karolinska Universitetssjukhuset and Karolinska Institutet,
Stockholm, Sweden; 10Hospital Universitario de Salamanca, Salamanca, Spain; 11Royal
Bournemouth and Christchurch Hospital, Bournemouth, United Kingdom; 12Royal North
Shore Hospital, Sydney, New South Wales, Australia; 13ASST Grande Ospedale Metropolitano
Niguarda, Milan, Italy; 14Dana-Farber Cancer Institute, Boston, MA, United States
of America; 15Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń,
Bydgoszcz, Poland; 16Hospital Clinic de Barcelona, Barcelona, Spain; 17FN Hradec Kralove,
Hradec Králové, Czechia; 18University of Washington/Seattle Cancer Care Alliance -
Clinical Research, Seattle, WA; 19Colorado Blood Cancer Institute, Denver, CA, United
States of America; 20AO Spedali Civili di Brescia, Lombardia, Italy; 21City of Hope
National Medical Center, Duarte, CA, United States of America; 22Ospedale Civile S.Maria
delle Croci, AUSL Ravenna, Italy; 23Vseobecna fakultni nemocnice v Praze, Prague,
Czechia; 24University Medical Center Utrecht, Utrecht, Netherlands; 25CCC Ulm - Universitätsklinikum
Ulm, Ulm, Baden-Württemberg, Germany; 26Sorbonne University, Pitié Salpêtrière Hospital,
Paris, France; 27Concord Repatriation General Hospital, Sydney, New South Wales, Australia;
28BeiGene USA, Inc., San Mateo, CA, United States of America; 29Royal Melbourne Hospital,
Parkville, Victoria, Australia
Background: ZANU is a potent and selective next-generation Bruton tyrosine kinase
inhibitor (BTKi) designed to have greater affinity to BTK while minimizing off-target
inhibition of TEC-and EGFR-family kinases. ASPEN (NCT03053440) is a randomized, open-label,
phase 3 study comparing ZANU with the first-generation BTKi IBR in pts with WM. We
present data with a median follow-up of 43 months.
Aims: To compare the efficacy and safety of ZANU vs IBR in pts with MYD88 mutant (MYD88
mut) WM and ZANU in pts with wild-type MYD88 (MYD88
wt) WM.
Methods: Pts with MYD88
mut WM were assigned to cohort 1 and randomized 1:1 to receive ZANU 160 mg twice daily
or IBR 420 mg once daily. Pts with MYD88
wt were assigned to cohort 2 and received ZANU 160 mg twice daily until disease progression.
Randomization was stratified by CXCR4 mutational status by Sanger sequencing and lines
of prior therapy (0, 1-3, or >3). All pts gave informed consent. The primary endpoint
was proportion of pts achieving very good partial response or better (VGPR + complete
response [CR]). Primary analysis occurred at 19 months median follow-up, and final
analysis is planned to occur ~4 years after the first pt enrolled.
Results: A total of 201 pts (102 ZANU; 99 IBR) were enrolled in cohort 1 and 28 pts
in cohort 2. Baseline characteristics in cohort 1 differed between pts treated with
ZANU vs IBR in CXCR4 mutations by next-generation sequencing (32% vs 20%, or 33 of
98 vs 20 of 92 available samples, respectively) and pts aged >75 years (33% vs 22%,
respectively). Median duration of treatment was 42 months (ZANU) and 41 months (IBR),
with 67% and 58% remaining on treatment, respectively. The VGPR+CR rate by investigator
was 36% with ZANU vs 22% with IBR (descriptive p = 0.02) in cohort 1, and 31% in cohort
2. One pt in cohort 2 obtained a CR. In pts with wild-type (65 ZANU; 72 IBR) or mutant
CXCR4 (33 ZANU; 20 IBR) from cohort 1, VGPR+CR rates with ZANU vs IBR were 45% vs
28% (p = 0.04) and 21% vs 5% (p = 0.15), respectively. Median progression-free survival
and overall survival were not reached.
Consistent with less off-target inhibition, rates of atrial fibrillation, diarrhea,
hypertension, localized infection, hemorrhage, muscle spasms, pneumonia, and adverse
events (AEs) leading to discontinuation or death were lower with ZANU vs IBR (Table);
neutropenia (including grade ≥3) was the only AE of interest that was higher with
ZANU (33.7%) vs IBR (19.4%). Rate of grade ≥3 infection was lower with ZANU (20.8%)
vs IBR (27.6%). AE incidence with ZANU was similar across cohorts 1 and 2.
Annual prevalence analysis of cohort 1 AEs showed reduced prevalence of hemorrhage
over time and lower prevalence with ZANU vs IBR at all intervals. In pts treated with
ZANU, neutropenia and infection prevalence decreased over time. Prevalence of infection
was lower in pts treated with ZANU vs IBR, and neutropenia was similar between arms
(8.8% vs 9.7%, respectively) at >24-36 months of treatment. Prevalence of atrial fibrillation
remained ≤5% and hypertension remained stable with ZANU, each with a lower prevalence
at all intervals vs an increasing trend seen with IBR.
Consistently, exposure-adjusted incidence rates of atrial fibrillation/flutter and
hypertension were lower with ZANU vs IBR (0.2 vs 0.8 and 0.5 vs 1.0 persons per 100
person-months, respectively; p < 0.05).
Image:
Summary/Conclusion: ASPEN is the largest phase 3 trial with head-to-head BTKi comparison
in WM. At a median follow-up of 43 months, ZANU was associated with a higher VGPR+CR
rate and demonstrated clinically meaningful advantages in long-term safety and tolerability
vs IBR.
P1162: ZANUBRUTINIB IN OLDER PATIENTS (PTS) WITH RELAPSED/REFRACTORY (R/R) MARGINAL
ZONE LYMPHOMA (MZL): SUBGROUP ANALYSIS OF THE MAGNOLIA STUDY
S. Opat1,*, B. Hu2, A. Tedeschi3, K. M. Linton4, P. McKay5, H. Chan6, J. Jin7, M.
Sun8, M. Sobieraj-Teague9, P. L. Zinzani10, M. Coleman11, P. Browett12, X. Ke13, C.
A. Portell14, C. Thieblemont15, K. Ardeshna16, F. Bijou17, P. Walker18, E. A. Hawkes19,
S.-J. Ho20, K.-S. Zhou21, M. Co22, J. Xu22, Z. Liang22, J. Anderson22, C. Tankersley22,
J. Huang22, J. Trotman23
1Monash Health and Monash University, Clayton, Victoria, Australia; 2Levine Cancer
Institute/Atrium Health, Charlotte, NC, United States of America; 3ASST Grande Ospedale
Metropolitano Niguarda, Milan, Italy; 4The Christie Hospital NHS Foundation Trust,
Manchester; 5Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom; 6North
Shore Hospital, Auckland, New Zealand; 7The First Affiliated Hospital, Zhejiang University,
Hangzhou, Zhejiang; 8Institute of Hematology & Blood Diseases Hospital, Chinese Academy
of Medical Sciences and Peking Union Medical College, Tianjin, China; 9Flinders Medical
Centre, Bedford Park, South Australia, Australia; 10Institute of Hematology “Seràgnoli”
University of Bologna, Bologna, Italy; 11Clinical Research Alliance/Weill Cornell
Medicine, Lake Success, NY, United States of America; 12Auckland City Hospital, Grafton,
New Zealand; 13Peking University Third Hospital, Beijing, China; 14University of Virginia
Health System, Charlottesville, VA, United States of America; 15APHP, Hôpital Saint-Louis,
Hemato-oncology, Paris University Diderot, Paris, France; 16University College London
Hospitals, London, United Kingdom; 17Institut Bergonié, Bordeaux, France; 18Peninsula
Private Hospital, Frankston, Victoria; 19Box Hill Hospital, Box Hill, Victoria; 20St.
George Hospital, Kogarah, New South Wells, Australia; 21Henan Cancer Hospital, Zhengzhou,
Henan, China; 22BeiGene (Beijing) Co., Ltd., Beijing, China and BeiGene USA, Inc.,
San Mateo, CA, United States of America; 23Concord Repatriation General Hospital,
University of Sydney, Concord, New South Wales, Australia
Background: MZL is the second most common lymphoma in older pts. Choosing an optimal
treatment can be challenging because of patient- or disease-related risk factors and
treatment-related toxicities (Curr Opin Oncol. 2019;31(5):386-393). Zanubrutinib is
a potent, irreversible next-generation Bruton tyrosine kinase (BTK) inhibitor designed
to maximize BTK occupancy and minimize off-target kinase inhibition, which may improve
efficacy outcomes and minimize toxicities, such as cardiac arrythmias and bleeding
events. Zanubrutinib received accelerated approval from the United States FDA for
the treatment of pts with R/R MZL (Haematologica. 2022;107(1):35-43).
Aims: We aim to present a subgroup analysis of efficacy and safety of zanubrutinib
in pts aged ≥65 years with R/R MZL enrolled in MAGNOLIA (BGB-3111-214; NCT03846427).
Methods: MAGNOLIA is a phase 2, multicenter, single-arm study of adults with R/R MZL
who had received ≥1 line of therapy including ≥1 CD20-directed regimen. All were treated
with zanubrutinib 160 mg twice daily until disease progression or unacceptable toxicity.
Use of long-term antiplatelet and anticoagulation agents was permitted. The primary
endpoint was overall response rate (ORR; complete response [CR] and partial response
[PR]) determined by an independent review committee (IRC) in accordance with the Lugano
classification. Secondary endpoints include ORR by investigator assessment (INV),
duration of response (DOR), progression-free survival (PFS), and safety. All pts gave
informed consent.
Results: As of 18 January 2021, a total of 68 pts were enrolled (Table). Forty (61%)
pts were ≥65 years old with a median age of 73 (range, 65-85); 18 pts were ≥75 years
old. Median number of prior therapies was 2 (range, 1-6) and 10 (25%) pts were refractory
to last therapy. Most pts received prior rituximab + cyclophosphamide + vincristine
+ prednisone (48%) or bendamustine + rituximab (30%), while 5 (13%) pts received rituximab
monotherapy. MZL subtypes included extranodal (n=17, 43%), nodal (n=14, 35%), and
splenic (n=8, 20%). Median duration of treatment was 14.4 months (mo; range, 0.9-19.6).
At a median follow-up of 15.8 mo (range, 2.8-21.8), ORR by IRC was 75% (CR 25%, PR
50%; Table). Responses were observed in all subtypes, with an ORR of 71%, 86%, and
75% in extranodal, nodal, and splenic subtypes, respectively (CR 41%, 21%, and 0%,
respectively). Median DOR and PFS were not reached; 15-month PFS was 87% and 12-month
DOR was 93%. Most (63%) pts are continuing zanubrutinib. Treatment discontinuation
due to disease progression was 28% by INV. Most common treatment-emergent adverse
events (AEs) observed in ≥20% of pts include contusion (28%), diarrhea (25%), and
constipation (20%). Grade ≥3 neutropenia occurred in 5% of pts. The most common infection
was upper respiratory tract infection (10%). Two (5%) pts discontinued zanubrutinib
due to unrelated fatal AEs (COVID-19 pneumonia and myocardial infarction in a patient
with pre-existing coronary artery disease). Atrial fibrillation/flutter and hypertension
occurred in 2 (5%) pts each and did not lead to treatment discontinuation. No pts
required dose reductions, or experienced major or serious hemorrhage.
Image:
Summary/Conclusion: The safety profile of zanubrutinib observed in older pts was consistent
with previously published results (Clin Cancer Res. 2021;27(23):6323-6332). Zanubrutinib
was well tolerated and effective, as demonstrated by a high response rate and durable
disease control in older pts with R/R MZL.
P1163: OUTCOMES OF HIGH-RISK LARGE B-CELL LYMPHOMA TREATED WITH STANDARD OF CARE CHEMOIMMUNOTHERAPY:
A NON-RANDOMIZED COMPARISON TO FRONTLINE CAR-T CELL THERAPY
O. Albanyan1,*, O. Castaneda-Puglianini1, J. Chavez2
1Blood and Marrow Transplant and Cellular Immunotherapy; 2Department of Malignant
Hematology, Moffitt Cancer Center, Tampa, United States of America
Background: High-risk LBCL is associated with poor prognosis after standard of care
(SOC) first-line anti-CD20 mAb-containing chemoimmunotherapy (CIT), therefore here
is a need for novel approaches. ZUMA-12 reported high overall response (ORR) and complete
response (CR) rates in high risk LBCL defined as IPI score of ≥3 and/or double or
triple-hit status [MYC and BCL2 and/or BCL6 translocations (double-hit/triple-hit
lymphoma – DHL/THL)] and positive interim PET scan (iPET+)
Aims: Here, we report the outcomes of these high risk LBCL patients (pts) who did
not qualify due to a negative (-) iPET or declined or were ineligible to be treated
in the ZUMA-12 clinical trial.
Methods: Eligible patients were older than 18 and had high-risk LBCL, defined as DHL/THL
status and/or an IPI score ≥3. Patients could have had an available iPET (or CT scan).
Patients did not pursue ZUMA-12 due to trial ineligibility (i.e., iPET-) or did not
want to pursue the trial. Patients continued SOC CIT per discretion of treating physician.
The primary endpoint was end of treatment ORR and CR. Secondary endpoints included
progression-free survival (PFS), overall survival (OS).
Results: During the time period ZUMA-12 was open, 61 pts were approached for the trial,
8 patients were excluded from the final analysis due to incomplete data. The remaining
53 pts did not qualify to ZUMA-12 due to iPET- (n=31, 58.4%), declined (n=10, 18.8%)),
ineligible (n= 8, 15.1%) and other (n= 4). Median age was 67 (26 – 78), 11 (20.1 %)
had DHL/THL, 46 (86.8 %) had IPI score 3-5. Patients were treated with DA-EPOCH-R
(n=11, 21%) and R-CHOP (n=41, 77%). The ORR and CR rates was 83 and 79.2%, respectively
in this cohort. Four patients did not complete therapy due to PD (n=3) and death for
other causes (n=1). The ORR and CR rates for the ZUMA-12 study were reported at 90
and 80%, respectively. In our study, the median PFS and OS was 27.4 months (95% CI:
23.7 – 31.1) and 30.8 months (95% CI: 27.7 – 33.8), and the 30-months PFS and OS were
60 and 71%, respectively. In comparison, the ZUMA-12 reported a PFS and OS that was
not reached.
Summary/Conclusion: In this nonrandomized study of high risk LBCL, the efficacy of
axi-cel in the ZUMA-12 was numerically higher than SOC CIT. The PFS and OS in our
cohort was lower despite majority of patients achieving early CR per iPET. This analysis
supports a potential trial of frontline CAR-T versus SOC CIT for high risk LBCL.
P1164: IMAGE-BASED DETECTION OF HIGH-GRADE B CELL LYMPHOMAS (DH-L)
I. Avivi1,2,*, C. Perry1,2, O. Ahron2,3, N. Hershkovitz4, D. Hershkovitz2,5, A. Avinoam6,
I. Gazy6, N. Paz-Yaacov6
1Hematology department, Tel Aviv Sourasky Medical Center; 2Sackler Faculty of Medicine.,
Tel Aviv University; 3Pathology department; 4Hematology department, Tel Aviv Medical
Center; 5Pathology department, Tel Aviv Sourasky Medical Center; 6Imagene AI, Tel
Aviv, Israel
Background: Aggressive B cell Non-Hodgkin Lymphomas (B-NHL) are divided into two main
categories: diffuse large B cell lymphoma (DLBCL) accounting for 90% of cases, and
high-grade B-cell lymphoma (HGBL). Diagnosis of high-grade lymphoma with MYC and BCL2
and/or BCL6 rearrangements (double-hit lymphoma-DHL) is confirmed by fluorescence
in situ hybridization (FISH) analysis, demonstrating c-MYC-rearrangement in combination
with BCL-2 / BCL-6 rearrangement in lymphoma cells. Accurate and rapid diagnosis of
DHL is obligatory, when considering more aggressive treatment regimens (other than
R-CHOP), suggested in these patients.
Aims: To establish a novel tool for diagnosing DLBCL and DHL, directly on the scanned
Hematoxylin and Eosin (H&E) biopsy slides, by applying digital imaging technologies
supported by machine learning algorithms.
Methods: H&E whole slide images, prepared from biopsies obtained mainly from lymph
nodes, but also from extra-nodal organs of patients with aggressive B cell lymphoma
histology, were collected from the pathology department at Tel-Aviv Medical center
(TASMC). Cases were randomly divided into a training (n=43) and a validation (n=35)
set. On-the-fly augmentation was applied to images, together with advanced Convolutional
Neural Network (CNN) analysis, to generate the aggressive B-NHL classifier (powered
by Imagene-AI). Proprietary multiple instance learning (MIL) algorithms and training
scheme, based on cases ranking, rather than absolute values, were applied. The classifier
was validated through a testing set in a blinded study scheme, and results were compared
to the FISH results for c-MYC and BCL2/6 rearrangements (Figure 1A).
Results: The classifier training set was composed of 36 DLBCL NOS and 7 DHL cases.
The aggressive B-NHL classifier was validated on a cohort of 35 cases, including 14
DHL cases (diagnosed by FISH) and 21 DLBCL cases. The model demonstrated 100% sensitivity
and 90.48% specificity, with an accuracy rate of 94.29% and Area Under the Curve (AUC)
of 0.99 (Figure 1B). Two cases were concluded as false positive, including one case
that demonstrated rearrangements in substantial percentages of cells, just below the
required positivity threshold.
Image:
Summary/Conclusion: Herein, we demonstrate for the first time a machine learning-based
genomic testing solution for the detection and classification of aggressive B-NHL,
based on H&E stained slide images. Implementation of this solution in clinical practice
can support the diagnosis of DHL, which currently has both technical and financial
challenges. Applying the proposed accessible and standardized AI-based method, can
improve and facilitate the diagnosis of DHL, thereby directly improving patient care.
P1165: BETALUTIN® IN PATIENTS WITH RELAPSED/ REFRACTORY DIFFUSE LARGE B-CELL LYMPHOMA
(DLBCL) NOT ELIGIBLE FOR AUTOLOGOUS STEM CELL TRANSPLANT
T. Illidge1,*, M. Beasley2, J. G. Codina3, F. Cavallo4, V. Pascal5, V. Wills6
1Haematology/Oncoloy, The Christie NHS Foundation Trust, Withington; 2Haematology/Oncoloy,
Bristol Haematology and Oncology Centre, Bristol, United Kingdom; 3Hospital Universitari
i Politècnic La Fe, Hospital Universitari i Politècnic La Fe, Valencia, Valencia,
Spain; 4Division of Hematology, Department of Molecular Biotechnology and Health Sciences,
University of Torino, University of Torino, A.O.U. Città della Salute e della Scienza
di Torino, Turin, Italy; 5Bioanalytics; 6Clinical Sciences, Nordic Nanovector ASA,
Oslo, Norway
Background: There remains a significant unmet medical need for the treatment of patients
with relapsed/refractory DLBCL. Betalutin® (lilotomab labelled with the beta-emitter
177Lu), targets the CD37 antigen which permits delivery of a therapeutic dose of radiation
directly to the DNA of tumour cells. Betalutin® is under development for the treatment
of relapsed/refractory Non-Hodgkin’s Lymphoma (NHL).
Aims: This was an open label Phase I, multi-centre, single ascending dose study of
four different treatment regimens of Betalutin® administered after rituximab pre-treatment
and lilotomab pre-dosing. The purpose of this study was to define the maximum tolerated
dose (MTD) of Betalutin®.
Methods: Eligible participants received 375 mg/m2 rituximab on Day -14 and escalating
doses of both lilotomab (60 and 100 mg/m2) and Betalutin® (10, 15 and 20 MBq/kg) on
Day 0. Participants were followed-up for dose limiting toxicities (DLTs) for up to
12 weeks. Participants were followed-up until disease progression, the start of further
anticancer treatment or until withdrawal from the study for any reason. Tumour response
was determined using Cheson et al, 2014. Clinical benefit was assessed using the ECOG
performance and quality of life (QoL) assessments using the Functional Assessment
of Cancer Therapy-Lymphoma (FACT-Lym) scale.
Results: A total of 16 participants (8 males and 8 females) with median age 75 years
(range: 45 to 94 years) received lilotomab and Betalutin® after pre-treatment with
rituximab. Overall ECOG scores at screening was 0 in 5 (31.3%), 1 in 7 (43.8%) and
2 in 4 (25.0%) participants. DLBCL stage at screening was Stage III or IV in 13 (81.3%).
Median time since last relapse was 1.3 months, and 11 (68.8%) participants were refractory
to the last anti-lymphoma therapy. Cell surface expression of CD37 on tumour cells
was demonstrated for all patients before treatment.
A single case of DLT (grade 4 neutropenia) was reported in an 83-year-old female at
the dose of 100 mg/m2 lilotomab and 20 MBq/kg Betalutin® on day 43; there was no fever
or infections associated with neutropenia; the event was treated with filgastrim and
resolved after 2 weeks on day 57. A total of 14 (87.5%) participants had at least
one treatment emergent adverse event (TEAE). Most events occurred between Days 1-92
and the most commonly reported events (in 4 [25.0%] participants each) were diarrhoea,
decrease in lymphocyte count, and decrease in neutrophil count. Grade ≥3 TEAEs were
reported in 10 (62.5%) participants, of which only four (25.0%) participants had treatment
related events (decrease in lymphocyte count - 2 participants, decrease in neutrophil
count - 1 participant and decrease in white blood cell count -1 participant). No SAEs
assessed were deemed to be related to lilotomab and Betalutin®, by the investigators.
Nine (56.3%) deaths were reported, all were determined to be related to disease progression
and none was considered related to the study drugs.
Two participants (one each from the 2 highest dose cohorts) had complete responses
that lasted 2.8 and 3.0 months, respectively.
Summary/Conclusion: Betalutin® is well tolerated in relapsed/ refractory DLBCL and
a MTD was not reached in this study. The initial efficacy and safety findings from
this study warrant further investigation of Betalutin® as a single agent or in combination
with a standard of care treatment in relapsed/refractory DLBCL.
References: Cheson et al, J Clin Oncol 2014:32: 3059-67
P1166: REPEAT BIOPSY IN RELAPSED OR REFRACTORY DIFFUSE LARGE B CELL LYMPHOMA: A NATIONWIDE
SURVEY AND RETROSPECTIVE STUDY
T. Berger1,2,*, K. R. Geiger1,2, M. Yeshurun1,2, A. Gafter-Gvili1,2,3, T. Shochat4,
R. Gurion1,2, P. Raanani1,2, O. Pasvolsky1,2
1Hematology division, Davidoff cancer center, Beilinson hospital, Rabin medical center,
Petah Tikva; 2Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv; 3Department
of Medicine A; 4Bio-Statistical Unit, Rabin Medical Center, Beilinson Campus, Petah-Tikva,
Israel
Background: Almost half of patients with diffuse large B-cell lymphoma (DLBCL) will
have relapsed/refractory (R/R) disease after frontline immunochemotherapy. Although
guidelines recommend histological confirmation of R/R disease, repeat biopsies are
not always performed.
Aims: To better appreciate the extent and reasoning for not performing repeat biopsies
in R/R DLBCL.
Methods: We conducted a two-part study: (1) using a nationwide electronic questionnaire
consisting of 8 clinical vignettes, we evaluated the views of practicing hematologists
in Israel regarding the need for repeat biopsies in suspected R/R DLBCL; (2) a single
center retrospective study was utilized to describe real-life clinical practice patterns
of performing procedures aimed at achieving tissue diagnosis for relapsed/refractory
aggressive NHL (aNHL) patients: we included all consecutive adult (≥18 years of age)
patients diagnosed with aNHL treated at a tertiary center in Israel, between 1/1/2013
and 1/1/2020, we identified patients who were diagnosed with R/R disease and analyzed
data pertaining to utilization of histological confirmation via repeat biopsy. Each
timepoint of management decision regarding biopsy performance was considered as a
separate “episode of R/R lymphoma”. We extracted data regarding reasons for not performing
the biopsies from the computerized system, according to the treating physician.
Results: In the survey part, all 64 participating physicians opted not to perform
a repeat biopsy in at least one clinical case scenario. Physicians’ age, gender, tenure
or clinical experience with DLBCL patients did not correlate with the decision to
perform a biopsy, whereas more physicians chose to perform biopsies in relapsed compared
to refractory disease cases.
In the retrospective part, 116 episodes of R/R aNHL among 61 patients were identified.
In 72% of these episodes a repeat biopsy was not performed, mostly due to low likelihood
of an alternative diagnosis or problematic location for biopsy. Patients with relapsed
disease were more likely to undergo biopsy, as compared to those with refractory disease
(47% vs 19%, p=0.002). Focusing on the group of patients with DLBCL (excluding primary
CNS lymphoma, primary mediastinal B cell lymphoma and Burkitt lymphoma), there were
43 patients with refractory (n=26) or relapsed (n=17) DLBCL. In 61.4% out of the 83
episodes of R/R disease in these 43 patients, a biopsy was not performed: 74.5% of
refractory DLBCL episodes versus 35.7% of relapsed DLBCL episodes (p=0.001). Stratifying
biopsy utilization for each patient according to the timing of event, i.e first, second
or third event of R/R disease, revealed that less biopsies were performed at more
advanced relapse episodes. The most common reason for not performing a repeat biopsy
was having stable radiological findings and other diagnoses deemed unlikely, as depicted
in Figure 1.
Image:
Summary/Conclusion: Our study suggests that contrary to guideline recommendations,
in clinical practice many patients do not undergo repeat biopsy in R/R DLBCL, especially
in refractory cases. Since this is a common dilemma faced in clinical practice, future
studies and recommendations should address the necessity of repeat biopsy, according
to patient and disease related characteristics.
P1168: CHARACTERISTICS PREDICTIVE OF PROGRESSION DURING FRONTLINE TREATMENT IN AGGRESSIVE
B-CELL LYMPHOMAS
A. Bock1,*, R. Mwangi2, M. Maurer2, J. Cerhan3, W. L. Ng1, R. King4, N. N. Bennani1,
S. Ansell1, T. Habermann1, T. Witzig1, Y. Wang1, G. Nowakowski1
1Division of Hematology; 2Division of Biostatistics; 3Division of Epidemiology, Department
of Quantitative Health Sciences; 4Department of Pathology, Mayo Clinic, Rochester,
United States of America
Background: Primary refractory diffuse large B-cell lymphoma (DLBCL) represents 10-15%
of DLBCL cases. The subgroup of refractory patients who have no response or progressive
disease (PD) by the end of frontline treatment (EOT) is a high-risk group with particularly
poor outcomes. It is challenging to identify such patients at diagnosis.
Aims: To identify clinical, pathological, and cytogenetic characteristics of patients
with progression during frontline treatment.
Methods: Adult patients with newly diagnosed DLBCL, including patients with high-grade
B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements (DHL/THL), between 2002
and 2019 and seen at Mayo Clinic Rochester were identified from the prospective Molecular
Epidemiology Resource (MER) cohort study of the University of Iowa/Mayo Clinic Lymphoma
SPORE. We analyzed patients who had no response or PD during treatment or by EOT (early
PD group) and compared to non-early PD (non-ePD) patients (including relapsed patients).
We compared demographics, clinical characteristics, cell of origin (COO), and MYC
rearrangements using Chi-square analysis.
Results: Out of 1,505 patients with DLBCL, there were 66 patients (4.3%) with early
PD (N=20 PD/stable disease at interim PET-CT, N=46 PD by EOT). At baseline, 61% were
male and the median age was 60. Early PD patients had more advanced stage III-IV (72.7%
vs 53.3%), elevated serum lactate dehydrogenase (LDH) (64.0% vs 47.3%), >1 extranodal
site involved (36.4% vs 23.3%), and International Prognostic Index (IPI) ≥3 (52.0%
vs 33.6%)(all p <0.05 compared to non-ePD). COO distribution was similar between early
PD and non-ePD patients. Among patients with complete pathological and cytogenetic
data available (early PD, n=47; non-ePD, n=813), DHL or THL was observed in 23.4%
of early PD patients compared to 9.2% in non-ePD patients (p=0.001). Of the DHL patients
in the early PD group (n=8), MYC/BCL2 rearrangement was present in 87.5%, MYC/BCL6
in 12.5%. Dual Myc/Bcl2 expressor (DEL) was present in 42.2% of early PD patients
(n=45) compared to 25.5% in non-ePD patients (n=736)(p=0.013). First-line treatment
regimens in the early PD group included R-CHOP (74.2%), R-EPOCH (7.6%), and other
immunochemotherapy (IC)(18.2%), which was similar to non-ePD patients.
Summary/Conclusion: High-risk characteristics including DHL/THL and DEL are significantly
associated with progressive disease during frontline treatment, suggesting that MYC-signaling
may mediate chemoresistance. Known high-risk clinical features including advanced
stage, elevated LDH, >1 extranodal site, and IPI ≥3 are also associated with early
PD. Early identification of patients at high risk for early PD through clinical, pathological,
and cytogenetic features is of utmost importance given the availability of clinical
trials incorporating novel therapies in both frontline and consolidative approaches.
P1169: ELEVATED CRP AT INFUSION OF AXICABTAGENE CILOLEUCEL PREDICTS HIGH-GRADE CAR-T
TOXICITY AND OUTPERFORMS MORE COMPLEX SCORES
S. Boyle1,*, A. Kuhnl1, M. Correia de Farias1, P. E. Patten1,2, D. Yallop1, R. Benjamin1,
V. Potter1, R. Sanderson1
1Dept of Haematology, King’s College Hospital; 2King’s College London, London, United
Kingdom
Background: Predicting CRS and ICANS prior to CAR-T infusion is desirable as it may
help to plan management. Two groups1,2 have recently shown that a modification of
the Endothelial Activation and Stress Index (EASIX) score, developed to predict toxicity
in bone marrow transplant recipients3, can predict CRS and ICANS severity in CAR-T
patients, however these have not been externally validated or are difficult to apply
in practice.
Aims: We aimed to determine whether the EASIX or related scores could predict CAR-T
toxicity in our patients and if not, then to identify other potential predictors from
the available clinical information.
Methods: We reviewed the medical records of adult patients who received axicabtagene
ciloleucel (axi-cel) as standard of care at our centre over a 2yr period, collecting
data on timing and severity of CRS and ICANS as well as baseline demographic and biochemical
factors. We calculated an EASIX score for all patients and an ‘EASIX-F’ score based
on the values provided by Greenbaum et al (EASIX-F1) and our own interquartile values
(EASIX-F2). We then performed a linear regression of each of: the EASIX score; EASIX-F1;
EASIX-F2; and other variables of interest with maximum grade of CRS, with a plan to
perform a multivariate analysis with any significant variables (p<0.05).
Results: 67 patients were eligible for analysis. Baseline demographics were similar
to those described on the licensing trial4. On univariate analysis (UVA), neither
EASIX score (p=0.10), nor either iteration of the EASIX-F score (p= 0.08 and 0.15
respectively), was associated with severity of CRS for our patients. Pre-infusion
C-Reactive Protein (D0 CRP) however appeared strongly associated with CRS severity,
both on UVA (p=0.001) and by multiple linear regression, where it was the only variable
identified that showed any association with CRS severity (p=0.003). Given the strength
of the D0 CRP and CRS association, we evaluated the association of D0 CRP with ICANS
severity which was significant (p=0.012) and D0 CRP with a combined outcome of ‘high
grade CAR-T toxicity’ (HGT), defined as development of either grade 3+ CRS or grade
3+ ICANS, where it was again strongly associated (p<0.001). We then dichotomised D0
CRP to determine whether we could identify a practically useful cut-off to predict
HGT. We looked at D0 CRP values of ≥50, ≥100 and ≥150 individually and all cut-offs
performed well with increasing likelihood of HGT with higher CRP values. With D0 CRP
≥50, probability of developing HGT was 42% vs 13% (OR 5.09, p=0.007); with CRP ≥100
it was 55% vs 14% (OR 7.2, p=0.006); and with CRP ≥150 it was 100% vs 13% (OR undefined,
p<0.001).
Finally, given these unexpected findings we performed a prospective review of the
19 axi-cel patients treated from our initial data-cut off to 1 Feb 2022 and found
that for these patients, a D0 CRP ≥100 or ≥150 predicted HGT well. HGT incidence was
75% vs 7% (OR 42, Fisher’s p=0.016) for both cut-offs because 3 of the 4 patients
with HGT had a CRP of >150 (Table 1). The single patient with HGT and D0 CRP <100
interestingly was a non-responder. For the full 86-patient cohort, D0 CRP ≥150 was
the best cut-off, predicting a 90% incidence of HGT vs 12% if CRP <150 (OR 42, p<0.001).
Image:
Summary/Conclusion: On retrospective analysis of 67 axi-cel patients, D0 CRP was more
strongly associated with CRS severity than more complex prognostic scores. D0 CRP
level, especially if ≥150, showed strong association with high grade CAR-T toxicity
and remained predictive in a 19-patient validation cohort. This has potential implications
for management of axi-cel patients.
P1170: INCIDENCE, TREATMENT AND OUTCOME OF PATIENTS WITH RICHTER’S SYNDROME: A POPULATION-BASED
COHORT STUDY IN THE NETHERLANDS
F. Huisman1, D. Al-sarayfi1, M. Bellido1, R. Mous2, J. S. Vermaat3, A. P. Kater4,
M. Nijland5, M. Brink6,*
1Department of Hematology, University Medical Center Groningen, University of Groningen,
Groningen; 2Department of Hematology, UMC Utrecht Cancer Center, Utrecht; 3Department
of Hematology, Leiden University Medical Center, Leiden; 4Department of Hematology,
Academic Medical Centre, Amsterdam; 5Department of Hematology, University Medical
Center Groningen, Groningen; 6Department of Research and Development, Netherlands
Comprehensive Cancer Organisation, Utrecht, Netherlands
Background: Richter’s syndrome is a rare aggressive transformation of chronic lymphocytic
leukaemia (CLL) into a diffuse large B-cell lymphoma (RS-DLBCL) or Hodgkin lymphoma.
Patients with RS-DLBCL have an average overall survival (OS) of 6-12 months. Survival
of RS-DLBCL patients previously treated for their CLL is inferior as compared to patients
with a previously untreated CLL. In recent years, management of CLL has changed with
the introduction of novel agents.
Aims: This study evaluates the clinical characteristics, treatment and survival of
RS-DLBCL patients in a contemporary era of CLL management in the Netherlands.
Methods: DLBCL patients ≥18 years diagnosed in 2014-2020 were identified in the Netherlands
Cancer Registry (NCR), with survival follow-up through February 1st, 2021. A prior
CLL, diagnosed in 2014-2019, was identified through cross-linkage with the NCR. For
these patients, detailed information on CLL and RS-DLBCL treatment was available.
Patients with CLL and DLBCL diagnosis within a time interval of 3 months were defined
as concurrent RS-DLBCL (concurrent RS). The primary endpoint was overall survival
(OS). OS was defined as the time between diagnosis and death from any cause and presented
for three subgroups of RS-DLBCL patients, 1) concurrent RS, 2) RS-DLBCL following
treated CLL (treated RS), and 3) RS-DLBCL following untreated CLL (untreated RS).
Results: A total of 110 patients with RS-DLBCL (median age 72 years, range 43-89 years;
61% males) were selected, including 22 (20%) concurrent RS, 38 (35%) treated RS, and
50 (45%) untreated RS. The majority of patients were diagnosed with stage III/IV RS-DLBCL,
i.e., 55% of the concurrent RS were diagnosed with advanced disease as compared to
70% and 79% of the untreated and treated RS, respectively. Median time from CLL to
RS diagnosis was for untreated RS 23.2 months (range, 3.4-74.4 months) and for treated
RS 17.4 months (range, 4.7-67.5 months).
Of the 38 treated RS patients, 30 patients had received rituximab in combination with
either chlorambucil (n=11), purine analogues (n=8) or chemotherapeutic regimens such
as cyclophosphamide-vincristine-prednisolonie (n=11). Seven patients received treatment
including ibrutinib or venetoclax, and 3 patients received treatment not including
one or more of the abovementioned regimens.
Regarding primary treatment of all RS-DLBCL patients, 19 (86%) out of 22 concurrent
RS patients received R-CHO(E)P, 2 patients received another R-based chemotherapy,
and 1 patient did not receive therapy. Of the 88 (un)treated RS patients, 59 (67%)
received R-CHO(E)P of whom 6 patients were consolidated with autologous stem cell
transplantation (SCT) and 2 patients with allogenic SCT. Of the 29 remaining patients,
17 (19%) were treated with a R-based chemotherapy regimen including R-PECC and R-bendamustine,
and 12 (14%) patients did not receive therapy.
The overall response rate (≥partial remission) of concurrent RS patients was superior
as compared to untreated and treated RS patients (86% vs. 70% vs. 55%, respectively;
p<0.01). With a median follow-up of 9.5 months (range 0.3-78.3), 3-years OS was 64%
for patients with concurrent RS, 53% for untreated RS patients and 5% for treated
RS patients (Figure; p<0.01).
Image:
Summary/Conclusion: In this contemporary population-based study, concurrent RS patients
have superior OS, while survival among treated RS patients is very poor. Our data
underline the importance of development of new strategies, specifically aiming at
treated RS patients.
P1171: REFLECT: PROSPECTIVE NON-INTERVENTIONAL STUDY ON THE EFFECTIVENESS AND SAFETY
OF SANDOZ RITUXIMAB (SDZ-RTX) WITH CHOP FOR PATIENTS WITH PREVIOUSLY UNTREATED CD20-POSITIVE
DIFFUSE LARGE B-CELL LYMPHOMA
M. Welslau1,*, B. Kubuschok2, J. Topaly3, B. Otremba4, T. Wolff5, G. Bryn6
1Department of Oncology, Klinikum Aschaffenburg, Aschaffenburg; 2Department of Hematology/Oncology,
Augsburg University Medical Center, Augsburg; 3Klinik für Hämatologie und Onkologie,
CaritasKlinikum Saarbrücken, Saarbrücken; 4Onkologische Praxis Oldenburg, Oldenburg;
5OncoResearch Lerchenfeld, Hamburg; 6Sandoz, Holzkirchen, Germany
Background: Sandoz biosimilar rituximab (SDZ-RTX) was approved in the EU for all the
indications of reference rituximab. REFLECT is a real-world, multicenter, open-label,
single-arm, non-interventional trial, and is the first prospective, post-approval
study of SDZ-RTX in combination with cyclophosphamide, doxorubicin, vincristine, and
prednisone (R-CHOP) as a curative therapy for patients with CD20-positive diffuse
large B-cell lymphoma (DLBCL).
Aims: The aim of the REFLECT study was to evaluate the 2-year effectiveness and safety
of SDZ-RTX as first-line treatment for DLBCL.
Methods: Treatment-naïve, CD20-positive adult patients (aged ≥18 years) with DLBCL
eligible for therapy with R-CHOP were treated with SDZ-RTX-CHOP every 2 or 3 weeks
for 6–8 cycles according to the product label, as per routine clinical practice. Data
was collected at enrollment, at every patient contact during the 12-month treatment
period, and at one time point ≥30 days after the last dose of R-CHOP. Complete response
was defined as the best result, as assessed by the treating physician using the Response
Evaluation Criteria in Lymphoma 2017 (RECIL criteria). Kaplan–Meier estimates of progression
free survival (PFS) were assessed at 24 months. No imputation for missing data was
used; endpoints were summarized descriptively. All patients provided written informed
consent prior to study entry. The cut-off for data collection was March 31, 2021.
Results: A total of 169 patients (52.1% female, mean [standard deviation] age 67.3
[13.4] years) with DLBCL were included in the full analysis set. At baseline, 19.5%
and 24.3% of patients had Ann Arbor disease stage III or IV, respectively, and most
patients (80.5%) had Eastern Cooperative Oncology Group performance score of 0 or
1. In terms of International Prognostic Index, 92 (54.5%) patients had a score of
≥2 at baseline. A total of 100 (59.2%) patients completed the 24-month observation
period. In total, 110 (65.1%; 95% confidence interval [CI] 57.4–72.3) patients achieved
complete response as the best response and 50 (29.6%; 95% CI 22.8–37.1) patients achieved
partial response. The overall response rate was 94.7% (95% CI 90.1–97.5). One-year
PFS was 84.9% (95% CI 78.2–89.6), while 2-year PFS was 78.5% (95% CI 70.9–84.4) (Figure
1); median PFS was not reached within the observational period. A total of 143 (84.6%)
patients experienced at least one adverse event (AE), 53 (31.4%) of which were suspected
to be related to the study drug. The most common AEs were anemia (24.3%), fatigue
(20.7%), polyneuropathy (17.2%), and nausea (12.4%). Serious AEs were reported in
63 (37.3%) patients, with those suspected to be related to the study drug reported
in 11 (6.5%) patients. AEs requiring dose interruption and/or dose change were reported
in 24 (14.2%) patients. There were 8 (4.7%) deaths recorded during the study, of which
3 (1.8%) occurred during the on-treatment period.
Image:
Summary/Conclusion: This real-world, post-approval study reconfirms that first-line
treatment of CD20-positive DLBCL with R-CHOP using SDZ-RTX is effective over a 24-month
observation period, with no unexpected safety signals. These data may help to broaden
patient access to rituximab-based chemotherapy and support the sustainability of cancer
care.
P1172: DUVELISIB IN PATIENTS WITH RELAPSED/REFRACTORY PERIPHERAL T-CELL LYMPHOMA FROM
THE PHASE 2 PRIMO TRIAL: UPDATED EXPANSION PHASE ANALYSIS
P. L. Zinzani, MD, PhD, J. Zain, MD2, M. Mead, BS, MD, C. Casulo, MD4, E. D. Jacobsen,
MD5, G. Gritti, MD, PhD6, L. Pinter-Brown, MD7, K. Isutzu, MD8, D. Cohan, MD9, M.
Daugherty, PharmD10, J. E. Brammer, MD11, N. Mehta-Shah, MD12, B. Pro, MD13, S. M.
Horwitz, MD14
1Institute of Hematology “Seràgnoli”, University of Bologna, Bologna, Italy; 2Department
of Hematology and Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer
Center, Duarte; 3Department of Medicine, Division of Hematology/Oncology, UCLA Medical
Center, Santa Monica; 4University of Rochester Medical Center, James P. Wilmot Cancer
Center, Rochester; 5Dana-Farber Cancer Institute, Boston, United States of America;
6Hematology and Bone Marrow Transplant Unit, Ospedale Papa Giovanni XXIII, Bergamo,
Italy; 7University of California-Irvine, Irvine, United States of America; 8National
Cancer Center Hospital, Tokyo, Japan; 9Secura Bio, Inc.; 10SecuraBio, Inc, Las Vegas;
11Division of Hematology, Department of Internal Medicine, The Ohio State University,
Columbus; 12Washington University School of Medicine in St. Louis, Saint Louis; 13Columbia
University Herbert Irving Comprehensive Cancer Center; 14Memorial Sloan Kettering
Cancer Center, New York, United States of America
Background: Peripheral T-cell lymphoma (PTCL) is a family of aggressive lymphomas,
with a short median overall survival when relapsed or refractory (R/R). Current single-agent
therapies for R/R PTCL have modest overall response rates (ORR) of <30%. Use of duvelisib
(DUV), an oral dual inhibitor of phosphatidylinositol 3-kinase (PI3K)-δ and PI3K-γ
isoforms, in PTCL is as an investigational agent only. Both Phase 1 data and prior
interim analyses of this Phase 2, open-label, multi-center, parallel cohort PRIMO
Trial (NCT03372057; supported by Secura Bio) demonstrated promising efficacy (ORR
~ 50%) of DUV in patients (pts) with R/R PTCL. Based on interim PRIMO data, DUV was
added to the National Comprehensive Cancer Network® T-cell Lymphoma Guidelines® (Version
1.2022, 12/22/21) as a Category 2A other recommended regimen for pts with R/R PTCL
of all subtypes. Here we present the latest PRIMO Expansion Phase (EP) data.
Aims: The primary objective of the PRIMO EP is to determine the efficacy of DUV given
at a recommended dose in pts with R/R PTCL; secondary objectives include additional
efficacy measures, safety, and pharmacokinetics.
Methods: EP eligibility criteria includes adults with pathologically confirmed PTCL,
defined per WHO criteria, after ≥ 2 cycles of 1 prior standard regimen, and a CD4
lymphocyte count of ≥ 50/mm3. Based on the dose optimization results, the dose in
the EP is DUV at 75 mg BID for 2 cycles, to maximize disease control, followed by
25 mg BID, to mitigate late toxicities, until progressive disease (PD) or unacceptable
toxicity. Pneumocystis jirovecii prophylaxis is required; herpes simplex and varicella
zoster virus prophylaxis are strongly recommended. The primary endpoint is ORR by
IRC assessment using the Lugano 2014 criteria; efficacy is assessed in all pts that
received at least 1 dose of DUV.
Results: This updated analysis of the EP included 101 pts (data cutoff Oct 1, 2021);
median 10.4 months follow-up from first dose. Pts had a median age of 67.0 years (range,
21-92) with a median of 3 prior lines of therapy (range 1-9). Fourteen pts remain
on treatment with 87 pts withdrawn from treatment for: PD (44), clinical deterioration
due to PD (4), adverse events (AE; 20), death (5), other (13), and withdrawal of consent
(1). The ORR by IRC was 49% with a CR rate of 34%; responses were seen across multiple
subtypes (Table 1 Median PFS was 3.6 months, though not yet fully mature. Five pts
discontinued therapy to undergo stem cell transplantation. There were 3 treatment-related
AEs associated with death (1 each): pneumonitis, Epstein-Barr associated lymphoproliferative
disorder, and sepsis. AEs of special interest ≥ Grade 3 (all causality, number of
events) were transaminase elevation increased (23), neutropenia (19), infections (10),
cutaneous reactions (9), diarrhea (7), pneumonia (2), and pneumonitis (1). There was
1 event of colitis (Grade 1). ALT and/or AST elevations were the most common AEs leading
to treatment discontinuations (15).
Image:
Summary/Conclusion: This updated analysis of 101 patients in the PRIMO EP demonstrated
an ORR of 49% and a CR rate of 34%, which compares favorably to currently available
single-agent options. Duvelisib was generally manageable with per-protocol dose modifications
in this population, showing an AE profile consistent with what has been observed previously
and no unexpected or novel toxicities. These data confirm duvelisib to be a promising
agent for a disease set with high unmet needs, poor prognoses, and limited effective
treatment options.
P1173: CLINICAL APPLICABILITY OF TARGETED NEXT-GENERATION SEQUENCING FOR PRECISION
MEDICINE IN DIFFUSE LARGE B CELL LYMPHOMA
D. Huang1,*, J. Li1, T. Yang1, Y. Nan1, L. Zhang1, Y. Xiang1, Q. Li1, Y. Liu1
1Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing,
China
Background: The cure rate of diffuse large B-cell lymphoma is only 60-70%, and new
treatment methods need to be found.
Aims: Comprehensive genomic analysis based on next-generation sequencing (NGS) allows
a better molecular characterization of diffuse large B-cell lymphoma, offering a roadmap
toward precision medicine.
Methods: We analyzed somatic alterations of 37 DLBCL tissue or blood samples by a
hybridization capture-based NGS panel targeting 188 lymphoma-related genes.
Results: Totally, 279 single-nucleotide variants and small insertions or deletions
affecting 103 genes were identified, with at least 1 mutation detected in 100% (37/37)
of cases. The most frequently mutated genes were those involved in epigenetic modifiers
(DNMT3A, KMT2B and KMT2D), activated signaling (TYK2, MYD88, NOTCH2 and BCR) and transcription
factors/transcriptional regulator (RUNX1, SPEN, IRF4, KLF2 and ASXL1). Significant
co-occurrences were shown between RUNX1 mutations with NOTCH2, IRF4, ARID1A and BCR
(P<0.05, respectively), BCR and IRF4 (P<0.01), etc. Additionally, we analyzed the
association of genomic aberrations with clinicopathological features in DLBCL. Patients
aged >60 years harbored less LRP1B mutations than those aged ≤60 years (5.3% vs. 33.3%,
P< 0.05). The mutation frequency of TP53 was significantly higher in LDH-high (>244 U/L)
group than that in LDH-low (≤244 U/L) group (40% vs. 6.25%, P<0.05). Compared with
germinal center B-cell (GCB), the mutation frequency of TYK was significantly lower
in non-GCB (5% vs. 36.4%, P<0.05). New potential therapeutic targets (CREBBP, EP300,
EZH2, SMARCB1, CD79A/B, BTK, CARD11, MYD88, THFAIP3, PIK3CA, PTEN and JAK) were identified
in 55% (11/20) of newly diagnosed patients and in 41.2% (7/17) of relapsed/refractory
patients. Ibrutinib, as a single agent, has demonstrated limited activity in DLBCL
with mutant MYD88 and wildtype CD79. In this study, we identified 3 patients with
mutant MYD88 and wildtype CD79A/B (8.1%), among whom 2 harbored MYD88 L265P variants
and 1 had MYD88 S219C mutation.
Summary/Conclusion: The utility of mutational profiling may facilitate the identification
of potential drug targets, which in turns may represent new therapeutic possibilities.
P1174: THE COMBINATION OF QUALITATIVE AND QUANTITATIVE METABOLIC PARAMETERS PREDICTS
PROGRESSION-FREE SURVIVAL IN PATIENTS WITH LARGE B-CELL LYMPHOMAS (LBCL) TREATED WITH
ANTI-CD19 CAR-T CELLS.
A. Dodero1,*, A. Guidetti1, A. Lorenzoni2, S. Pizzamiglio3, G. Argiroffi2, P. Verderio3,
A. Chiappella1, F. Bagnoli4, C. Carniti1, M. Pennisi1, A. Alessi2, P. Corradini1
1Hematology; 2Nuclear Medicine; 3Unit of Bioinformatics and Biostatistics, Fondazione
IRCCS Istituto Nazionale dei Tumori; 4Hematology, Policlinico di Milano, Ospedale
Maggiore Ca’ Granda, Milano, Italy
Background: Autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy is
an effective therapy for 40% of patients (pts) with relapsed/refractory (R/R) LBCL.
Qualitative and quantitative metabolic parameters from the 18-FDG PET-CT scan are
tools able to predict outcomes in lymphomas, but their value in the CAR-T setting
is still under investigation.
Aims: Prognostic role of PET-CT performed before lymphodepletion and 1 month after
CAR-T cell infusion, analyzed by qualitative and quantitative parameters
Methods: We conducted an observational a prospective study on pts affected by R/R
aggressive lymphomas receiving axicabtagene ciloleucel (axi-cel) or tisagenlecleucel
(tisa-cel). PET-CT was evaluated before lymphodepletion, at 1 month (M1) and at 3
months (M3) post-infusion using visual assessment with a 5-point Deauville score (DS).
Quantitative PET parameters were evaluated with PET-VCAR software (GE Healthcare).
The contour of pathological lesions was delineated using semiautomatic contouring
systems including a gradient-based algorithm. SUVmax, SUVmean, Metabolic tumor volume
(MTV), and Total lesions glycolysis (TLG) were automatically recorded. Total MTV (TMTV)
and total TLG (TTLG) were calculated as the sum of all individual lesions. The percentage
change in FDG uptake of each parameter between baseline and M1 was defined as Δ=(baseline-M1)/baseline
x 100%. Logistic and Cox regression models were applied to evaluate the association
between PET parameters at baseline, at M1 and in terms of Δ with respect to clinical
response and progression free survival (PFS), respectively.
Results: 47 pts were analyzed: median age was 55 years (range, 22-70); histologies
included diffuse-large B-cell lymphomas (n=33) and primary mediastinal B-cell lymphomas
(n=14); pts received treatment with axi-cel (n=31) or tisa-cel (n=16). Before infusion,
18 (38%) had high LDH, 14 (30%) had extranodal disease and 16 (34%) had bulky disease.
Majority of pts [n=41 (84%)] received bridging therapy. At a median follow-up of 12
months (range, 3-29 months), 38 patients are alive and 9 died of disease progression
with an estimated 1-year PFS and OS of 46% (95%CI; 30%-60%) and 83% (95%CI; 66%-92%),
respectively. Baseline values of SUVmax, but not SUVmean, TTLG, TMTV correlated with
poor clinical response [HR 1,49; (95%CI; 1,01-2,2), p=0,04]. By univariate logistic
model, a higher ΔSUVmean between baseline and M1 was associated with better response
at M3 [odds ratio 3,49, (95%CI; 1,25-9,77), p=0.02]. Univariable Cox analyses showed
that a higher ΔSUVmean and ΔTMTV between baseline and M1 were associated with a better
PFS [HR 0,63, (95%CI; 0,41-0,96), p=0,03; HR 0,67 (95%CI; 0,48-0,94), p=0,02], respectively.
In terms of visual assessment, pts with DS 4-5 at M1 had a significantly higher risk
of failure as compared to those with DS 1-3 [HR 2,92, (95%CI; 1,12-7,64), p=0,02].
A combination of M1 DS score and ΔSUV mean showed a better prognosis for patients
with DS1-3 (PFS 60%), an intermediate prognosis for those with DS4/5 and high ΔSUVmean
(PFS 49%) and a poor prognosis for those with DS4/5 and low ΔSUVmean (PFS 22%), [p=0,009].
In a multivariable analyses, evaluating clinical prognostic factors and PET parameters,
a higher risk of failure was observed in pts with high LDH [HR 3,2 (95%CI; 1,30-8,14),
p=0,01] whereas a higher ΔSUVmean was associated to a better PFS [HR 0,61 (95%CI,
0,41-0,92), p=0,02].
Summary/Conclusion: Assessment of DS combined with variation of SUVmean at M1 provides
informations about early risk of CAR-T failure. A validation group are needed to extend
and confirm our observations.
P1175: REAL LIFE OUTCOMES OF RELAPSED/REFRACTORY DIFFUSE LARGE B CELL LYMPHOMA: A
SINGLE CENTRE EXPERIENCE
I. Dogliotti1,*, M. Clerico1, F. Vassallo2, S. Ragaini1, V. Peri1, B. Botto2, C. Consoli1,
L. Orsucci2, R. Freilone2, S. Ferrero1, A. Lonardo1, G. De Luca1, D. Ferrero1, F.
Cavallo1
1Division of Hematology 1U, Department of Biotechnology and Health Sciences, University
of Torino; 2Division of Hematology 2, University Hospital AOU Città della Salute e
della Scienza, Torino, Italy
Background: Nearly 40% diffuse large B cell lymphoma (DLBCL) patients relapse after
first line treatment (Crump, Blood 2017) and around 15% are chemorefractory (Feugier,
JCO 2005). Despite modern treatments availability, outcome of these patients remains
suboptimal (Crump, Blood 2017).
Aims: With the observational retrospective study STRIDER, we aimed at determining
real-life outcome of relapsed/refractory (R/R) DLBCL patients.
Methods: Patients were selected based on local diagnosis of DLBCL between Jan 2010
and Dec 2019 within Hematology Units at the University Hospital Città della Salute
e della Scienza, Torino, Italy. R/R disease was documented by biopsy, imaging, or
clinical evaluation; refractoriness was defined as reappearance or progression of
DLBCL within 12 months from initial diagnosis. Demographics, laboratory, imaging,
pathological and clinical data were collected retrospectively from hospital health
records.
Results: Overall, 413 DLBCL patients aged >18 years old were included. 151 (36%) patients
had an event of relapse (n=51 patients, 12%) or refractory disease (n=100, 24%). Median
age was 67 (range 21-89) years for non R/R patients (NRR) vs 72 (19-89) for R/R group
(p <0.001). Advanced stage (stage 3-4: 68% vs 87%, p <0.001), baseline B symptoms
(28 vs 43%, p <0.001), higher LDH level (p <0.001) and >1 extranodal site involvement
(39 vs 53%, p =0.007) were linked to R/R disease, and consequently, IPI score highly
affected R/R disease distribution. Kidney or adrenal involvement was less frequent
in the NRR group (6% vs 11%, p =0.042). Central nervous system (CNS) involvement was
present at diagnosis in only 8/413 (2%) patients, while median CNS-IPI score was 2
vs 3 for the 2 groups (p <0.001). Expression of immunohistochemical markers was not
significantly different between groups. Treatment intent was curative (R-CHOP/COMP,
R-Magrath, R-DA-EPOCH, MATRIX) for 391/413 (95%) patients; R/R group included reduced
dose or palliative intent treatments (11 vs 2%, p <0.001). For CNS prophylaxis, intrathecal
methotrexate was used in 78/413 (19%) patients, while intravenous in 22 (5%) patients.
After first line completion, 99/413 (24%) patients received consolidative radiotherapy
on previous bulky areas, while 4 (1%) received upfront autologous stem cell transplantation
(ASCT). Interim response was not different, but end of treatment response was inferior
for R/R group (p <0.001). After a median follow up of 50 months, median overall survival
(OS) from initial diagnosis was NR for NRR patients, 38 months (IQR 25-64) for relapsed,
11 (5-20) for refractory patients (Figure 1). Median progression free survival (PFS)
was 21 (IQR 16-30) months for relapsed vs 6 (4-9) for refractory patients. Treatments
performed in II line were platinum-based (rituximab + OxDHA/DHAP/ICE/GemOx) for 50
patients (22 relapsed, 28 refractory), lenalidomide for 17 patients (5 vs 12); 14
patients were enrolled in clinical trials. Palliative intent treatments (oral chemotherapy
+/- rituximab) were administered to 17 patients. II line consolidation with RT was
performed in 11, ASCT consolidation in 6 patients. After II line, best response was
CR in 29%, PR in 17% (ORR 45%); 45 patients received a third line, 10 more than three.
Image:
Summary/Conclusion: Our retrospective series confirmed good survival for NRR patients,
but 36% patients were R/R after first line, with refractory patients showing a dismal
outcome despite intensive treatments. Survival analyses after II line and multivariate
analysis are ongoing and will be presented at the meeting.
P1176: RITUXIMAB IN PRIMARY CNS LYMPHOMA – LONG TERM FOLLOW-UP OF THE PHASE III HOVON
105/ALLG NHL 24 STUDY
J. Doorduijn1,*, S. Issa2, B. van der Holt3, M. Minnema4, T. Seute5, M. Durian6, G.
Cull7, M. van der Poel8, W. Stevens9, J. Zijlstra10, M. Nijland11, K. Mason12, A.
Beeker13, D. Brandsma14, M. van den Bent15, M. Gonzalez16, D. de Jong17, J. Bromberg15
1Hematology, Erasmus MC Cancer Institute Rotterdam, Rotterdam, Netherlands; 2Hematology,
Middlemore Hospital, Auckland, New Zealand; 3HOVON Data Center, Erasmus MC, Rotterdam;
4Hematology; 5neurology, University Medical Center Utrecht, Utrecht; 6Hematology,
ETZ Hospital, Tilburg, Netherlands; 7Hematology, Sir Charles Gairdner Hospital and
PathWest Laboratory Medicine, Nedlands, Australia; 8Hematology, University Medical
Center, Maastricht; 9Hematology, Radboud University Medical Center, Nijmegen; 10Hematology,
Amsterdam UMC, Amsterdam; 11Hematology, University Medical Center Groningen, Groningen,
Netherlands; 12Hematology, Royal Melbourne Hospital, Melbourne, Australia; 13Hematology,
Spaarne Gasthuis, Haarlem; 14neurology, Netherlands Cancer Institute, Amsterdam; 15neurology,
Erasmus MC Cancer Institute Rotterdam, Rotterdam, Netherlands; 16pathology, Royal
Melbourne Hospital, Melbourne, Australia; 17pathology, Amsterdam UMC, Amsterdam, Netherlands
Background: The efficacy of rituximab in Primary CNS Lymphoma (PCNSL) is still under
debate. We performed an international randomized phase III study to investigate the
efficacy of rituximab when added to methotrexate, BCNU, teniposide and prednisolone
(MBVP) in PCNSL. The primary endpoint, event-free survival (EFS) at one year, was
similar in both treatment groups and was previously reported (Bromberg et al, Lancet
Oncology 2019; 20: 216-228).
Aims: Here we present long-term follow up results with a median follow-up of 82 months.
Methods: Between August 2010 and May 2016 200 newly-diagnosed, non-immunocompromised
patients with PCNSL aged 18-70 years and WHO performance status 0-3 were randomized
between treatment with MBVP chemotherapy with (arm B) or without (arm A) rituximab.
The rituximab was given weekly in the first MBVP cycle, fortnightly in the second
(in total 6 rituximab administrations). Responsive patients received consolidation
with high-dose cytarabine, and patients aged ≤ 60 were subsequently treated with low-dose
WBRT if in CR/CRu; in case of PR with an additional boost on the tumor. Patients >
60 were not irradiated. All patients gave written informed consent.
Results: The modified intention-to-treat (m-ITT) population consisted of 199 eligible
patients, 55% were men. The median age was 61 yrs (range 26-70), the median WHO performance
status 1 (range 0-3). The primary endpoint EFS at one year was 49% (95% CI 39-58)(MBVP)
vs 55% (95% CI 44-64) (R-MBVP). The EFS at 5 years was 25% (17-34) vs 36% (27-46)
respectively, hazard ratio (HR) 0.85, 95% CI 0.61-1.18, p=0.33 (adjusted for age and
WHO performance status). The progression-free survival (PFS) at one and 5 years were
58% (47-67) and 29% (21-39) (MBVP) and 65% (54-73) and 43% (33-53) (R-MBVP)) (HR 0.73,
95% CI 0.52-1.02, p=0.07).
80 patients were still alive. Overall survival (OS) at 5 years for MBVP and R-MBVP
was 49% (39-59) and 53% (43-63) respectively. The median OS was 57 months (95% CI
38-75) in the MBVP arm and 85 months (95% CI 43–104) in the R-MBVP arm (HR 0.87, 95%
CI = 0.61-1.26, p=0.47). A total of 111 patients had progression or relapse, 63 after
MBVP and 48 after R-MBVP. 79% of these patients received further treatment. The median
OS after progression/relapse was 9.7 months (5.9-19.9) in the MBVP arm, and 6.1 months
(2.4-13.1) in the R-MBVP arm (HR 1.25, 95% CI 0.83-1.87, p=0.29).
119 patients died, 64 in the MBVP arm and 55 in the R-MBVP arm. Causes of death were
PCNSL in 69% of the patients (both arms), complication of treatment (6% vs 5%), secondary
malignancy (5% vs 2%) and other or unknown causes (20% vs 24%). Age was the strongest
prognostic factor for EFS, PFS and OS in multivariate analysis.
An unplanned subgroup analysis by age (≦60 vs > 60 yrs) showed a significant different
EFS at one year for younger patients with R-MBVP, not found in patients > 60 yrs (Bromberg
et al, 2019). With longer follow up the 5-year EFS in the younger group was 28% (16-41)(MBVP)
vs 55% (40-68) (R-MBVP), HR 0.48, 95% CI 0.28-0.81, p=0.006. 5-year PFS was 31% (18-44)
versus 57% (42-70), HR 0.47, 95% CI 0.27-0.81, p=0.007. The OS at 5 years was 54%
(39-68) vs 70% (55-81) respectively (HR 0.65, 95% CI 0.36-1.19, p=0.16).
Image:
Summary/Conclusion: In the modified-ITT population we found no statistically significant
benefit of the addition of rituximab to MBVP on EFS, PFS and OS in patients with PCNSL,
even after a long follow-up of median 82 months. Therefore, the results of this study
do not support the use of rituximab with MBVP in the treatment of primary CNS lymphoma.
P1177: PROGNOSTIC VALUE OF TP53 VARIANTS IN CHILDREN WITH MATURE B-CELL LYMPHOMA TREATED
ACCORDING TO REDUCED-INTENSITY B-NHL-2010M PROTOCOL
V. Egor1,*, A. Yuliya1, K. Brenning1, A. Itov1, D. Abramov2, A. Sharlai2, M. Senchenko2,
D. Konovalov2, G. Novichkova1, N. Miakova1
1Oncohematology; 2Pathology, Dmitry Rogachev National Medical Research Center Of Pediatric
Hematology, Oncology and Immunology, Moscow, Russia
Background: Reduced-intensity rituximab-based therapy leads to long-term remission
in more than 80-85% of pediatric patients with B-cell non-Hodgkin lymphoma (B-NHL),
but in case of relapse or refractoriness the prognosis in poor with less then 20%
survival rate. Therefore, the identification of adverse prognostic factors is critically
important. Recent data show that the status of the TP53 gene may be of prognostic
value in children with B-NHL. We evaluated the clinical significance of TP53 abnormalities
in children with B-NHL enrolled in the Russian-Belorussian B-NHL-2010mab multicenter
clinical trial.
Aims: To investigate the prognostic value of TP53 gene status in pediatric B-NHL receiving
reduced-intensity therapy
Methods: Due to reference status of pathology department in Dmitry Rogachev National
Medical Research Center for Pediatric Hematology, Oncology, and Immunology, samples
from 79 pediatric B-NHL patients (77 with Burkitt lymphoma and 2 with diffuse large
B-cell lymphoma) treated according to B-NHL-2010mab protocol in 2012-2021 were included
in this study. The median age was 9 years (range 2-17 years), m:f ratio was 6:1. 19
patients (24%) had limited stage and 60 patients (76%) had advanced-stage of disease.
DNA was extracted from FFPE tissue, TP53 variants were detected using Sanger sequencing
of exons 5 to 8 according to the International Agency for Research on Cancer (IARC)
protocol. B-NHL samples were obtained following informed consent from participants
or their parents/guardians.
Results: Median follow up was 16 months (range 5-117 months). The 5-year event free
and overall survival for all patients was 86,3% (95%CI=78, 3-94%) and 86,5% (95%CI=78,3-94,8%)
respectively. DNA suitable for sequencing was obtained from 66 samples. TP53 pathogenic
variants were present in 26% of cases and were associated with a significantly inferior
event free and overall survival compared to those without a TP53 abnormality: 45,3%
(95%CI = 21-69,6%) vs 97,9% (95%CI=93,9-100%) (p < 0.001) and 47,1% (95%CI=21,8-72,4%)
vs 97,9% (95%CI= 93,9-100%) respectively (p = 0.001). 13 patients with unknown TP53
status are alive without relapsed of disease.
Summary/Conclusion: Our findings show the independed prognostic value of TP53 pathogenic
variants in pediatric B-NHL patients treated with rituximab-based reduced-intensity
therapy. Therefore, given the poor survival rate of patients with TP53 mutations,
this marker can be used to define high risk groups within pediatric B-NHL, those who
need new therapeutic approaches to achieve better results. It is necessary to study
this prognostic factor in large groups of patients.
P1178: BRENTUXIMAB VEDOTIN IN COMBINATION WITH LENALIDOMIDE AND RITUXIMAB IN PATIENTS
WITH RELAPSED/REFRACTORY DLBCL: SAFETY AND EFFICACY RESULTS FROM THE SAFETY RUN-IN
PERIOD OF THE PHASE 3 ECHELON-3 STUDY
N. L. Bartlett1,*, C. A. Yasenchak2, K. K. Ashraf3, W. N. Harwin4, J. Orcutt5, P.
Kuriakose6, P. L. Zinzani7,8, A. Mamidipalli9, K. Fenton9, C. Glenn9, G. Nowakowski10
1Washington University School of Medicine, St. Louis, MO; 2Willamette Valley Cancer
Institute and Research Center/US Oncology Research, Eugene, OR; 3Hem-Onc Assocs, Birmingham,
AL; 4Florida Cancer Specialists and Research Institute, Fort Myers, FL; 5Charleston
Hematology Oncology Associates, Charleston, SC; 6Henry Ford Health System, Detroit,
MI, United States of America; 7Institute of Hematology “L. e A. Seràgnoli”, University
of Bologna; 8Department of Specialized Medicine, Diagnostic and Experimental, University
of Bologna, Bologna, Italy; 9Seagen Inc., Bothell, WA; 10Division of Hematology, Mayo
Clinic, Rochester, MN, United States of America
Background: Patients (pts) with relapsed/refractory (R/R) diffuse large B-cell lymphoma
(DLBCL) who relapse after or are ineligible for hematopoietic stem cell transplant
(HSCT) or chimeric antigen receptor T cell (CAR-T) therapy generally have poor outcomes.
Preclinical data provide the rationale for combining brentuximab vedotin (BV), lenalidomide
(len), and rituximab for the treatment of R/R DLBCL. BV+len showed promising clinical
activity in a Phase 1 trial with a 57% overall response rate (ORR), 10.2-month median
progression-free survival, and 14.3-month overall survival (Ward 2021).
Aims: We report the results of the open-label safety run-in that was conducted prior
to the randomized portion of the study.
Methods: ECHELON-3 (NCT04404283) is a randomized, double-blind, placebo-controlled,
active-comparator, multicenter Phase 3 study. Prior to randomization, an open-label
safety run-in was conducted; pts received BV (1.2 mg/kg) and rituximab (375 mg/m2)
both q3w, and len 20 mg qd. Pts must have received ≥2 prior lines of therapy and be
previously treated with or were ineligible for HSCT or CAR-T, ECOG score ≤2, and PET-avid,
bidimensional measurable disease (>1.5 cm by CT). Pts with negligible CD30 expression
(<1%) were eligible. Response was assessed by the investigator according to the Lugano
Classification Revised Staging System (Cheson 2014).
Results: 10 pts with R/R DLBCL were enrolled. Median age was 70.5 years, 7 pts were
male, and all had an ECOG ≤1. Median prior lines of therapy was 3 (range, 2 to 6);
no pts received prior HSCT and 6 pts received prior CAR-T.
At a median follow-up of 6.9 months (range, 2.3 to 14.1), the most common treatment
emergent adverse events (TEAE) were fatigue (n=5), anemia (n=4) and constipation (n=4).
Grade ≥3 events were experienced by 8 pts, most commonly anemia and pneumonia (n=3
each), and neutropenia and thrombocytopenia (n=2 each). Serious adverse events were
observed in 7 pts. Seven pts each had BV and rituximab dose modifications, and 6 pts
had a len dose modification. The most common reasons for any dose modification were
anemia (n=3), neutropenia, peripheral neuropathy, or pneumonia (n=2 each). 4 pts discontinued
treatment due to an AE, 2 of which were treatment related (n=1 each of Grade 2 fatigue;
Grade 3 anemia). There was 1 death due to a TEAE that was not treatment related.
The ORR (best response) was 70%, including 5 pts with a complete metabolic response
and 2 pts with a partial metabolic response; 3 pts had progressive disease. Responses
were seen in both CD30 (+) and (-) pts, as well as in 4 pts who received prior CAR-T.
Summary/Conclusion: This novel triplet regimen appears active in R/R DLBCL with an
acceptable safety profile. The randomized portion of the study is currently enrolling.
P1179: EFFECTIVENESS OF CENTRAL NERVOUS SYSTEM PROPHYLAXIS STRATEGIES IN DIFFUSE LARGE
B-CELL LYMPHOMA
F. Z. Eraslan1, T. Toptas2,*, T. Tuglular2, I. Atagunduz2
1Internal Medicine; 2Hematology, Marmara University School of Medicine Hospital, İSTANBUL,
Turkey
Background: Central nervous system recurrence is seen in 5% of patients diagnosed
with DLBCL. CNS relapse is associated with survival limited to months. Therefore,
determining the patients who are at high risk of CNS relapse and selecting the most
efficient prophylactic approach is one of the mainstays of treatment. IV-MTX and IT-MTX
are widely used in CNS prophylaxis. Nevertheless, the efficiency of IV-MTX and IT-MTX
in preventing CNS relapse was not proven in large study groups or in prospective studies.
Aims: Our study aims to verify the efficiency of current CNS prophylaxis in preventing
CNS relapse in DLBCL patients. Additionally, association of CNS relapse with survival
and the correlation of CNS-IPI score, age, albumin, origin of cell, and DHL/DEL status
with cumulative CNS relapse incidence.
Methods:
The data from 190 patients were analyzed retrospectively. Among the 89 patients who
had been classified as high-risk group for CNS relapse, seven were administered IT-MTX,
17 received IV-MTX, and remaining 65 patients received no prophylaxis.
Results: Median age was 57(18-88) and male to female ratio was almost equal (male:
48.8%). At the time of diagnosis, 66.8% of the patients were defined to have advanced
stage disease. All of the cohort included 89 patients (46.8%) who were at high risk
for CNS relapse. Cumulative CNS relapse risk at one-year was estimated as 12.2%, 9.5%,
4.5% for patients received IV-MTX, IT-MTX and no prophylactic treatment, respectively.
Two-year cumulative CNS relapse risk was 24.5%, 12.2%, 6.0% for patients who received
IV-MTX, IT-MTX and no prophylaxis, respectively (p=0.008). The frequency of DEL or
DHL were higher in patients who received IV-MTX (41.1%). CNS relapse rate was 25%
and 50% for patients who did not receive prophylaxis and who did, respectively. In
patients with DEL, CNS relapse rate (33.3%) did not differ for those who received
prophylaxis and those who did not. In our study, we found overall CNS relapse incidence
6.8% in DLBCL. Median time to CNS relapse was 12 months in all cohort. Median overall
survival after occurrence of CNS relapse was two (95% CI: 0-4.9) months. We found
that the patients who had high CNS-IPI score were at six times greater risk of CNS
relapse than those with low and intermediate CNS-IPI score (9.0% and 1.5% after 12
months, 11.6% and 2% after 24 months) (p=0.03). Despite the univariate analyses revealed
a statistically significant association, multivariate analysis has shown that albumin
level is not an independent predictor for CNS relapse. Additionally, we have shown
that CNS-IPI score, DHL/DEL status are strong predictors of CNS relapse in DLBCL patients.
Image:
Summary/Conclusion: Independent predictors of CNS relapse in DLBCL are DHL/DEL status
and CNS-IPI score. No predictive relationship was proven to exist between CNS relapse
and gender, albumin levels, cell of origin classification by Hans-algorithm. Patients
with CNS relapse had poor prognosis (median survival two months). IV-MTX prophylaxis
has been shown to have no preventive effect on CNS relapse in high risk DLBCL population.
Due to this data, significant changes in prophylactic approach against CNS relapse
in high risk DLBCL patients is required.
P1180: ARE EXTRANODAL LIMITED-STAGE DIFFUSE LARGE B-CELL LYMPHOMA MORE AGRESSIVE THAN
NODAL FORMS?
C. Fernandes1,*, S. Alves1, F. Moita1, I. Dlouhy1, S. Esteves2, M. Gomes da Silva1
1Hematology; 2Clinical Research Unit, Instituto Português de Oncologia de Lisboa Francisco
Gentil, Lisboa, Portugal
Background: Limited-stage diffuse large B-cell lymphoma (DLBCL) is associated with
favourable outcomes with overall survival of 70-80% at 10 years. Stage-modified IPI
(sm-IPI) predicts the risk of relapse and death based on clinical factors that do
not include extranodal (EN) involvement, although in some studies it has been associated
with worse outcomes.
Aims: To analyse the characteristics and outcomes of limited-stage DLBCL in a single
center series and evaluate the prognostic impact of EN involvement.
Methods: We conducted a retrospective analysis of consecutive patients with newly
diagnosed stage I-II DLBCL treated with curative intent at a single centre from 2015
to 2019. Primary central nervous system (CNS) lymphoma, primary testicular lymphoma,
primary mediastinal B-cell lymphoma and transformed lymphoma were excluded. Response
to therapy was assessed according to Lugano criteria. Overall (OS) and progression
free survival (PFS) were calculated by Kaplan-Meier method and comparisons done by
the log rank test. Cumulative incidence of relapse was calculated using competing
risk models and Gray test for group comparisons. Cox regression was used to identify
prognostic factors for PFS.
Results: Of 152 patients identified, 53 (35%) had EN disease and 99 (65%) were nodal
(N). The most common EN sites were stomach (30%), sinus/nasal cavity (13%), skin and
soft tissue (9%) and breast (9%). Seventy-two (47%) had low risk sm-IPI (45 N; 27
EN). Clinical characteristics were similar between both groups, except for stage (stage
I in 50.9% of EN vs 32.3% of N cases) and bulky disease (EN 15.1% vs N 37.4%).
All patients received R-CHOP (median 6 cycles; range 1-8). Eight patients, all with
EN involvement (3 sinus/nasal cavity, 2 breast, 2 bone and 1 orbit), received CNS
prophylaxis. Seventeen out of 152 patients received additional radiotherapy (RT),
71% of whom had nodal involvement; 16/17 were irradiated due to bulky disease and/or
treatment with abbreviated immunochemotherapy. Globally, 89% patients had a complete
response (CR) to treatment: 85% in the N and 96% in the EN group (p=0.212). Nine patients
relapsed at a median of 18 (7-52) months after obtaining CR (6 from the N and 3 from
the EN group). The 4-year cumulative incidence of relapse was similar: 11.2% (95%CI
9.0-13.9%) in EN and 6.7% (95%CI 5.9-7.5%) in N group (p=0.778). No gastric lymphomas
relapsed. The most common sites of relapse were lymph nodes (3), soft tissue (3) and
CNS (2). Nineteen (14 N; 5 EN) patients died at a median of 12 (0-47) months after
starting treatment, mostly (52.6%) due to lymphoma progression. With a median follow-up
of 43 months, 4-year OS and PFS were 87% and 82%, respectively. No significant survival
differences were observed in patients with N or EN presentation with a 4-year PFS
of 81.8% and 81.3% (p=0.428), respectively. On multivariable analysis adjusting for
RT, sm-IPI and bulky disease, EN involvement was not associated with a decreased PFS
compared with N involvement (HR 0.77; 95%CI 0.31-1.90; p=0.570).
Image:
Summary/Conclusion: Our study corroborates the very favourable outcomes of localized
DLBCL. We could not find differences in PFS according to N and EN presentations when
controlling for other prognostic factors. These results do not support different treatment
strategies for EN disease; however, due to limited sample size the role of RT and
the prognostic impact of specific locations cannot be fully ascertained.
P1181: REAL-WORLD CHARACTERISTICS AND CLINICAL OUTCOMES IN RELAPSE/REFRACTORY DIFFUSE
LARGE B-CELL LYMPHOMA POST CAR-T FAILURE
C. Flores Avile1,*, L. Liao2, L. Wilson1, A. Lau2, L. Chen2
1Genesis Research, Hoboken; 2ADC Therapeutics, New Providence, United States of America
Background: Progressive disease following chimeric antigen receptor T-cell (CAR-T)
therapy for diffuse large B-cell lymphomas (DLBCL) is a common scenario. There are
limited treatment options after CAR-T failure with a poor prognosis for patients at
this stage in their disease. The effectiveness of existing treatment options following
CAR-T failure is still being investigated in the real-world setting.
Aims: To further understand the clinical outcomes of CAR-T failure in relapse/refractory
(RR) DLBCL patients in the real-world setting.
Methods: This retrospective analysis identified adult patients diagnosed with RR-DLBCL
[01/01/2014 – 03/31/2021] who received CAR-T therapy and experienced a subsequent
disease progression or death. COTA’s Real World Evidence (RWE) database is comprised
of longitudinal, HIPAA-compliant data abstracted from electronic health records (EHR)
from over 200 sites of care in US (60% academic, 40% community). The first post CAR-T
therapy was categorized as checkpoint inhibitor +/- other therapies (CPI), investigational
therapies, tafasitamab +/- lenalidomide, polatuzumab-containing regimen (pola-containing),
lenalidomide +/- anti-CD20, BTK inhibitors (BTKi), chemotherapy/chemoimmunotherapy
(CT/CIT), allogenic stem-cell transplant (allo-SCT), or anti-CD20 monoclonal antibody.
Overall response rate (ORR), complete response (CR), and overall survival (OS) were
reported for treatment groups with at least 5 patients.
Results: Of the 97 CAR-T patients identified, 57 (59%) patients failed CAR-T therapy
due to receiving subsequent line of therapy, having documented progression event,
or death. Patients who failed CAR-T were 67% male and on average 59 years old. Within
a median follow-up of 12.4mo, 44 (77%) initiated further therapy whereby 7 (16%) initiated
investigational therapies, 9 (20%) CPI, 7 (16%) tafasitamab +/- lenalidomide, 6 (14%)
pola-containing, 5 (11%) lenalidomide +/- anti-CD20, 3 (7%) CT/CIT, 3 (7%) anti-CD20
monoclonal antibody, 2 (5%) BTKi, and 2 (5%) allo-SCT as their first post CAR-T therapy.
Of these patients, 48% received more than two lines of therapies after CAR-T. Response
rates of select first therapies received after CAR-T are detailed in Table 1. Outside
clinical trials, ORR and CRs were highest for lenalidomide +/- anti CD20 (60% & 20%),
followed by CPI (33% & 0%), pola-containing (33% & 0%) and tafasitamab +/- lenalidomide
(14% & 0%). OS of first post CAR-T therapy was highest for lenalidomide+/- anti CD20
(12.5 mo), then CPI (11 mo), pola-containing therapy (6.37 mo), and tafasitamab +/-
lenalidomide (2.7 mo).
Image:
Summary/Conclusion: There is no existing standard of care after patients fail CAR-T
therapy. Although further research is warranted in a larger sample population, poor
clinical outcomes in treatment response and longevity were observed with existing
treatment options. There is still a high unmet need for more effective therapies after
CAR T-cell therapy has failed.
P1182: REAL-WORLD CHARACTERISTICS AND CLINICAL OUTCOMES IN RELAPSE/REFRACTORY DIFFUSE
LARGE B-CELL LYMPHOMA PATIENTS WHO RECEIVED CAR-T THERAPY
L. Liao1, L. Nastoupil2, L. Wilson3, C. Flores Avile3,*, T. Kilavuz1, L. Chen1
1ADC Therapeutics, New Providence; 2Department of Lymphoma–Myeloma, Division of Cancer
Medicine, University of Texas, Houston; 3Genesis Research, Hoboken, United States
of America
Background: A significant proportion of patients diagnosed with diffuse large B-cell
lymphoma (DLBCL) experience refractory or relapse (RR) disease. Approval of chimeric
antigen receptor T-cell (CAR-T) therapies has resulted in a novel therapeutic option
for eligible patients with RR-DLBCL. However, progressive disease post CAR-T remains
a common scenario as patient identification, timing, and effectiveness of CAR-T in
the real-world setting is still evolving.
Aims: To further understand clinical outcomes of standard of care CAR-T in RR-DLBCL
in clinical practice.
Methods: This retrospective analysis identified adult patients diagnosed with RR-DLBCL
[01/01/2014 – 03/31/2021] who received CAR-T therapy. COTA’s Real World Evidence (RWE)
database is comprised of longitudinal, HIPAA-compliant data abstracted from the electronic
health records (EHR) of healthcare provider sites, representing diverse treatment
U.S settings from over 200 sites of care; roughly 60% of patients are seen at academic
sites and 40% are seen at community sites. Patients were categorized as having received
CAR-T therapy in 2L, 3L, 4L, or 5L. Baseline characteristics was reported for CAR-T
patients. Best response rate, treatment failure, and overall survival (OS) were reported
by line of therapy. Disease characteristics were derived from the EHR, including the
presence of high-grade lymphoma (positive rearrangement in C-MYC and BCL-2 or BCL-6
biomarkers) and primary refractory disease (2L started within 6 months not due to
patient preference, drug shortage, insurance reasons, toxicity, or pandemic reasons).
CAR-T treatment failure was defined as the earliest of death, initiation of subsequent
line of therapy, or documented progression event after CAR-T.
Results: A total of 97 CAR-T patients were identified whereby 17 received CAR-T therapy
in a clinical trial setting and were excluded from this real-world evidence study.
Of the 80 patients that remained, 10 (13%) received CAR-T in 2L, 31 (39%) in 3L, 24
(30%) in 4L, and 15 (19%) in 5L+. CAR-T patients had a mean age of 57 years, most
were male (58%), 16% were diagnosed with high-grade lymphoma, and 60% were primary
refractory. Median time from diagnosis to initiation of CAR-T was 15 months. Overall,
38% of patients achieved a complete response with a decrease in response in later
lines (2L: 70%, 3L: 58%, 4L: 29%, 5L+: 33%). Within a median follow up of 13.5 months
(2L: 13.5 mo, 3L: 15.2 mo, 4L: 12.8 mo, 5L+: 9.5 mo), treatment failure occurred in
45% of patients, with an increase in later lines (2L: 20%, 3L: 48%, 4L: 63%, 5L+:
80%). Median OS was 31.2 months (Not reached (NR); 2L: NR, 3L: NR, 4L: 26.3 mo, 5L+:
13.5 mo) with unequal survival probabilities across lines of therapy (Log-rank test:
p=0.004) (Figure 1).
Image:
Summary/Conclusion: CAR T-cell therapies are considered a major advance in DLBCL,
yet over half of those patients eventually fail. Outcomes are inferior in later lines
with a decrease in complete response rates and shorter survival by line of therapy,
thus, highlighting the need to provide CAR T-cell therapies in earlier settings.
P1183: CAR-T MORPHOLOGICAL CHARACTERISTICS AND DYNAMICS IN PERIPHERAL BLOOD, EVALUATED
BY A NOVEL MICROSCOPY PLATFORM
G. Fridberg1,*, B.-Z. Katz1, D. Benisty1, C. Perry1, R. Ram1, I. Avivi1
1Hematology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
Background: Chimeric Antigen Receptor T-cells (CAR-T) therapy becomes a preferable
therapeutic approach in patients with relapsed and refractory (R/R) non-Hodgkin’s
large B cell lymphomas (LBCL) who failed ≥2 therapeutic regimens. However, there are
no commercial assays for routine CAR-T measurements and clinical studies often employ
PCR or FACS, both expensive and relatively complicated. Detection of CAR-T in peripheral
blood smear (PBS) is challenging, mostly due to the lack of data regarding their morphology
prior to transfusion and the low sensitivity of currently available technologies.
As oppose to the currently available digital morphology, Scopio Labs Full Field Morphology
(FFM) is a novel digital microscopy platform that provides high-resolution images
combined with a full field analysis, incorporating artificial intelligence capabilities,
thereby enabling the detection and classification of rare cells in PBS, including
CAR-T cells.
Aims: The dynamics of CAR-T quantity and subtypes in PBS were evaluated, and correlated
with responsiveness to treatment and treatment-related toxicities.
Methods: Morphological library of CAR-T cells was established from cells obtained
directly from the CAR-T transfusion-bag (Fig. 1A). FFM analysis of consecutive PBS
samples obtained from 26 R/R DLBCL patients, treated either with tisagenlecleucel
(tisa-cel) or axicabtagene ciloleucel (axi-cel) at the Tel Aviv Sourasky Medical Center
between October 2019-October 2020 was performed.
Results: We identified in the CAR-T cell transfusion bags 5 distinct CAR-T morphological
sub groups prior to transfusion: regular, activated, apoptotic, multinucleated and
cells in mitosis (Fig. 1A).
First, 79 smears, obtained from 18 patients, were examined with the FFM platform vs
the CellaVision DM1200 system. Overall, 24,247 WBC were detected by the FFM platform
(average 303 cells per PBS), compared with 4,335 detected by CellaVision DM1200 system
(average 54.4 cells per PBS). Detection of CAR-T was superior with the FFM platform
(median difference per PBS - 2, IQR 0-6, p<0.001). Then, PBS from 26 patients, 21
(80.7%) treated with tisa-cel, and 5 (19.2%) treated with axi-cel, were analyzed by
the FFM platform. The absolute number of CAR-T cells in the PBS post infusion was
higher in patients treated with axi-cel (across days 1 to 15). The average numbers
of total, regular, and activated CAR-T cells, measured on day 5 post CAR-T-transfusion,
were all higher in patients that obtained CR vs those who did not (Fig. 1B). Longer
duration of cytokine release storm (CRS) was associated with higher number of CAR-T
cells with activated morphology, and a lower presence of apoptotic cells on day 14.
Higher number of apoptotic cells on day 14 were associated with a deferred CRS onset,
while lower number of activated morphology cells on day 14 was associated with earlier
onset of CRS. There was a positive correlation between the number of cells, measured
on day 7 by FACS, and time to peak of regular morphological CAR-T cells (r=0.67, p=0.034,
n=10).
Image:
Summary/Conclusion: In this study, we show that CAR-T cells has heterogeneous morphology
identified in PBS. Specific morphological features in specific days were associated
with outcome and therapy related toxicities, emphasizing that CAR-T monitoring require
quantitative, qualitative and repeated measurements. Thus, morphological CAR-T surveillance
using FFM might serve as an available, simple and inexpensive method to provide clinically
relevant insights.
P1184: PHASE I TRIAL OF MB-CART2019.1 IN PATIENTES WITH RELAPSED OR REFRATORY B-CELL
NON-HODGKIN LYMPHOMA: 2 YEAR FOLLOW-UP REPORT
P. Borchmann1,*, A. Lohneis1, P. Gödel1, H. Balke-Want1, C. Schmid2, F. Ayuk3, S.
Holtkamp4, L. Hanssens4, G. Zadoyan4, B. Friedrichs4, M. Assenmacher5, I. Bürger5,
D. Schneider6, T. Overstijns4, C. Scheid7, U. Holtick7, S. Miltenyi5, M. Hallek1
1Department of Internal Medicine I, University Hospital of Cologne, Cologne; 2II.
Med. Klinik Hämatologie/Onkologie, Universitätsklinikum Augsburg, Augsburg; 3Department
of Stem Cell Transplantation, Universitätsklinikum Hamburg-Eppendorf, Hamburg; 4Miltenyi
Biomedicine GmbH; 5Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany; 6Lentigen
Technology Inc., Gaithersburg, United States of America; 7Leukapheresis and Stem Cell
Transplant Unit, Department of Internal Medicine I, University Hospital of Cologne,
Cologne, Germany
Background: A tandem chimeric antigen receptor (CAR) targeting CD20 and CD19 (pLTG1497)
has shown superior efficacy as compared to each single-CAR in pre-clinical models.
MB-CART2019.1 consists of autologous CD4 and CD8 enriched T-cells, which are transduced
with a lentiviral vector that encodes the CAR construct for both CD20 and CD19 incorporating
a 4-1BB co-stimulatory domain.
Aims: This first-in-human, Phase I study had the objective to assess feasibility,
safety and toxicity of ex vivo generated MB-CART2019.1 in mainly elderly patients
(pts) with relapsed or refractory (r/r) CD20 and CD19 positive aggressive B-cell Non-Hodgkin
lymphoma (B-NHL) without curative treatment option (NCT03870945).
Methods: The trial included two predefined dose levels (DL1 1.0x106 and DL2 2.5x106
CAR T-cells/kg body weight (BW), respectively). In DL1 pts with multiple prior treatment
lines were included while in DL2 the patient population was limited to pts with r/r
DLBCL and 1 prior line of treatment who were transplant-ineligible. Fludarabine and
cyclophosphamide were used for lymphodepletion. Infusion of fresh MB-CART2019.1 was
scheduled 14 days after leukapheresis. The primary endpoint was to evaluate the maximum
tolerated dose of MB-CART2019.1 as determined by dose limiting toxicities (DLT). Secondary
endpoints included adverse events, overall response rate (ORR), maximum concentration
(Cmax), area under the curve (AUCd0-d28), and persistence of CAR T-cells, measured
by flow-cytometry.
Results: A total of 12 pts, 6 per dose level, have been treated in the trial after
obtaining informed consent. Median age was 72 y (range 20, 78 y), with 8 pts >70 y.
Histologies included aggressive B-NHL (9), transformed follicular lymphoma (2), and
mantle cell lymphoma (1). No grade ≥3 cytokine release syndrome (CRS) or neurotoxicity
were observed. Haematotoxicity was very limited with no anemia or thrombocytopenia
≥grade 3 beyond day 28 and intermittent neutropenia ≥grade 3 in only two pts beyond
week 8 after treatment. No DLT were observed.
The ORR was 75% (3/6 pts in DL1 and 6/6 pts in DL2) with 5/12 pts (3/6 pts in DL1
and 2/6 pts in DL2) achieving PET-CT negative complete remission (CR) (CR-group).
Among those 5 pts, all had ongoing CR as radiologically documented by PET-CT or CT
at 12 months and all completed a 2-year follow-up visit without evidence of relapse
as per investigator assessment. Patients with only partial response or stable disease
as best overall response ultimately progressed.
All pts with CR had a Cmax ≥450 cells/µL with mean Cmax 1,092.5 cells/µL (range 460.1,
3,147.0) while pts with no CR had lower values with mean Cmax of 111.0 cells/µL (3.9,
458.0). Mean AUCd0-d28 was 7,901 d*cells/µL (2,399.2; 19,574.7) in the CR-group as
opposed to mean AUCd0-d28 of 942.0 d*cells/µL (39.3; 3,471.62) in the non-CR group.
Mean time of CAR T-cell persistence in the CR-group was 491 days (274, 736 d) and
all 5 CR pts had detectable CAR T-cells beyond month 6.
Summary/Conclusion: As reported previously, recommended dose of MB-CART2019.1 is 2.5x106
CAR T-cells/kg BW. Importantly, all 5 pts with CR were still in CR 12 months after
treatment and have now completed the 2-year follow-up visit without evidence of relapse.
MB-CART2019.1 is currently challenging conventional immune-chemotherapy in a randomized
Phase II trial for elderly, non-transplant eligible pts with first progression or
relapse of aggressive B-NHL (NCT04844866).
P1185: ANTITUMOR ACTIVITY AND SAFETY OF ZANUBRUTINIB COMBINED WITH R-CHOP REGIMEN
IN THE TREATMENT OF NEWLY DIAGNOSED NON-GCB DLBCL WITH EXTRANODAL INVOLVEMENT: A PROSPECTIVE
PHASE II STUDY.
C. Li1,2,*, H. Geng1, X. Zong1,2, J. Zhou1,2, Y. Zhang1, J. Li1, S. Jia1, D. Wu1,2
1National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of
Hematology, The First Affliated Hospital of Soochow University; 2Institute of Blood
and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow
University, suzhou, China
Background: Approximately one-third of Diffuse large B-cell lymphoma(DLBCL) arises
primarily from extranodal sites,less than 70% showed extranodal involvement in at
least 1 site.Patients with multiple extranodal involvement indicates adverse clinical
outcomes,and non-GCB subtype is another poor prognostic factor.Study shows that the
rates of complete remission(CR),five-year OS,five-year PFS were significantly lower
for the non-GCB subtype(77.5% vs 52.6%,78% vs 54%,76% vs 48%).Patients who failed
in initial treatment had poor prognosis and short overall survival.In the genetic
taxonomy, with non-GCB tumors enriched for MCD and BN2 subtype are suitable for the
treatment of BTK inhibitor.Zanubrutinib has shown very promising future in newly diagnosed
and relapsed/refractory B-cell lymphoma.
Aims: We conducted a single-arm,phase II trial for newly diagnosed non-GCB DLBCL with
extranodal involvement to evaluate the efficacy and safety of the zanubrutinib combined
with R-CHOP(number NCT 04835870).
Methods: 20 patients enrolled in this prospective single-center study with newly diagnosed
non-GCB DLBCL from October 2020 to February 2022.Patients received therapies:R-CHOP(intravenously
rituximab(375mg/m2 on Day0), cyclophosphamide(750mg/m2on Day1),doxorubicin(50mg/m2
on Day1),vincristine(1.4mg/m2 on Day1),and oral prednisone(50mg/day Day1-5) with Zanubrutinib(160mg
bis in die orally) regimen. Particularly,patients with a weak constitution or older
than 70 years old were administered ZR-miniCHOP.
Results: 20 patients received ZR-CHOP regimen treatments, Patients had a median age
of 60 years(31~83years) and 4 patients were over 70 years old,11 patients (55%) had
IPI-PS ≥3 score,16 patients (80%) had stage III/IV disease,13 patients (65%) had ≥2
extranodal organs involved,2 patients (10%) had bulky disease and 9 patients (45%)
were double-expression lymphoma confirmed by immunohistochemistry and FISH (Table
1). The median follow-up time of all patients was 8.7 months(0.4~16.1months) and there
are 12 patients received 6 ~ 8 cycles of treatment who can evaluate the efficacy.
The objective response rate(ORR) of 12 patients was 91.67%(11/12),11 patients(91.67%)
reached complete remission(CR),of which 9 achieved CR at the interim evaluation, and
1 patient(8.3%) reached progression disease(PD).The median OS and PFS of the 12 patients
who finished treatment were 10.7months(5.6~16.1months) and 10.2months(5.6~16.1months).One
patient relapsed(Figure 1).13(65%) patients reduced the dose during treatment, the
most common AEs were hematological toxicity, fatigue and apositia. all-grade and grade
3 or higher hematological toxicity were 100% and 65%,11 patient observed grade 3 or
higher neutropenia and 3 patients had severe pneumonia during treatment. Hemorrhage
were observed in 3 patients,2 patients developed atrial fibrillation after the first
medication.No patient had fatal adverse events(Table2).
Image:
Summary/Conclusion: Our results showed that Zanubrutinib plus R-CHOP(ZR-CHOP) is a
safe and effective scheme for this part of non-GCB DLBCL patients with extranodal
involvement.It indicate that the addition of zanubrutinib to the standard first-line
treatment can enable these patients to achieve remission in the early stage of treatment
and without significantly increasing toxicity.
P1186: A LARGE FRENCH REAL WORLD MULTICENTRIC PROSPECTIVE COHORT OF PATIENTS WITH
LYMPHOMA (REALYSA STUDY): DESCRIPTION OF THE DIFFUSE LARGE B CELL LYMPHOMA PATIENTS
IN REAL WORLD IN FRANCE
H. Ghesquieres1,*, F. Cherblanc2, A. Belot2, V. Camus3, K. Thokagevistk4, K.-K. Bouabdallah5,
C. Esnault4, L.-M. Fornecker6, S. Micon4, F. Bijou7, C. Haioun8, N. Morineau9, L.
Ysebaert10, G. Damaj11, S. Le Gouill12, S. Guidez13, F. Morschhauser14, C. Thiéblemont15,
A. Chauchet16, R. Gressin17, F. Jardin3, C. Fruchart18, G. Labouré19, L. Fouillet20,
P. Lionne-Huyghe21, A. Bonnet22, L. Lebras23, S. Amorim24, C. Leyronnas25, G. Olivier26,
R. Guieze27, T. Lamy28, V. Launay29, B. Drenou30, O. Fitoussi31, L. Detourmignies32,
J. Abraham33, C. Soussain34, F. Lachenal35, P. Fogarty2, P. Cony-Makhoul2, A. Bernier2,
S. Le Guyader-Peyrou36, A. Monnereau36, F. Boissard37, C. Rossi38
1Hopital Lyon Sud, Pierre Benite; 2LYSARC, Pierre-Benite; 3Centre Henri Becquerel,
Rouen; 4Roche, Boulogne-Billancourt; 5CHU Haut Lévêque, Pessac; 6ICANS, Strasbourg;
7Institut Bergonié, Bordeaux; 8Hôpital Henri Mondor, Créteil; 9CHD Vendée, La Roche
sur Yon; 10IUCT Oncopôle, Toulouse; 11Institut d’Hématologie de Basse Normandie, Caen;
12Centre Hospitalier Universitaire de Nantes, Nantes; 13Centre Hospitalier Universitaire
de Poitiers, Poitiers; 14CHRU de Lille, Lille; 15Hôpital Saint-Louis, Paris; 16CHU
Jean Minjoz, Besancon; 17Centre Hospitalier Universitaire Michallon, La Tronche; 18Centre
Hospitalier de Dunkerque, Dunkerque; 19Centre Hospitalier de Libourne, Libourne; 20Institut
de Cancérologie Lucien Neuwirth, Saint-Priest-en-Jarez; 21Centre Hospitalier d’Arras,
Arras; 22Centre Hospitalier Bretagne Atlantique, Vannes; 23Centre Léon Bérard, Lyon;
24Hôpital Saint-Vincent-de-Paul, Lille; 25Groupe Hospitalier Mutualiste de Grenoble,
Grenoble; 26Centre Hospitalier de Niort, Niort; 27CHU de Clermont-Ferrand, Clermont-Ferrand;
28CHU Pontchaillou, Rennes; 29Centre Hospitalier Yves Le Foll, Saint-Brieuc; 30Groupe
Hospitalier Mulhouse et Sud Alsace, Mulhouse; 31Polyclinique Bordeaux Nord Aquitaine,
Bordeaux; 32Centre Hospitalier de Roubaix, Roubaix; 33Centre Hospitalier Universitaire
de Limoges, Limoges; 34Institut Curie, Saint-Cloud; 35Centre Hospitalier Pierre Oudot,
Bourgoin-Jallieu; 36Université de Bordeaux, Bordeaux, France; 37Roche - F.Hoffman-La
Roche Ltd, Basel, Switzerland; 38Centre Hospitalier Universitaire Francois Mitterand,
Dijon, France
Background: In France, there were an estimated 18.000 incident lymphoma cases in 2018.
Currently, most of the knowledge comes from clinical trials (CT), with stringent inclusion
criteria and poor representativeness. Real world data (RWD) is essential to complement
CT data, but major challenges such as feasibility and data quality remain. In this
context, the REal world dAta in LYmphoma and Survival in Adults (REALYSA) multicentric
prospective cohort started in 2018 (NCT03869619), with a recruitment objective of
6000 patients and a 9-year follow-up (FU).
Aims: The aims of this study are (i) to evaluate the capacity of a real-world program
to provide high-quality data that translate into meaningful clinical endpoints, and
(ii) to describe the demographic and clinical characteristics as well as therapeutic
management and treatment effectiveness of 1st line (1L) DLBCL patients.
Methods: Patients diagnosed with 7 histological lymphoma subtypes are included in
REALYSA at diagnosis after signing informed consent. Patients receive standard routine
care. Data collection includes demographic, clinical, quality of life and epidemiological
data at inclusion and every 6 to 12 months. A biobank is also constituted. A strong
data validation system inspired from clinical research standards, including care pathway
visualisation tools, is regularly running. Meaningful real-life endpoints were derived,
such as end of treatment (EoT) evaluation and event-free survival (EFS).
We present here the results of a proof-of-concept analysis for REALYSA patients with
a DLBCL diagnosis and who received a 1L treatment.
Results: Thirty-five hospitals/clinics are currently participating to REALYSA across
France. As of Jan 31st 2022, 1217 patients with DLBCL were included in REALYSA with
a mean number of 60 patients per month; 645 epidemiological questionnaires were collected;
and there were more than 1500 samples in the biobank.
The analysis was conducted on 645 DLBCL patients included in REALYSA before March
31st 2021. Median age was 66.3 years [54-75]. Most patients were male (344; 53.3%),
with advanced-stage disease (Ann-Arbor stage III/IV for 472 patients (73.3%)), extra-nodal
locations (499; 77.4%) and elevated LDH level (402; 64.0%). The international prognostic
index was 2 to 5 for 486 patients (76.2%). Treatments received at 1L were R-CHOP (482;
74.7%), R-miniCHOP (86; 13.3%), high dose anthracycline-based regimen (62; 9.6%) and
non-anthracycline based regimen (15; 2.3%). Among patients with EoT evaluation done
(603), 483 (80.1%) had a complete response, 51 (8.5%) a partial response, 7 (1.2%)
a stable disease and 62 (10.3%) a progressive disease. Of note, EoT evaluation was
performed by PET/scan for 92% of patients. Median FU was 9.9 months [4.5-17.2]. Results
for effectiveness at 12 months were: (i) EFS of 77.9% [95% CI 73.8-81.4]; (ii) PFS
of 79.7% [75.6-83.1]; and (iii) overall survival of 90.0% [86.5-92.5]. With the limitation
of the short FU, and using backward stepwise regression model, we confirmed that EFS
was associated with age, ECOG PS, LDH level and bulk disease.
Summary/Conclusion: This analysis showed that REALYSA, the first nationwide real-life
cohort on lymphoma, is able to provide high-quality data, with results for 1L DLBCL
patients consistent with the literature, including recent phase 3 trials. These data
will be useful for numerous purposes, including a better characterization of lymphoma
population in France (clinic, epidemiology, biology), as well as innovative study
designs (e.g. new outcome endpoints, synthetic control arm).
P1187: END OF TREATMENT 18F FDG-PET/CT RESPONSE IS PROGNOSTIC FOR OVERALL AND PROGRESSION-FREE
SURVIVAL IN PERIPHERAL T-CELL LYMPHOMA: RESULTS OF THE UK NCRI PHASE 2 RANDOMISED
CHEMO-T TRIAL PET/CT SUBSTUDY
M. Gleeson1,2,*, C. Arias2, D. Cunningham2, C. Peckitt2, Y. Du2, N. Hujairi2, Y. M.
To2, H.-C. Chen2, S. Patel2, I. Chau2, P. Johnson3, K. M. Ardeshna4, A. Wotherspoon2,
A. Attygalle2, E. A. Hawkes5,6, M. P. Macheta7, G. P. Collins8, J. Radford9, A. Forbes10,
A. Hart11, S. Montoto12, P. McKay13, K. Benstead14, N. Morley15, N. Kalakonda16, Y.
Hasan17, D. Turner18, S. Chua2
1Guy’s and St. Thomas’ NHS Foundation Trust, London; 2The Royal Marsden NHS Foundation
Trust, London and Surrey; 3Cancer Research UK Centre, University of Southampton, Southampton;
4University College London Hospitals, London, United Kingdom; 5Olivia Newton John
Cancer Research Centre, Austin Health; 6Monash University, Melbourne, Australia; 7Blackpool
Victoria Hospital, Blackpool; 8Oxford Cancer and Haematology Centre Churchill Hospital,
Oxford; 9University of Manchester and the Christie NHS Foundation Trust, Manchester;
10Royal Cornwall Hospital, Truro; 11New Victoria Hospital, Glasgow; 12St Bartholomew’s
Hospital, London; 13Beatson West of Scotland Cancer Centre, Glasgow; 14Gloucestershire
Hospitals NHS Foundation Trust, Cheltenham; 15Nick.morley@sth.nhs.uk, Sheffield; 16The
Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool; 17Sandwell and West Birmingham
Hospitals NHS Trust, Birmingham; 18Torbay Hospital, Torquay, United Kingdom
Background: The role of 18F FDG PET (PET) in peripheral T-cell lymphoma (PTCL) remains
under evaluation. The UK NCRI phase 2 randomised CHEMO-T trial compared cyclophosphamide,
doxorubicin, vincristine and prednisolone (CHOP) with gemcitabine, cisplatin and methylprednisolone
(GEM-P) in previously untreated PTCL patients. The primary endpoint, to demonstrate
a superior complete response (CR) rate for GEM-P by contrast-enhanced CT (CECT) at
end of treatment (EOT) was not met.
Aims: To investigate the role of PET in determining baseline bone marrow (BM) involvement
in PTCL and as a prognostic tool at EOT.
Methods: Patients required a baseline PET (acquired as per protocol) within 28 days
of randomisation and a PET at EOT at least 28 days post chemotherapy. Site accreditation,
data collation, and quality control for PET imaging was performed by the UK PET Core
Lab. BM biopsy was required within 6 weeks of randomisation. PET scans were centrally
reviewed at The Royal Marsden by 2 expert nuclear medicine physicians (YD and NH).
Baseline BM involvement by PTCL on PET was evaluated. PET response was assessed according
to the revised response criteria for malignant lymphoma and by the Deauville score
(DS) 5-point scale. Where discordance existed between reviewers, a third reviewer
(SC) adjudicated. EOT PET response and PET status (DS 1-3=PET-negative, DS 4-5=PET-positive)
were correlated with PFS and OS.
Results: Baseline PET data was available for 82/84 patients, 80/82 (98%) had FDG-avid
disease (n=2 with non-avid disease (n=1 AITL, n=1 EATL) were excluded). Clinical characteristics
were similar to the main trial cohort: median age=63 yrs, male=73% and high-risk IPI=23%.
PTCL subtypes were AITL n=33 (41%), PTCL-NOS n=32 (40%), ALK- ALCL n=14 (18%) and
EATL n=1 (1%). For 20/79 (25%) patients with biopsy-proven BM infiltration 9/19 (47%)
had BM uptake on baseline PET. Of 20/80 patients (25%) with BM uptake on baseline
PET, 9/20 (45%) had biopsy-confirmed infiltration. PET had a sensitivity of 47% and
a specificity 81% for determination of BM involvement. EOT PET was available for 69/80,
n=4 were excluded as data was acquired following a later line (n=3) or complete imaging
was not retrievable (n=1); therefore 65/80 had evaluable EOT PET data. The median
number of days from EOT to PET was 41. At EOT, PET response was CR n=41 (63%), partial
response (PR) n=15 (23%), stable disease n=0 and progressive disease (PD) n=9 (14%);
41 (63%) were PET-negative and 24 (37%) were PET-positive. At a median follow-up of
27 months, PET-negative vs. PET-positive patients at EOT had a 2-yr OS of 72% vs.
52% [HR 0.58; 95% CI 0.24–1.41), p=0.231] and 2-yr PFS of 55% and 29% [HR 0.45 (95%
CI 0.23–0.88), p=0.021] respectively. EOT PET response correlation with 2-yr OS was
CR=72%, PR=67%, PD=29%; and with 2-yr PFS was CR=55%, PR=32%, PD=25%. A CR by PET
(vs. PD) was associated with improved OS [HR 0.245 (95% CI 0.083–0.730), p=0.012]
(Figure 1) and PFS [HR 0.265 (95% CI 0.102–0.687), p=0.006]. Adjusting for stratification
parameters at randomisation (IPI risk group, PTCL subtype) PET response remained prognostic
for both OS and PFS while EOT PET status (positive vs. negative) did not. CECT response
remained prognostic for PFS only.
Image:
Summary/Conclusion: Our findings indicate that PET has insufficient sensitivity for
detection of BM involvement for baseline staging. Furthermore, while both EOT CECT
and PET response were prognostic for PFS and OS, only PET response remained prognostic
for OS following adjustment for stratification factors suggesting it is a superior
prognostic tool in PTCL.
P1188: PERSONALIZED TREATMENT ACCORDING TO BIOLOGICAL PROFILE IMPROVES OUTCOME OF
NEWLY DIAGNOSED PATIENTS AFFECTED BY DIFFUSE LARGE B-CELL LYMPHOMA: A REAL-LIFE EXPERIENCE
A. Guidetti1,2,*, R. Pasquale3, A. Dodero1, A. Chiappella1, L. Devizzi1, L. Farina1,
C. Rusconi1, M. Pennisi1, P. Corradini1,2
1Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori; 2Pathophysiology and
Transplantation, University of Milano; 3Hematology, Ospedale Maggiore Policlinico,
Milano, Italy
Background: Diffuse Large B-Cell Lymphoma (DLBCL) is a heterogeneous disease characterized
by biological and clinical aspects conditioning different prognosis. Chemo-immunotherapy
treatment with R-CHOP proved to be less effective in patients with double hit (DH)/triple
hit (TH) or double expressor (DE) DLBCL who require more intensive regimens
Aims: To investigate survival outcomes of DLBCL patients treated according to biological
profile in the real-life clinical practice at Istituto Nazionale dei Tumori in Milano.
Primary objective was to verify whether personalized treatment could overcome well
known differences in Progression Free Survival (PFS) observed between DE, DH/TH and
non DE/DH/TH patients when they are uniformly treated with R-CHOP
Methods: We retrospectively collected clinical and survival data of all newly diagnosed
DLBCL patients referred at our Hematology Division from January 2013 to August 2021.
All DLBCL patients are evaluated for BCL2, BCL6 and MYC expression in immunohistochemistry
and gene rearrangements by fluorescent in situ hybridization, and they are assigned
to treatment based on the biologic profile: non DE/DH/TH patients receive 6 cycles
R-CHOP-21 + 2R, DE patients receive 6 cycles R-DA-EPOCH and DH/TH receive R-CODOX-M/R-IVAC.
Central Nervous System (CNS) prophylaxis with lumbar puncture and/or intravenous high
dose methotrexate is reserved for DE or DH patients and for those with CNS-IPI 3-5
Results: Since 2013, 243 patients affected by DLBCL at diagnosis were consecutively
treated according to biological risk: 139 non DE/DH/TH patients received R-CHOP, 80
DE patients received R-DA-EPOCH and 24 high risk patients received R-CODOX-M/R-IVAC
(n= 13 DH/TH patients and n=11 DE patients with CNS IPI 3-5). Median age was 62 years
(range, 21-86), 178 patients (73%) presented in stage 3-4 and 97 patients (40%) had
an IPI score of 3-5. The three groups were not significantly different in terms of
clinical characteristics, except for median age, which was higher in the R-CHOP group
(66 years vs. 58 and 57 years in R-DA-EPOCH and R-CODOX-M/R-IVAC groups, respectively).
Despite the toxicity expected with an intensified therapy, 95% of patients completed
the treatment and no toxic deaths were registered. Febrile neutropenia was observed
in 3%, 15% and 29% of patients in RCHOP, RDAEPOCH and RCODOX-M/R-IVAC, respectively.
At a median follow-up of 42 months (range, 2-106 months), the estimated 3 years PFS
and OS for the entire population were 80% and 89%, respectively. Patients with low
IPI, limited stage and germinal center B cell of origin had significant better PFS
as compared to the rest. Three years PFS and OS according to biological entity and
relative treatment group were not significantly different, being 84%, 77% and 66%,
(p=ns) and 93%, 85% and 74%, (p=ns) in non DE/DH/TH, DE and high risk patients, respectively.
Similar PFS between non DE/DH/TH, DE and high risk patients were observed also in
the subgroup of patients with IPI3-5 with PFS of 77%, 65% and 57%, respectively (p=ns).
CNS prophylaxis was administered to 177 patients (73%) and 9 cases of CNS recurrence
(4%) were observed
Summary/Conclusion: In this study, we describe the feasibility of a personalized treatment
according to biological risk in a real-life setting. This approach seems to overcome
the poor outcome of DLBCL patients at high risk of relapse and ameliorate the prognosis
of the overall DLBCL population. Biological characteristics should be carefully considered
at diagnosis in order to give patients a personalized program and improve their outcome
P1189: INITIAL SAFETY RUN-IN RESULTS OF THE PHASE III POLARGO TRIAL: POLATUZUMAB VEDOTIN
PLUS RITUXIMAB, GEMCITABINE, AND OXALIPLATIN IN PATIENTS WITH RELAPSED/REFRACTORY
DIFFUSE LARGE B-CELL LYMPHOMA
A. McMillan1,*, C. Haioun2, J.-M. Sancho3, A. Viardot4, A. Rodriguez Izquierdo5, E.
M. Donato Martin6, A. M. García-Sancho7, J. Sandoval-Sus8, H. Tilly9, E. Vandenberghe10,
J. Hirata11, P. Choudhry11, Y. M. Chang12, L. Musick11, M. Matasar13
1Centre for Clinical Haematology, Nottingham University Hospitals NHS Trust, Nottingham,
United Kingdom; 2Lymphoid Malignancies Unit, Groupe Hospitalier Henri Mondor, Créteil
Cedex, France; 3ICO-HU Germans Trias I Pujol, Barcelona, Spain; 4University Hospital
of Ulm, Ulm, Germany; 5Hospital Universitario 12 de Octubre Servicio de Hematología,
Madrid; 6Hospital Universitario Dr Peset Servicio de Hematología, Valencia; 7Hospital
Clínico Universitario de Salamanca Servicio de Hematología, Salamanca, Spain; 8Moffitt
Cancer Center at Memorial Healthcare Institute, Pembroke Pines, United States of America;
9Centre Henri Becquerel and University of Rouen, Rouen, France; 10St James Hospital
Cancer Clinical Trials Office, Dublin, Ireland; 11Genentech, Inc., South San Francisco,
United States of America; 12Hoffmann-La Roche Ltd, Mississauga, Canada; 13Memorial
Sloan Kettering Cancer Center, New York, United States of America
Background: Transplant-ineligible patients with relapsed/refractory diffuse large
B-cell lymphoma (R/R DLBCL) have a poor prognosis (Gisselbrecht C, et al. Br J Haematol
2018). Several treatment options are available, including platinum-based chemotherapies
such as oxaliplatin plus rituximab and gemcitabine (R-GemOx). Adding polatuzumab vedotin
to R-GemOx (Pola-R-GemOx) may improve outcomes for patients with a continued unmet
medical need. Pola-R-GemOx is an investigational combination. The safety of polatuzumab
vedotin and platinum-based therapy combinations must be considered as both are associated
with neuropathy. POLARGO (NCT04182204; MO40598) is a Phase III, multicenter, open-label,
randomized trial evaluating the safety and efficacy of Pola-R-GemOx vs R-GemOx in
patients with R/R DLBCL.
Aims: To present the results from the safety run-in stage of POLARGO.
Methods: The primary endpoint is the safety and tolerability of polatuzumab vedotin
(1.8 mg/kg) + R-GemOx (R, 375 mg/m2; Gem, 1000 mg/m2; Ox, 100 mg/m2) given every 21
days for up to 8 cycles. Safety was assessed by the incidence, nature, and severity
of adverse events (AEs; NCI CTCAE v5.0), with a focus on peripheral neuropathy (PN).
Dose interruptions and reductions were used to assess tolerability. Granulocyte-colony
stimulating factor was given as primary prophylaxis with each cycle (C) of therapy;
anti-infective prophylaxis for pneumocystis and herpes virus was mandatory. All patients
provided informed consent.
Results: As of October 26, 2021, 15 patients were enrolled, and 11 (73%) received
≥4 cycles of Pola-R-GemOx. Median age was 76 (range 47–87) years, 10 (