22
views
0
recommends
+1 Recommend
0 collections
    1
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Discovery and functional characterization of a germline, CSF2RB-activating mutation in leukemia

      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 Colony stimulating factor 2 receptor beta (CSF2RB) is the shared beta-chain receptor and essential signaling component for IL-3, IL-5 and GM-CSF receptor activation. In the context of GM-CSF signaling, the ligand-specific alpha chain (CSF2RA) complexes with CSF2RB and GM-CSF ligand, forming a dodecameric complex in which the proximity of CSF2RB subunits allows associated JAK2 kinases to trans-phosphorylate 1 . CSF2RB signals through several pathways including JAK2/STAT5, PI3K/mTOR and MEK/ERK to promote survival, proliferation and differentiation (reviewed in 2 ). Both spontaneous transformation 3, 4 and random mutagenesis 4, 5 screens have shown that CSF2RB mutations can result in ligand-independent activation in vitro. However, these predicted oncogenic mutations have never been observed clinically. In fact, the only phenotypic CSF2RB mutations reported are recessive, loss-of-function, germline mutations in pulmonary alveolar proteinosis 6 . As oncology moves toward an age of personalized and molecularly-targeted treatment it will require the identification and in vitro characterization of targetable genetic lesions. Here we describe the first case of a leukemia patient harboring a germline, CSF2RB-activating point mutation (R461C) and identify small molecule inhibitors with therapeutic potential against this mutation. We obtained a pediatric T-cell acute lymphoblastic leukemia (T-ALL, Supplemental Figure 1A) with informed consent approved by the Institutional Review Boards of Oregon Health & Science University and Erasmus University Medical Center - Sophia Children’s Hospital. We isolated mononuclear cells and performed deep sequencing as previously described 7 , uncovering the CSF2RB R461C mutation at a 54% allele burden (Supplemental Figure 1B and C). The patient also presented with somatic NOTCH1-truncation, NOTCH1 missense, and PTEN point mutations, all recurrent and leukemia-associated mutations 8 (other observed mutations in Supplemental Table 1). R461C was later confirmed by Sanger sequencing as a heterozygous, germline variant using a minimal residual disease day 79 sample for normal DNA (Supplemental Figure 1D). CSF2RB R461C has never been previously described in cancer patients (including public databases) but is listed as a SNP in the 1000 Genomes database 9 (rs371045078) at a low allele frequency (MAF=0.000998, 5 observations in 1000 Genomes 9 ; MAF=0.000272, 33 observations in the Exome Aggregation Consortium 10 ) in exclusively heterozygous cases. Prior work has established that cancer-predisposing mutations exist in the germline of healthy individuals at frequencies as high as 1.1%, but those frequencies elevate to over 8% in children and adolescents with cancer, most of which demonstrate no family history of cancer 11 . Congenital mutations in CSF3R have also been observed to precede the development of acute myeloid leukemia 12 . In addition, there are numerous reports of families with gain-of-function germline mutations in receptors with similar biology as CSF2RB (e.g., CSF3R 13 , EPOR 14 , MPL 15 ) and in many of these pedigrees the affected family members do not develop overt leukemia, though they sometimes exhibit elevated blood counts. The location of this variant, within the putative membrane-spanning portion of the receptor, is of particular interest as this region is a hot spot for activating mutations in vitro 5 . Previously, mutations altering the number of membrane-spanning or membrane-adjacent unpaired cysteines have been shown to activate the IL-7 receptor in T-ALL 16 and RET in medullary thyroid carcinoma 17 . CSF2RB has been shown to be partially activated in vitro by replacement of the extracellular domain with a short, cysteine-containing sequence 4 . Lastly, multiple sequence alignment of CSF2RB homologues demonstrated that arginine 461 is conserved across most mammals (Supplemental Figure 2) increasing the potential that R461C could have functional relevance and cooperate with canonical leukemogenic mutations. Accordingly, we hypothesized that R461C could activate CSF2RB-signaling and contribute to the patient’s leukemia. We cloned CSF2RB R461C into the pMXs-IRES-Puro plasmid and transfected into IL-3-dependent murine Ba/F3 cells by electroporation. Stably transfected cells were selected using two weeks of continual 2µg/ml puromycin selection prior to performing IL-3 withdrawal transformation assays as previously described 7 . R461C conferred factor-independent growth while wild-type (WT) CSF2RB did not (Figure 1A, Supplemental Figure 3). The full CSF2RB transgene was sequence confirmed in two biologically replicate, IL-3 independent cell lines and used for further studies. We identified that CSF2RB R461C expression resulted in an accumulation of surface CSF2RB protein, relative to WT, prior to IL-3 withdrawal when assayed by flow cytometry (Supplemental Figure 4). Furthermore, immunoblot analysis showed that the R461C protein is significantly increased in IL-3-independent cells relative to WT cells grown in IL-3 (Figure 1B, details and all antibodies included Supplemental methods and Supplemental Table 2). As both exogenous versions of CSF2RB were expressed by identical promoters we rationalized that the accumulation was due to changes in CSF2RB protein stability. We performed cycloheximide time course experiments using 100ug/ml cycloheximide in DMSO followed by staining with PE-conjugated anti-CSF2RB to determine alterations in surface protein stability. The R461C mutant possessed a prolonged surface half-life relative to WT CSF2RB (6.5 vs 3.6 hours; Figure 1C). Given the presence of a novel cysteine and increased stability in the R461C mutant, we investigated disulfide-linked receptor oligomerization by immunoblot under non-reducing conditions. Comparing the WT- and R461C-expressing cells, we observed CSF2RB-containing complexes of ~500 kDa that showed a reduced intensity when 2-mercaptoethanol was added to the lysate (normally CSF2RB runs at 125 kDa, Figure 1D), indicating R461C enables novel disulfide interactions with additional CSF2RB monomers or other endogenous receptors. Previous studies have shown that mutations within receptor transmembrane domains can result in constitutive receptor oligomerization and activation 15–17 . The role of the CSF2RB transmembrane domain in receptor stability and signal transduction has not been characterized 1 . Even the boundaries of the membrane-spanning region have not been empirically studied, though it is commonly assumed to span residues 444–460 based on early domain modeling 5 . Various modern models consistently predict a much larger domain (Supplemental Figure 5), including residue 461, suggesting R461C might be within the transmembrane domain and alter receptor-receptor interactions. Another possibility is that the loss of a charged residue near the membrane boundary could result in shifting or fluidity of the transmembrane domain of the mutant receptor. Using immunoblot and co-immunoprecipitation analysis, we showed that transformed Ba/F3 cells expressing CSF2RB R461C exhibited constitutive receptor phosphorylation and signaled through several pathways, including STAT5, PI3K/mTOR1, and MEK/ERK (Figure 2A and B). To identify therapeutically targetable pathways we utilized a 104 small molecule inhibitor library screen, as previously described 7, 18 , and compared IC50 values for R461C-expressing cells to WT-expressing cells. JAK inhibitors (Tofacitinib, Ruxolitinib, and AZD1480) constituted the three top hits in the screen (Figure 2C, Supplemental Figure 6A–C, full screen Supplemental Table 3). These treatments were specific and cytotoxic, inducing apoptosis in R461C-expressing cells (Figure 2D and Supplemental Figure 6D). While these inhibitors have a differential specificity for targeting JAK kinases (Tofacitinib: JAK2/3, Ruxolitinib: JAK1/2, AZD1480: JAK2), the strongest target is JAK2. We also observed reduced levels of total JAK2 in R461C cells (Figure 2B), which is consistent with JAK2 activation and then degradation through a STAT5/SOCS-1 negative feedback loop 19 . These results suggest a dependency on the JAK/STAT pathway that is consistent with ligand-independent activation of CSF2RB in R461C-expressing cells. Targeted inhibition of JAK2 using AZD1480 quickly reduced levels of phosphorylated STAT5, validating the specific nature of the inhibition and highlighting JAK2 as the key mediator of CSF2RB R461C signaling (Figure 2E). In sequencing 449 primary hematopoietic malignancies, we found 7 additional CSF2RB mutations (Supplemental Table 4) but none were membrane spanning and all 7 were incapable of transforming Ba/F3 cells (data not shown). Several other CSF2RB-activating mutations have been observed in vitro, but have not been observed in primary patient samples. CSF2RB R461C is a rare germline variant and recent studies of germline cancer-predisposing mutations have not included CSF2RB 11 , possibly because of the lack of previous evidence showing the oncogenic potential of CSF2RB. Our data would support the inclusion of CSF2RB in future studies. CSF2RB R461C is a transforming mutation in vitro, and further investigation is necessary to determine its status as a predisposing or cooperative congenital mutation. CSF2RB has previously been shown to become factor-independent through in vitro mutagenesis screens. One such screen successfully predicted R461C to be an active variant 5 (though insufficient to transform Ba/F3 cells in a short-term withdrawal assay), demonstrating the potential for predictive mutagenesis screens. In spite of this in vitro evidence, there has never before been a reported case of cancer with a CSF2RB-activating mutation. We have demonstrated for the first time that a targetable CSF2RB variant found in a human leukemia confers factor-independent growth, receptor phosphorylation and accumulation, and constitutive JAK/STAT pathway activation. Our findings contribute to ongoing efforts to identify potential germline predisposition mutations in pediatric cancers, and are consistent with observations that genetic alterations activating cytokine receptor pathways are common in leukemia. Our research highlights the need for basic research to investigate and characterize the functional role of the CSF2RB transmembrane domain in signaling and recycling of the receptor. Finally, JAK inhibitors blocked the growth of R461C-transformed cells, thereby providing a therapeutic rationale to consider JAK inhibitors in any future cases of CSF2RB-activated leukemias. Supplementary Material 1 2

          Related collections

          Most cited references14

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

          Oncogenic CSF3R mutations in chronic neutrophilic leukemia and atypical CML.

          The molecular causes of many hematologic cancers remain unclear. Among these cancers are chronic neutrophilic leukemia (CNL) and atypical (BCR-ABL1-negative) chronic myeloid leukemia (CML), both of which are diagnosed on the basis of neoplastic expansion of granulocytic cells and exclusion of genetic drivers that are known to occur in other myeloproliferative neoplasms and myeloproliferative-myelodysplastic overlap neoplasms. To identify potential genetic drivers in these disorders, we used an integrated approach of deep sequencing coupled with the screening of primary leukemia cells obtained from patients with CNL or atypical CML against panels of tyrosine kinase-specific small interfering RNAs or small-molecule kinase inhibitors. We validated candidate oncogenes using in vitro transformation assays, and drug sensitivities were validated with the use of assays of primary-cell colonies. We identified activating mutations in the gene encoding the receptor for colony-stimulating factor 3 (CSF3R) in 16 of 27 patients (59%) with CNL or atypical CML. These mutations segregate within two distinct regions of CSF3R and lead to preferential downstream kinase signaling through SRC family-TNK2 or JAK kinases and differential sensitivity to kinase inhibitors. A patient with CNL carrying a JAK-activating CSF3R mutation had marked clinical improvement after the administration of the JAK1/2 inhibitor ruxolitinib. Mutations in CSF3R are common in patients with CNL or atypical CML and represent a potentially useful criterion for diagnosing these neoplasms. (Funded by the Leukemia and Lymphoma Society and others.).
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Kinase pathway dependence in primary human leukemias determined by rapid inhibitor screening.

            Kinases are dysregulated in most cancers, but the frequency of specific kinase mutations is low, indicating a complex etiology in kinase dysregulation. Here, we report a strategy to rapidly identify functionally important kinase targets, irrespective of the etiology of kinase pathway dysregulation, ultimately enabling a correlation of patient genetic profiles to clinically effective kinase inhibitors. Our methodology assessed the sensitivity of primary leukemia patient samples to a panel of 66 small-molecule kinase inhibitors over 3 days. Screening of 151 leukemia patient samples revealed a wide diversity of drug sensitivities, with 70% of the clinical specimens exhibiting hypersensitivity to one or more drugs. From this data set, we developed an algorithm to predict kinase pathway dependence based on analysis of inhibitor sensitivity patterns. Applying this algorithm correctly identified pathway dependence in proof-of-principle specimens with known oncogenes, including a rare FLT3 mutation outside regions covered by standard molecular diagnostic tests. Interrogation of all 151 patient specimens with this algorithm identified a diversity of kinase targets and signaling pathways that could aid prioritization of deep sequencing data sets, permitting a cumulative analysis to understand kinase pathway dependence within leukemia subsets. In a proof-of-principle case, we showed that in vitro drug sensitivity could predict both a clinical response and the development of drug resistance. Taken together, our results suggested that drug target scores derived from a comprehensive kinase inhibitor panel could predict pathway dependence in cancer cells while simultaneously identifying potential therapeutic options.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Familial essential thrombocythemia associated with a dominant-positive activating mutation of the c-MPL gene, which encodes for the receptor for thrombopoietin.

              One Japanese pedigree of familial essential thrombocythemia (FET) inherited in an autosomal-dominant manner is presented. A unique point mutation, serine 505 to asparagine 505 (Ser505Asn), was identified in the transmembrane domain of the c-MPL gene in all of the 8 members with thrombocythemia, but in none of the other 8 unaffected members in this FET family. The Ba/F3 cells expressing the mutant Asn505 acquired interleukin 3 (IL-3)-independent survival capacity, whereas those expressing wild-type Ser505 did not. The autonomous phosphorylation of Mek1/2 and Stat5b was observed in the mutant Ba/F3 cells in the absence of IL-3. The former was also found in platelets derived from the affected individual in the absence of thrombopoietin. These results show that the Asn505 is an activating mutation with respect to the intracellular signaling and survival of the cells. This is the first report of FET deriving from a dominant-positive activating mutation of the c-MPL gene.
                Bookmark

                Author and article information

                Journal
                8704895
                5536
                Leukemia
                Leukemia
                Leukemia
                0887-6924
                1476-5551
                19 April 2016
                27 April 2016
                September 2016
                01 March 2017
                : 30
                : 9
                : 1950-1953
                Affiliations
                [1 ]Cancer Biology Program, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
                [2 ]Oregon Health & Science University, Knight Cancer Institute, Portland, OR
                [3 ]Howard Hughes Medical Institute, Portland, OR
                [4 ]Department of Cell & Developmental Biology, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
                [5 ]Department of Pediatric Oncology, Erasmus University Medical Center, Sophia Children’s Hospital, Rotterdam, Netherlands
                [6 ]Division of Hematology and Medical Oncology, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
                Author notes
                Corresponding Author: Anupriya Agarwal, Address: OHSU Mailcode L592, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, agarwala@ 123456ohsu.edu , 503-494-7599, 503-494-3688
                Article
                NIHMS777047
                10.1038/leu.2016.95
                5014660
                27118405
                7ddfc12d-87f4-4a63-968b-f21416dc48ce

                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