21
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Ongoing Clonal Evolution in Chronic Myelomonocytic Leukemia on Hypomethylating Agents: A Computational Perspective

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          To the Editor Chronic myelomonocytic leukemia (CMML) is a clinically heterogeneous myeloid neoplasm that combines myelodysplastic and myeloproliferative features, and carries a poor prognosis due to progression to acute myeloid leukemia or complications of cytopenias. TET2, SRSF2 and ASXL1 are the most commonly mutated genes in CMML, but somatic variants in additional genes have been identified 1–3 . Allogeneic stem cell transplantation is potentially curative, but most patients are ineligible due to advanced age and/or co-morbidities. Hypomethylating agents (HMAs) such as 5-azacitidine (5-Aza) induce responses in ~40% of CMML patients, but their impact on survival remains debatable 4 . While TET2 mutations have been reported to predict HMA response in myelodysplastic syndromes, data from patients treated with decitabine suggest that epigenetic profiles rather than somatic mutations govern response to HMAs in CMML 5, 6 . Previous work described unchanged mutant allele burden in CMML in patients responding to HMAs 1 , but detailed analyses of clonal evolution in relation to HMA response have not been reported. We applied SubcloneSeeker computational analysis algorithm to whole exome sequencing (WES) and single nucleotide polymorphism (SNP) array data to uncover clonal architecture and evolution in CMML patients treated with 5-Aza on a prospective trial 7 . Compared to targeted sequencing, this approach allows reconstruction of clonal architecture using all somatic mutation calls, including driver and passenger mutations, in an unbiased fashion. We studied a total of 46 samples obtained from twelve patients treated with single-agent 5-Aza for up to two years. All patients provided informed consent for the study. Ten patients were treated on a prospective clinical study (NCT01350947) and two in analogy to the study protocol. Median age at presentation was 70 years. Ten patients had CMML-1 and two had CMML-2. Three patients (25%) achieved complete remission (CR) and four (33%) partial remission (PR), one had stable disease (SD), and one had no response (International Working Group 2006 response criteria). Three patients (25%) progressed after a period of SD (Supplementary Table 1). Mononuclear cells (MNCs) or monocytes were isolated from bone marrow or blood samples prior to treatment, at 3- and 6-month intervals on therapy, and at the end of study or disease progression. Cultured mesenchymal stromal cells (n=4), skin fibroblasts (n=6) or fluorescence-activated cell-sorted CD3+ lymphocytes (n=2) were used as sources of constitutional DNA in CMML patients. WES was performed on paired tumor and control samples, with median of 3 longitudinal tumor samples per patient (n=46). Copy number variation (CNV) and loss of heterozygosity (LOH) were analyzed by whole-genome SNP arrays (Infinium Omni2.5-8 v1.3, Illumina). Variant allele frequencies (VAFs) were corrected for proportion of lymphocytes in MNC samples. A median of 39 (range: 10 – 95) somatic mutations per patient exome was detected, with average read depth (DP) of >200X per sample. The most common variants were non-synonymous missense single-nucleotide variants (SNVs) (90%), followed by frameshift insertions/deletions and stop-gain mutations (Supplementary Figures 1a-c). The variants were predominantly transitions (65%) with transition-transversion ratio of 1.86, similar to other myeloid and lymphoid malignancies 8, 9 . Mutations in TET2 and SRSF2 were each found in 62% of the patients. Additional mutations detected in genes associated with CMML included ASXL1 (38%), RUNX1 (38%), CBL (31%), KRAS (23%), DNMT3A (15%) and NRAS (15%) (Figure 1a). TET2, SRSF2 and ASXL1 mutations persisted across pre- and post-treatment samples (Figure 1a). Two patients showed copy neutral (CN)-LOH. CNV and mutations with corresponding VAFs in longitudinal samples are summarized in Supplementary Table 2. Our data revealed higher median somatic mutations per exome than a previous report 1 , probably related to higher average read depth. However, somatic mutation burden was remarkably stable despite response to 5-Aza. Of 477 total somatic mutations identified prior to 5-Aza, 98% were still detectable in the last follow-up samples and only 13 new variants were acquired on therapy. To map clonal architecture, we used the SubcloneSeeker computational algorithm to construct a set of clonal trees by clustering all somatic variants with similar VAFs and calculating their cellular prevalence values 10 . Compatible trees from multiple longitudinal samples at different time points were merged to establish a unified model of clonal evolution in each patient (Supplementary Methods). Clonal evolution patterns were studied in patients without LOH, based on changes in relative proportions of parental clones, pre- and post-treatment (Figure 1a, Supplementary Table 4). In patients P01 to P03, baseline clonal architectures remained relatively stable with proportional changes between parental clones and progeny subclones on 5-Aza (Figure 1b, Supplementary Figure 2a). In patients P04 to P06, we saw increasing shifts from parental clones to progeny subclones on therapy (Supplementary Figures 2b-d). In patients P07 to P10, clonal architecture was characterized by expansion of maximally mutated progeny subclones on 5-Aza (Figure 1c, Supplementary Figures 2e-f). We next delineated clonal evolution in patients with CN-LOH. Patient P11 harbored subclones bearing SRSF2 P95H, RUNX1 L144Q and two CBL variants (CBL C384Y, CBL C416Y) at presentation (Figure 2a). While in SD on 5-Aza treatment, the patient acquired chromosome 11q CN-LOH, with uniparental disomy of CBL C384Y. These subclones expanded after acquisition of additional RUNX1 mutations and became dominant at disease progression. A similar pattern of clonal evolution was observed in patient P12 with CR. At 3 months on 5-Aza, CN-LOH of chromosome 12 led to elimination of KRASA146V-containing subclones, with reversion to native KRAS; and focal CN-LOH in chromosome 17 led to reduction of SRSF2 P95H-containing subclones. At 6 months, clonal architecture was largely simplified to subclones containing TET2 V239fs and SH2B3 V402M (Figure 2b). Our data illustrate early clonal dominance and clonal heterogeneity with co-existence of parental and progeny populations at baseline. TET2, SRSF2 and ASXL1 mutations were detected as co-founding events in individual subclones across different CMML patient samples, and their VAFs were not altered despite clinical response, as previously described11. Distinct evolution patterns were observed, ranging from relative preservation of baseline clonal architecture to expansion and dominance of progeny subclones through successive acquisition of mutations or via LOH. As an example, in patient P11 with CN-LOH, disruption of the ring finger domain critical for E3 ligase activity via acquired biallelic CBL C384Y mutation correlated with myelomonocytic expansion 12, 13 . Subsequent acquisition of inactivating RUNX1 mutations further enabled dominance of these CBL C384Y-bearing subclones. Overall, global suppression of myelomonocytic cells was achieved after 4 cycles of 5-Aza, with re-expansion of lymphocytes to a median of 24% in patients with CR or PR (n=6) (Supplementary Figure 3). However, clonal evolution patterns did not correlate with response to HMAs. Clonal evolution with expansion of maximally mutated progeny subclones occurred in 5 out of 8 patients with favorable clinical response, while progeny subclones evolved and expanded with successive acquisition of secondary mutations or LOH events in 2 out of 3 patients with disease progression. This suggests that subclones within the CMML compartment continue to evolve irrespective of clinical response, and that response is governed by complex genetic signals and epigenetic mechanisms. Consistent with a previous report 11 , our study highlights that current understanding of CMML biology is predominantly mechanistic, and accurate correlation of specific pathogenic clones with clinical response has yet to be determined. Next generation sequencing (NGS) panels are increasingly used for molecular monitoring in CMML, but VAFs of somatic mutations alone do not adequately reflect clonal heterogeneity. Single cell sequencing provides the maximum resolution for delineation of clonal architecture, but is limited by costs and potential allelic bias 14 . Analysis of single cell colonies may be influenced by specific cytokines used in the cultures. While current clinical practice is to monitor hematologic parameters and VAFs of mutations in CMML patients, we demonstrate that subclonal hierarchies and evolution can be delineated at high resolution from standard bulk NGS data using the SubcloneSeeker computational algorithm. Inclusion of all somatic mutations rather than only driver mutations is required to solve clonal architecture with high confidence. This strategy provides deeper insights into the hierarchy of acquisition and distribution of founding and secondary mutations within individual subclones in CMML, without resorting to single cell analysis. The predictive accuracy of the computational algorithm has recently been validated in a similar study for drug-resistant breast cancer subclones, using single-cell genotyping experiments 15 . Until single-cell sequencing technologies become widely available for routine testing, computational reconstruction of clonal evolution represents the most dynamic platform available for delineation of specific mutations and subclones in leukemia, and may become a useful tool for response monitoring and potentially therapeutic decision making. Ongoing clonal evolution despite apparent clinical remission highlights gaps in the current mechanistic understanding of HMA therapy in CMML. Integration of epigenetic evolution and the influence of tumor microenvironment into clonality studies may establish strategies to unravel biological complexity and identify novel therapeutic targets in CMML. Supplementary Material 1

          Related collections

          Most cited references11

          • Record: found
          • Abstract: found
          • Article: not found

          Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study.

          Drug treatments for patients with high-risk myelodysplastic syndromes provide no survival advantage. In this trial, we aimed to assess the effect of azacitidine on overall survival compared with the three commonest conventional care regimens. In a phase III, international, multicentre, controlled, parallel-group, open-label trial, patients with higher-risk myelodysplastic syndromes were randomly assigned one-to-one to receive azacitidine (75 mg/m(2) per day for 7 days every 28 days) or conventional care (best supportive care, low-dose cytarabine, or intensive chemotherapy as selected by investigators before randomisation). Patients were stratified by French-American-British and international prognostic scoring system classifications; randomisation was done with a block size of four. The primary endpoint was overall survival. Efficacy analyses were by intention to treat for all patients assigned to receive treatment. This study is registered with ClinicalTrials.gov, number NCT00071799. Between Feb 13, 2004, and Aug 7, 2006, 358 patients were randomly assigned to receive azacitidine (n=179) or conventional care regimens (n=179). Four patients in the azacitidine and 14 in the conventional care groups received no study drugs but were included in the intention-to-treat efficacy analysis. After a median follow-up of 21.1 months (IQR 15.1-26.9), median overall survival was 24.5 months (9.9-not reached) for the azacitidine group versus 15.0 months (5.6-24.1) for the conventional care group (hazard ratio 0.58; 95% CI 0.43-0.77; stratified log-rank p=0.0001). At last follow-up, 82 patients in the azacitidine group had died compared with 113 in the conventional care group. At 2 years, on the basis of Kaplan-Meier estimates, 50.8% (95% CI 42.1-58.8) of patients in the azacitidine group were alive compared with 26.2% (18.7-34.3) in the conventional care group (p<0.0001). Peripheral cytopenias were the most common grade 3-4 adverse events for all treatments. Treatment with azacitidine increases overall survival in patients with higher-risk myelodysplastic syndromes relative to conventional care.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Design and Analysis of Single-Cell Sequencing Experiments.

            Recent advances in single-cell sequencing hold great potential for exploring biological systems with unprecedented resolution. Sequencing the genome of individual cells can reveal somatic mutations and allows the investigation of clonal dynamics. Single-cell transcriptome sequencing can elucidate the cell type composition of a sample. However, single-cell sequencing comes with major technical challenges and yields complex data output. In this Primer, we provide an overview of available methods and discuss experimental design and single-cell data analysis. We hope that these guidelines will enable a growing number of researchers to leverage the power of single-cell sequencing.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Mutation allele burden remains unchanged in chronic myelomonocytic leukaemia responding to hypomethylating agents

              The cytidine analogues azacytidine and 5-aza-2'-deoxycytidine (decitabine) are commonly used to treat myelodysplastic syndromes, with or without a myeloproliferative component. It remains unclear whether the response to these hypomethylating agents results from a cytotoxic or an epigenetic effect. In this study, we address this question in chronic myelomonocytic leukaemia. We describe a comprehensive analysis of the mutational landscape of these tumours, combining whole-exome and whole-genome sequencing. We identify an average of 14±5 somatic mutations in coding sequences of sorted monocyte DNA and the signatures of three mutational processes. Serial sequencing demonstrates that the response to hypomethylating agents is associated with changes in DNA methylation and gene expression, without any decrease in the mutation allele burden, nor prevention of new genetic alteration occurence. Our findings indicate that cytosine analogues restore a balanced haematopoiesis without decreasing the size of the mutated clone, arguing for a predominantly epigenetic effect.
                Bookmark

                Author and article information

                Journal
                8704895
                5536
                Leukemia
                Leukemia
                Leukemia
                0887-6924
                1476-5551
                12 January 2018
                27 March 2018
                September 2018
                27 September 2018
                : 32
                : 9
                : 2049-2054
                Affiliations
                [1 ]Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, USA
                [2 ]Department of Hematology, Singapore General Hospital, Singapore
                [3 ]Eccles Institute of Human Genetics, The University of Utah, Salt Lake City, UT, USA
                [4 ]Centre for Hematology, Department of Medicine, Imperial College, London, UK
                [5 ]Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT, USA
                Author notes
                Correspondence: Michael W. Deininger, MD, PhD, The University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope, Room 4280, Salt Lake City, Utah 84112-5550, 801/581-6363 phone, 801/585-0900 fax, michael.deininger@ 123456hci.utah.edu
                Article
                NIHMS933711
                10.1038/s41375-018-0050-z
                6128729
                29588547
                f921a94a-3485-47fe-9999-db9c659e05bc

                Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms

                History
                Categories
                Article

                Oncology & Radiotherapy
                Oncology & Radiotherapy

                Comments

                Comment on this article