Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Apr 1.
Published in final edited form as: Hematol Oncol Clin North Am. 2019 Dec 11;34(2):379–391. doi: 10.1016/j.hoc.2019.10.003

Targeting aberrant splicing in myelodysplastic syndromes: biologic rationale and clinical opportunity

Andrew M Brunner 1, David P Steensma 2
PMCID: PMC7045909  NIHMSID: NIHMS1542430  PMID: 32089217

Introduction

Myelodysplastic syndromes (MDS) are cancers of the bone marrow with diverse clinical presentations, collectively characterized by clonally restricted hematopoiesis, cytopenias, dysplastic cell morphology, functional blood cell defects, and a risk of clonal evolution including progression to acute leukemia.1-3 Patients with MDS face complications related to or exacerbated by bone marrow failure and have a high symptom burden,4,5 and higher-risk forms of MDS have a poor prognosis with a median survival measured ranging from months to a few years.6,7 Treatment of lower-risk MDS is often focused on alleviating symptoms related to ineffective clonal hematopoiesis – most commonly centering on amelioration of anemia – while in higher-risk MDS, therapy is usually intended to alter the course of disease and prolong survival.8 Currently, therapeutic options for MDS are limited, and no new therapies have been approved by regulatory authorities for MDS-related indications for nearly 15 years,9 although that may change soon.

While almost all patients with MDS have somatic mutations in hematopoietic cells (mostly in genes encoding epigenetic modifiers, spliceosome components, and transcription factors) to date, no available therapies are particularly specific for a given molecularly defined subset of MDS. For instance, while the majority of patients with MDS with del(5q) are likely to have improvements to their transfusion burden when treated with lenalidomide10, some do not, and conversely a smaller proportion of patients without del(5q) may also see improvement in transfusion needs and transfusion independence during lenalidomide therapy.11,12 Similarly, the hypomethylating agents (HMAs) azacitidine or decitabine are routinely employed as disease-modifying chemotherapeutics for patients with higher risk disease, but mutation patterns have only modest effect on clinical response and such data are not used in practice for treatment decision-making.13,14,15 New therapies that selectively target malignant clones while sparing healthy bone marrow (and ideally thus providing favorable toxicity profile) are desperately needed.

A challenge in developing new clinical therapeutics has been the limitations of preclinical in vivo models of MDS16 – as a result, many therapeutics in MDS to date have been “borrowed” from acute myeloid leukemia or other hematological malignancies in which such models are better developed. This barrier to drug development has been changing thanks to a greater understanding of the genetic composition of MDS and novel mouse models that provide insight into MDS genetics.17-19 Sequencing efforts have identified frequent recurrent mutations in MDS that allow for the identification of clonal origin and how disease evolves over time.3,20-22 These efforts identified frequent, recurrent mutations in pre-mRNA splicing that appear to be early events in the development of MDS.23-27 Splicing factor mutations are present in more than half of MDS and chronic myelomonocytic leukemia (CMML) cases, and are associated with specific phenotypic findings, such as SF3B1 and ring sideroblasts,28,29 or SRSF2 enrichment in CMML.30

In this review, we discuss the rationale for targeting pre-mRNA splicing in MDS, including pre-clinical insights into the effects of splicing factor mutations and how they may present therapeutic vulnerabilities, and we summarize early clinical data from studies in progress. The spliceosome is an appealing target due to the prevalence of splicing factor mutations in MDS, presence of mutations in early malignant progenitors, and potential for exploiting synthetic lethality strategies. Nonetheless, splicing is such a fundamental biological process that the therapeutic window of splicing modulation may be narrow, and much work is needed to understand how to target splicing and test novel therapeutic combinations in order to change the MDS treatment paradigm.

Preclinical Rationale

Somatic mutations in pre-mRNA splicing factors are seen in a number of cancers, including solid tumors (notably uveal melanoma) and lymphoid malignancies such as chronic lymphocytic leukemia (CLL).31,27,32,33,34 These mutations are enriched in myeloid neoplasms, specifically in MDS, CMML, and AML (especially secondary AML arising from MDS). In MDS, mutations in splicing factors occur most often in SF3B1, U2AF1, SRSF2, and less frequently ZRSR2.25 Combined, splicing factor mutations represent the most frequently acquired recurrently mutated gene in MDS,35 and thus are of interest as possible therapeutic targets.36

Splicing factor mutations result in characteristics splicing alterations which can then be measured as a biomarker.37 Normal splicing proceeds through the identification of a 5’ splice site and 3’ splice site at either end of each intron to be spliced together, as well as a branch point sequence upstream of the 3’ splice site and polypyrimidine tract that are critical for the identification of exons and splicing accuracy.38 In the initial steps of mRNA splicing, the 5’ site is identified and bound to the branch point sequence by the U1 and U2 snRNPs, the latter of which contains SF3B1 which binds to the branch point sequence. U2AF1 is involved in recognizing the 3’ splice site, while SRSF2, a member of the serine/arginine (SR) protein family, functions as a splicing enhancer, with a role in exon recognition. ZRSR2 appears to have a role analogous to U2AF1 but as part of the minor splicing complex. Mutations in these splicing factors result in characteristic alterations in pre-mRNA splicing based on these functions:39 mutations in SF3B1 result in alternative branchpoint usage,40,41 mutations in U2AF1 are associated with altered splice site recognition,42 and mutations in SRSF2 result in altered recognition of exon splicing enhancers, inducing altered splicing of a number of proteins including critical regulators of hematopoiesis.43

At the same time, splicing does not happen in a vacuum; rather, splicing is coupled with RNA transcription in a dynamic fashion.44-46 This has the effect that any perturbations in splicing can cascade into the normal processing of mRNA, potentially slowing elongation. Alterations in RNA transcription may lead to increased RNA:DNA hybrid structures termed R-loops,47 which can effect DNA integrity and leading to single strand breaks.48,49 In response, ataxia telangiectasia and rad3 related (ATR) signaling is activated, with eventual R-loop resolution.50 The downstream effects of alternative splicing on such integral cellular functions as pre-mRNA transcription may provide further targets for therapeutic intervention in splicing factor mutated cells.

Mutual Exclusivity of Splicing Factor Mutations

Nearly all MDS cases can be characterized by at least one recurrent somatic mutation using a relatively small panel of ~100 myeloid genes of interest,35,51-53 the most frequently mutated family of genes being those in the splicing apparatus.51 Interestingly, these mutations are almost always mutually exclusive of one another;26,31,54 meaning that (with rare exception) a patient sample will harbor only one splicing mutation. Instances with more than one splicing factor mutation identified may often represent separate subclonal events. In addition, splicing factor mutations are heterozygous events, and the presence of the wild-type allele appears necessary for cell survival.55 These factors suggest that mutations in the splicing apparatus, although frequent in MDS, nonetheless result in a state of dependence upon some wildtype splicing function, with strong negative selection for homozygous mutations or mutations in multiple splicing factor genes in the same cell.

Splicing Factor Mutations as Early Clonal Events

Serial sequencing of MDS patient samples over time shows that dynamic clonal changes may occur and can be associated with changes in clinical and pathologic behavior.56,57 Some mutations, including those commonly seen in secondary AML such as FLT3, PTPN11, WT1, IDH1, NPM1, IDH2 and NRAS, are more commonly late events, occurring around the time of disease transformation.57 This contrasts with splicing mutations which are usually present early in the course of disease.23,58,59 Mutations in SF3B1, SRSF2, or U2AF1 tend to persist after treatment even when clinical response occurs,58,60 further supporting the hypothesis that these mutations are present in an early progenitor and critical to the initial development of MDS. This also suggests that therapies targeting splicing factor mutations may be more likely to impact the malignant progenitor clone and perhaps thus more likely to elicit durable responses.

Preclinical Models of Splicing Factor Mutant MDS

Efforts to recapitulate genomically accurate murine models of myelodysplasia have led to new models for testing therapeutic combinations for treating MDS and greater insights into its pathogenesis (Figure 1).61-63 One such murine model utilized inducible SRSF2 P95H, a mutation hotspot in human MDS. In this murine model, the investigators showed that homozygous loss of SRSF2 resulted in failed hematopoiesis, but conditional expression of P95H mutations in SRSF2 yielded a myeloid dysplasia phenotype.43 Splicing factor mutations do not appear to result in loss of function (which may be lethal), but rather alter splicing preferences; in the SRSF2 murine model, disruption of SRSF2 function resulted in alternative splicing of several key hematopoietic regulators like EZH2. In a different inducible murine model of U2AF1 S34F, heterozygosity for this mutation recapitulated an MDS phenotype and resulted in aberrant splicing involving genes recurrently mutated in MDS and AML, among others.64 Curiously, these inducible models of splicing factor mutations can recapitulate some aspects of the MDS phenotype, but splicing mutant cells have a competitive disadvantage with wildtype cells in co-transplantation experiments, in contrast to what is seen in human MDS (Figure 1).

Figure 1.

Figure 1.

General Schema of Splicing Factor Mutated MDS Murine Models.

A prototype murine model of MDS. MDS progenitor cells may be introduced into mice through a number of methods, including using patient samples or engineering mice with inducible splicing factor mutations. After infusion/induction, these cells can be tested either comparing wildtype (WT) or mutant (SFMUT) populations alone, or in combination with a drug of interest (e.g. spliceosome modulator, ATR inhibitor). Competitive transplantation studies show exhaustion of the clones harboring splicing factor mutations, that is hastened in homozygous mutations. Spliceosome modulation appears to improve survival when administered to mice with splicing factor mutated leukemic xenografts.

Synthetic Lethality in Splicing Factor Mutant Cells

A key question raised by the frequency of splicing factor mutations is whether and how these mutations could be targeted for therapeutic effect. Using murine models with mutated SRSF2 or U2AF1, the effects of specific chemotherapies have been testing for signals of selective activity against mutant cells (Figure 1).62,63 In one study, mice engineered to express the SRSF2 P95H mutation received the spliceosome modulator E7107, an SF3B1 inhibitor. In this model, there was no significant difference in survival between SRSF2P95H/+ mice and SRSF2WT mice, but those mice with heterozygous SRSF2 P95H mutations who were exposed to E7107 had prolonged survival.62 Similarly, in a study evaluating hematopoietic cells expressing U2AF1S34F/+, exposure to sudemycin compounds, which bind to the SF3B1 protein in the spliceosome, attenuated expansion of U2AF1 mutated progenitor cells in a mouse transplant model.63 These and other studies have led to a hypothesized mechanism of action for spliceosome modulators: there is a degree of “tolerable” splicing dysfunction within a cell, but further alteration of splicing results in a state where the cell can no longer compensate and leads to selective cell death (Figure 2).

Figure 2.

Figure 2.

Putative Therapeutic Mechanism of Spliceosome Modulator Therapies in Splicing Factor Mutated Cells.

At diagnosis, patients with MDS have largely malignant hematopoiesis arising from MDS progenitors; in splicing factor mutated MDS, normal splicing function will be diminished and maintained by the wildtype allele. Modulators of the spliceosome are thought to further alter splicing; healthy progenitor cells are able to “tolerate” this modulation, while cells with a splicing factor mutation have excessive spliceosome dysfunction leading to cell death. This hypothetically results in the expansion of the healthy cell compartment and restoration of normal hematopoiesis.

Interaction with Transcription and DNA Damage

Because splicing occurs in conjunction with mRNA transcription, alterations in splicing can affect the efficiency and integrity of transcription. As noted above, alternative splicing slows the replication fork and results in DNA:RNA hybrid structures termed “R-loops.” R-loops are unstable structures which displace the non-hybridized DNA strand, resulting in single-stranded DNA which activates a DNA damage response mediated through ataxia telangiectasia and Rad3 related (ATR) signaling.48,65,66 Interrupting this DNA damage response may hamper genomic integrity and lead to selective apoptosis of splicing factor mutant cells.67-69 Alternatively, cumulative DNA damage as well as the buildup of alternatively spliced isoforms may yield tumor enriched antigens that enhance immunogenicity on their own or in combination with immuno-oncologic therapies.70,71

Clinical Studies

Several therapies have recently entered clinical trials to test the preclinical rationale of targeting the spliceosome itself or associated pathways (Table 1). Although clinical results are preliminary, early signals from these studies are likely to greatly influence our understanding of spliceosome function in both wildtype and mutated cells, including the role of splicing factor mutations in MDS pathogenesis and treatment.

Table 1.

Active Clinical Trials for Splicing-Factor Mutated MDS

Drug Target NCT Number Notes
H3B-8800 Small molecule modulator of SF3B1 Dose escalation on two schedules: 5 days on, 9 days off, or 21 of 28 days.
GSK3326595 Reversible inhibitor of PRMT5 Dose escalation followed by three parallel study expansion arms in monotherapy or in combination with azacitidine
JNJ-64619178 Inhibitor of PRMT5 Dose escalation and tolerability, evaluating in lower-risk MDS
AZD6738 Small molecular ATR inhibitor Single agent AZD6738 in higher-risk MDS, CMML, and lower-risk MDS

Modulating Spliceosome Function

Some of the first human studies to evaluate a spliceosome modulator investigated the compound E7107. Two phase 1 studies of E7107 enrolled patients with advanced solid tumors (, ).72,73 Although no patients on these studies had a diagnosis of MDS, the toxicities encountered may be informative to spliceosome modulators in general. A total of 56 patients were enrolled between the two studies; the maximum tolerated dose of E7107 as an intravenous infusion was 4.3mg/m2 when given on days 1 and 8 of a 21-day cycle, or 4mg/m2 when dosed on days 1, 8, and 15 of a 28-day cycle. Dose-limiting toxicities (DLTs) across the two studies included gastrointestinal toxicity (diarrhea, vomiting, nausea, abdominal cramping), anorexia, dehydration, and myocardial infarction. Also noted were three patients who experienced irreversible visual toxicity; one with blurred vision and central scotomas, and two others with vision loss; these events resulted in cessation of further study of this agent. Of note, investigators did observe a dose dependent change in mRNA splicing in cells from E7107 treated patients.73

H3B-8800 is an orally bioavailable small molecule modulator of SF3B1 which shares a pladienolide chemical backbone but is molecularly distinct from E7107. Preclinical data confirmed selective cytotoxic activity against splicing factor mutated leukemia in xenograft models.74 H3B-8800 has been studied in a phase I dose escalation trial enrolling patients with AML, MDS, and CMML, with preliminary data reported to date ().75,76 A total of 81 patients were enrolled in dose escalation cohorts evaluating doses of 1-40mg administered either 5 days on/9 days off or on days 1-21 of a 28 day cycle. DLTs included bone marrow failure in a patient with lower-risk MDS treated at 7mg on the 5 days on/9 days off schedule, and nausea and QTcF interval prolongation at 20mg on the schedule of days 1-21 of 28. There was a dose-dependent increase in plasma levels of H3B-8800, as well as evidence of on-target alternative splicing in hematopoietic cells proportional to dose increases.76 The drug was generally tolerable with several patients receiving treatment for over two years duration.

Other spliceosome modulators are under development including sudemycin D6,77 Pladienolide-B, and FD-895, although none have yet been tested in human subjects;78 there is also significant interest in novel combination therapies incorporating spliceosome modulation with other therapies in MDS.

Inhibition of PRMT5

Protein arginine methyltransferase 5 (PRMT5) is essential to the formation of small nuclear ribonucleoproteins (snRNPs) and the spliceosome machinery. Inhibition of PRMT5 results in increased alternative splicing patterns,79 and also may impair homologous recombination DNA repair.80 As such, PRMT5 is being explored as a target among tumors with spliceosome mutations.

GSK3326595 is a specific and reversible inhibitor of PRMT5 and has preclinical activity in a number of tumors; cell lines treated with GSK3326595 showed increased alternative splicing, and p53 activation due to alternative splicing of MDM4 was observed.81 This compound has been studied in a phase I dose escalation study in solid tumors and non-Hodgkin’s lymphoma ()82 and more recently is under investigation in myeloid neoplasms including AML, MDS, and CMML ().

Another PRMT5 inhibitor in early human study is JNJ-64619178. This agent is orally bioavailable and shows preclinical efficacy in a number of tumors including AML.83 A study in patients with non-Hodgkin lymphoma and advanced solid tumors, as well as lower-risk MDS, is currently enrolling ().

Splicing Induced Immune Responses

Splicing factor mutations may result in generation of clonal cell-restricted neoantigens that can be recognized by the immune system.71 A number of studies are actively investigating the role of immunotherapy in MDS as monotherapy or in combination with hypomethylating agents, including the anti-CTLA-4 therapy ipilimumab, and the anti-PD-1 or PD-L1 therapies nivolumab, pembrolizumab, and atezolizumab.84,85 While not specific to MDS harboring splicing factor mutations per se, analysis of mutation patterns and clinical responses in patients enrolled on these studies will be informative. Preliminary data suggests relatively low single agent response rates to immune checkpoint inhibitors, restricted largely to arms containing CTLA-4 inhibition.84 Moreover, a number of patients have experienced immune-related toxicities, including pulmonary complications, that will need to be mitigated if these agents have a future in MDS.

DNA Damage Response

As noted, splicing factor mutations have effects on RNA transcription resulting in the activation of a DNA damage response. Early data suggests that this occurs by recruitment of RPA to the area of R-loop formation, activating ATR signaling and the CHK1-Wee1 axis.68 Inhibiting this DNA damage response may provide another target in splicing factor mutated as well as splicing wildtype MDS.

AZD6738 is an orally bioavailable selective inhibitor of ATR that has been studied in solid tumors as monotherapy and, since it may potentiate chemotherapy-induced DNA damage, paired with a number of other agents,86,87 including other DNA repair agents like poly-(ADP ribose) polymerase (PARP).88 Mutations in the spliceosome result in constitutively increased R-loop formation and resultant ATR activation compared to wildtype cells.68 Blunting the DNA damage response by inhibiting ATR results in synthetical lethal death preferentially in splicing factor mutated cells.67 A clinical trial of AZD6738 in MDS and CMML is currently underway to test this hypothesis (). Other targets in this pathway, such as CHK1 or Wee1, may provide avenues for future study as well.

Summary/Discussion

The discovery of recurrent genetic mutations in MDS patient samples promises to bring an immense arsenal of targeted therapies to this extremely heterogeneous and treatment-refractory group of hematologic malignancies. A curious finding is the central, prominent role that mutations in the spliceosome machinery seem to play in this disease. These mutations occur early in the evolution of MDS and persist during treatment and over time. Splicing factor mutations are mutually exclusive of one another, and while each mutated splicing factor results in distinct and different downstream effects on splice isoforms, there are a number of common mis-spliced regulatory proteins. For all that has been discovered regarding splicing factor mutations in MDS, numerous questions remain, including how spliceosome mutations confer a clonal advantage and how best to exploit these mutations in the treatment of patients with this cancer. Nonetheless, the field is dynamic and evolving, with a number of currently enrolling trials targeting the pathways outlined in this review. Moreover, the identification of multiple potential targets against splicing factor mutations suggests the promise of novel:novel targeted combinations for patients with MDS, which may spare traditional chemotherapies altogether.

Synopsis.

Myelodysplastic syndromes are enriched for somatic mutations in the pre-mRNA splicing apparatus, with recurrent acquired mutations most commonly occurring in SF3B1, SRSF2, U2AF1, or ZRSR2. These mutations appear to be early events in the pathogenesis of disease, and given their frequency and central role in leukemogenesis, are of interest as potential therapeutic targets. Currently, clinical trials are exploring targets that either directly affect the spliceosome (splicing modulators e.g. H3B8800 or PRMT5 inhibitors e.g. GSK3326595) or that exploit vulnerabilities created by alternative splicing (ATR inhibitors e.g. AZD6738). Future research is needed to explore novel approaches and combinations of therapies and understand how these mutations lead to clonal dominance.

Key Points.

  1. Myelodysplastic syndromes (MDS) are enriched for somatic mutations in proteins involved in pre-mRNA splicing, specifically SF3B1, SRSF2, U2AF1, or ZRSR2

  2. Alterations in pre-mRNA splicing in MDS may expose therapeutic vulnerabilities related to splicing itself or to effects caused by altered splicing in the cells

  3. Identification of multiple targets in splicing factor mutated cells may lead to novel drugs or novel drug combinations for the treatment of MDS and other myeloid neoplasms in the future

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Steensma DP. Myelodysplastic Syndromes. Mayo Clinic Proceedings. 2015;90(7):969–983. doi: 10.1016/j.mayocp.2015.04.001 [DOI] [PubMed] [Google Scholar]
  • 2.Brunner AM, Steensma DP. Recent advances in the cellular and molecular understanding of myelodysplastic syndromes: implications for new therapeutic approaches. Clin Adv Hematol Oncol. 2018;16(1):56–66. [PMC free article] [PubMed] [Google Scholar]
  • 3.da Silva-Coelho P, Kroeze LI, Yoshida K, et al. Clonal evolution in myelodysplastic syndromes. Nat Commun. 2017;8:15099. doi: 10.1038/ncomms15099 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Brunner AM, Blonquist TM, Hobbs GS, et al. Risk and timing of cardiovascular death among patients with myelodysplastic syndromes. Blood Adv. 2017;1(23):2032–2040. doi: 10.1182/bloodadvances.2017010165 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Steensma DP. Graphical representation of clinical outcomes for patients with myelodysplastic syndromes. Leukemia & Lymphoma. 2016;57(1):17–20. doi: 10.3109/10428194.2015.1061191 [DOI] [PubMed] [Google Scholar]
  • 6.Greenberg P, Cox C, LeBeau MM, et al. International Scoring System for Evaluating Prognosis in Myelodysplastic Syndromes. Blood. 1997;89(6):2079–2088. [PubMed] [Google Scholar]
  • 7.Greenberg PL, Tuechler H, Schanz J, et al. Revised International Prognostic Scoring System for Myelodysplastic Syndromes. Blood. 2012;120(12):2454–2465. doi: 10.1182/blood-2012-03-420489 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Steensma DP. Myelodysplastic syndromes current treatment algorithm 2018. Blood Cancer Journal. 2018;8(5):47. doi: 10.1038/s41408-018-0085-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Sekeres MA, Steensma DP. Boulevard of Broken Dreams: Drug Approval for Older Adults With Acute Myeloid Leukemia. JCO. 2012;30(33):4061–4063. doi: 10.1200/JCO.2012.44.2962 [DOI] [PubMed] [Google Scholar]
  • 10.List A, Dewald G, Bennett J, et al. Lenalidomide in the Myelodysplastic Syndrome with Chromosome 5q Deletion. New England Journal of Medicine. 2006;355(14):1456–1465. doi: 10.1056/NEJMoa061292 [DOI] [PubMed] [Google Scholar]
  • 11.Raza A, Reeves JA, Feldman EJ, et al. Phase 2 study of lenalidomide in transfusion-dependent, low-risk, and intermediate-1–risk myelodysplastic syndromes with karyotypes other than deletion 5q. Blood. 2008; 111(1): 86–93. doi: 10.1182/blood-2007-01-068833 [DOI] [PubMed] [Google Scholar]
  • 12.Toma A, Kosmider O, Chevret S, et al. Lenalidomide with or without erythropoietin in transfusion-dependent erythropoiesis-stimulating agent-refractory lower-risk MDS without 5q deletion. April 2016. https://hal-univ-rennes1.archives-ouvertes.fr/hal-01321412. Accessed July 15, 2019. [DOI] [PubMed]
  • 13.Itzykson R, Kosmider O, Cluzeau T, et al. Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syndromes and low blast count acute myeloid leukemias. Leukemia. 2011;25(7): 1147–1152. doi: 10.1038/leu.2011.71 [DOI] [PubMed] [Google Scholar]
  • 14.Bejar R, Lord A, Stevenson K, et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood. 2014;124(17):2705–2712. doi: 10.1182/blood-2014-06-582809 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Schroeder MA, DeZern AE. Do somatic mutations in de novo MDS predict for response to treatment? Hematology. 2015;2015(1):317–328. doi : 10.1182/asheducation-2015.1.317 [DOI] [PubMed] [Google Scholar]
  • 16.Beurlet S, Chomienne C, Padua RA. Engineering mouse models with myelodysplastic syndrome human candidate genes; how relevant are they? Haematologica. 2013;98(1):10–22. doi: 10.3324/haematol.2012.069385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhou T, Kinney MC, Scott LM, Zinkel SS, Rebel VI. Revisiting the case for genetically engineered mouse models in human myelodysplastic syndrome research. Blood. 2015;126(9):1057–1068. doi: 10.1182/blood-2015-01-624239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Song Y, Rongvaux A, Taylor A, et al. A highly efficient and faithful MDS patient-derived xenotransplantation model for pre-clinical studies. Nature Communications. 2019; 10. doi: 10.1038/s41467-018-08166-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Suragani RNVS, Cadena SM, Cawley SM, et al. Transforming growth factor-β superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat Med. 2014;20(4):408–414. doi: 10.1038/nm.3512 [DOI] [PubMed] [Google Scholar]
  • 20.Hsu J, Reilly A, Hayes BJ, et al. Reprogramming identifies functionally distinct stages of clonal evolution in myelodysplastic syndromes. Blood. 2019;134(2):186–198. doi: 10.1182/blood.2018884338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Polgarova K, Vargova K, Kulvait V, et al. Somatic mutation dynamics in MDS patients treated with azacitidine indicate clonal selection in patients-responders. Oncotarget. 2017;8(67): 111966–111978. doi: 10.18632/oncotarget.22957 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Uy GL, Duncavage EJ, Chang GS, et al. Dynamic Changes in the Clonal Structure of MDS and AML in Response to Epigenetic Therapy. Blood. 2015;126(23):610–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Graubert TA, Shen D, Ding L, et al. Recurrent mutations in the U2AF1 splicing factor in myelodysplastic syndromes. Nat Genet. 2012;44(1):53–57. doi : 10.1038/ng.1031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Makishima H, Visconte V, Sakaguchi H, et al. Mutations in the spliceosome machinery, a novel and ubiquitous pathway in leukemogenesis. Blood. 2012;119(14):3203–3210. doi: 10.1182/blood-2011-12-399774 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Saez B, Walter MJ, Graubert TA. Splicing factor gene mutations in hematologic malignancies. Blood. 2017;129(10):1260–1269. doi: 10.1182/blood-2016-10-692400 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Yoshida K, Sanada M, Shiraishi Y, et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature. 2011;478(7367):64–69. doi: 10.1038/nature10496 [DOI] [PubMed] [Google Scholar]
  • 27.Quesada V, Conde L, Villamor N, et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nature Genetics. 2012;44(1):47. doi: 10.1038/ng.1032 [DOI] [PubMed] [Google Scholar]
  • 28.Malcovati L, Karimi M, Papaemmanuil E, et al. SF3B1 mutation identifies a distinct subset of myelodysplastic syndrome with ring sideroblasts. Blood. 2015;126(2):233–241. doi: 10.1182/blood-2015-03-633537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Papaemmanuil E, Cazzola M, Boultwood J, et al. Somatic SF3B1 Mutation in Myelodysplasia with Ring Sideroblasts. New England Journal of Medicine. 2011;365(15): 1384–1395. doi: 10.1056/NEJMoa1103283 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Meggendorfer M, Roller A, Haferlach T, et al. SRSF2 mutations in 275 cases with chronic myelomonocytic leukemia (CMML). Blood. 2012;120(15):3080–3088. doi: 10.1182/blood-2012-01-404863 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Seiler M, Peng S, Agrawal AA, et al. Somatic Mutational Landscape of Splicing Factor Genes and Their Functional Consequences across 33 Cancer Types. Cell Reports. 2018;23(1):282–296.e4. doi: 10.1016/j.celrep.2018.01.088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Scott LM, Rebel VI. Acquired Mutations That Affect Pre-mRNA Splicing in Hematologic Malignancies and Solid Tumors. J Natl Cancer Inst. 2013; 105(20):1540–1549. doi: 10.1093/jnci/djt257 [DOI] [PubMed] [Google Scholar]
  • 33.Bejar R Splicing Factor Mutations in Cancer. Adv Exp Med Biol. 2016;907:215–228. doi: 10.1007/978-3-319-29073-7_9_9 [DOI] [PubMed] [Google Scholar]
  • 34.Martin M, Maßhöfer L, Temming P, et al. Exome sequencing identifies recurrent somatic mutations in EIF1AX and SF3B1 in uveal melanoma with disomy 3. Nature Genetics. 2013;45(8):933. doi: 10.1038/ng.2674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Papaemmanuil E, Gerstung M, Malcovati L, et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013; 122(22):3616–3627. doi: 10.1182/blood-2013-08-518886 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lee SC-W, Abdel-Wahab O. Therapeutic targeting of splicing in cancer. Nat Med. 2016;22(9):976–986. doi: 10.1038/nm.4165 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Pellagatti A, Armstrong RN, Steeples V, et al. Impact of spliceosome mutations on RNA splicing in myelodysplasia: dysregulated genes/pathways and clinical associations. Blood. 2018;132(12):1225–1240. doi: 10.1182/blood-2018-04-843771 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Yoshimi A, Abdel-Wahab O. Molecular Pathways: Understanding and Targeting Mutant Spliceosomal Proteins. Clin Cancer Res. 2017;23(2):336–341. doi: 10.1158/1078-0432.CCR-16-0131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Shiozawa Y, Malcovati L, Galli A, et al. Aberrant splicing and defective mRNA production induced by somatic spliceosome mutations in myelodysplasia. Nature Communications. 2018;9(1):3649. doi: 10.1038/s41467-018-06063-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Alsafadi S, Houy A, Battistella A, et al. Cancer-associated SF3B1 mutations affect alternative splicing by promoting alternative branchpoint usage. Nat Commun. 2016;7:10615. doi: 10.1038/ncomms10615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Darman RB, Seiler M, Agrawal AA, et al. Cancer-Associated SF3B1 Hotspot Mutations Induce Cryptic 3’ Splice Site Selection through Use of a Different Branch Point. Cell Rep. 2015;13(5):1033–1045. doi: 10.1016/j.celrep.2015.09.053 [DOI] [PubMed] [Google Scholar]
  • 42.Ilagan JO, Ramakrishnan A, Hayes B, et al. U2AF1 mutations alter splice site recognition in hematological malignancies. Genome Res. 2015;25(1):14–26. doi: 10.1101/gr.181016.114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kim E, Ilagan JO, Liang Y, et al. SRSF2 Mutations Contribute to Myelodysplasia by Mutant-Specific Effects on Exon Recognition. Cancer Cell. 2015;27(5):617–630. doi: 10.1016/j.ccell.2015.04.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Schor IE, Gómez Acuña LI, Kornblihtt AR. Coupling between transcription and alternative splicing. Cancer Treat Res. 2013;158:1–24. doi: 10.1007/978-3-642-31659-3_1 [DOI] [PubMed] [Google Scholar]
  • 45.Montes M, Becerra S, Sánchez-Álvarez M, Suñé C. Functional coupling of transcription and splicing. Gene. 2012;501(2):104–117. doi: 10.1016/j.gene.2012.04.006 [DOI] [PubMed] [Google Scholar]
  • 46.Bentley DL. Coupling mRNA processing with transcription in time and space. Nature Reviews Genetics. 2014;15(3):163–175. doi: 10.1038/nrg3662 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hamperl S, Cimprich KA. The contribution of co-transcriptional RNA:DNA hybrid structures to DNA damage and genome instability. DNA Repair. 2014;19:84–94. doi: 10.1016/j.dnarep.2014.03.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Crossley MP, Bocek M, Cimprich KA. R-Loops as Cellular Regulators and Genomic Threats. Molecular Cell. 2019;73(3):398–411. doi: 10.1016/j.molcel.2019.01.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Aguilera A, Gómez-González B. Genome instability: a mechanistic view of its causes and consequences. Nature Reviews Genetics. 2008;9(3):204–217. doi : 10.1038/nrg2268 [DOI] [PubMed] [Google Scholar]
  • 50.Cimprich KA, Cortez D. ATR: an essential regulator of genome integrity. Nat Rev Mol Cell Biol. 2008;9(8):616–627. doi: 10.1038/nrm2450 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 2014;28(2):241–247. doi: 10.1038/leu.2013.336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lindsley RC. Uncoding the genetic heterogeneity of myelodysplastic syndrome. Hematology. 2017;2017(1):447–452. doi: 10.1182/asheducation-2017.1.447 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Thol F, Kade S, Schlarmann C, et al. Frequency and prognostic impact of mutations in SRSF2, U2AF1, and ZRSR2 in patients with myelodysplastic syndromes. Blood. 2012;119(15):3578–3584. doi: 10.1182/blood-2011-12-399337 [DOI] [PubMed] [Google Scholar]
  • 54.Yoshida K, Ogawa S. Splicing factor mutations and cancer. Wiley Interdisciplinary Reviews: RNA. 2014;5(4):445–459. doi: 10.1002/wrna.1222 [DOI] [PubMed] [Google Scholar]
  • 55.Fei DL, Motowski H, Chatrikhi R, et al. Wild-Type U2AF1 Antagonizes the Splicing Program Characteristic of U2AF1-Mutant Tumors and Is Required for Cell Survival. PLoS Genet. 2016;12(10):e1006384. doi: 10.1371/journal.pgen.1006384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Steensma DP, Bejar R, Jaiswal S, et al. Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes. Blood. 2015;126(1):9–16. doi: 10.1182/blood-2015-03-631747 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Makishima H, Yoshizato T, Yoshida K, et al. Dynamics of clonal evolution in myelodysplastic syndromes. Nat Genet. 2017;49(2):204–212. doi: 10.1038/ng.3742 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Mossner M, Jann J-C, Wittig J, et al. Mutational hierarchies in myelodysplastic syndromes dynamically adapt and evolve upon therapy response and failure. Blood. 2016;128(9):1246–1259. doi: 10.1182/blood-2015-11-679167 [DOI] [PubMed] [Google Scholar]
  • 59.Mian SA, Rouault-Pierre K, Smith AE, et al. SF3B1 mutant MDS-initiating cells may arise from the haematopoietic stem cell compartment. Nat Commun. 2015;6:10004. doi : 10.1038/ncomms10004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wu S-J, Kuo Y-Y, Hou H-A, et al. The clinical implication of SRSF2 mutation in patients with myelodysplastic syndrome and its stability during disease evolution. Blood. 2012; 120(15):3106–3111. doi: 10.1182/blood-2012-02-412296 [DOI] [PubMed] [Google Scholar]
  • 61.Mupo A, Seiler M, Sathiaseelan V, et al. Hemopoietic-specific Sf3b1-K700E knock-in mice display the splicing defect seen in human MDS but develop anemia without ring sideroblasts. Leukemia. 2017;31(3):720–727. doi: 10.1038/leu.2016.251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lee SC-W, Dvinge H, Kim E, et al. Modulation of splicing catalysis for therapeutic targeting of leukemias with spliceosomal mutations. Nat Med. 2016;22(6):672–678. doi: 10.1038/nm.4097 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Shirai CL, White BS, Tripathi M, et al. Mutant U2AF1-expressing cells are sensitive to pharmacological modulation of the spliceosome. Nat Commun. 2017;8. doi: 10.1038/ncomms14060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Shirai CL, Ley JN, White BS, et al. Mutant U2AF1 Expression Alters Hematopoiesis and Pre-mRNA Splicing In Vivo. Cancer Cell. 2015;27(5):631–643. doi: 10.1016/j.ccell.2015.04.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Chen L, Chen J-Y, Huang Y-J, et al. The Augmented R-Loop Is a Unifying Mechanism for Myelodysplastic Syndromes Induced by High-Risk Splicing Factor Mutations. Molecular Cell. 2018;69(3):412–425.e6. doi : 10.1016/j.molcel.2017.12.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Flynn RL, Zou L. ATR: a Master Conductor of Cellular Responses to DNA Replication Stress. Trends Biochem Sci. 2011;36(3):133–140. doi: 10.1016/j.tibs.2010.09.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Nguyen HD, Leong WY, Li W, et al. Spliceosome Mutations Induce R Loop-Associated Sensitivity to ATR Inhibition in Myelodysplastic Syndromes. Cancer Res. 2018;78(18):5363–5374. doi: 10.1158/0008-5472.CAN-17-3970 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Nguyen HD, Yadav T, Giri S, Saez B, Graubert TA, Zou L. Functions of Replication Protein A as a Sensor of R Loops and a Regulator of RNaseH1. Mol Cell. 2017;65(5):832–847.e4. doi: 10.1016/j.molcel.2017.01.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Nguyen HD, Zou L, Graubert TA. Targeting R-loop-associated ATR response in myelodysplastic syndrome. Oncotarget. 2019;10(27):2581–2582. doi : 10.18632/oncotarget.26851 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Kahles A, Lehmann K-V, Toussaint NC, et al. Comprehensive Analysis of Alternative Splicing Across Tumors from 8,705 Patients. Cancer Cell. 2018;34(2):211–224.e6. doi: 10.1016/j.ccell.2018.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hoyos LE, Abdel-Wahab O. Cancer-Specific Splicing Changes and the Potential for Splicing-Derived Neoantigens. Cancer Cell. 2018;34(2):181–183. doi: 10.1016/j.ccell.2018.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hong DS, Kurzrock R, Naing A, et al. A phase I, open-label, single-arm, dose-escalation study of E7107, a precursor messenger ribonucleic acid (pre-mRNA) splicesome inhibitor administered intravenously on days 1 and 8 every 21 days to patients with solid tumors. Invest New Drugs. 2014;32(3):436–444. doi: 10.1007/s10637-013-0046-5 [DOI] [PubMed] [Google Scholar]
  • 73.Eskens FALM, Ramos FJ, Burger H, et al. Phase I Pharmacokinetic and Pharmacodynamic Study of the First-in-Class Spliceosome Inhibitor E7107 in Patients with Advanced Solid Tumors. Clin Cancer Res. 2013;19(22):6296–6304. doi: 10.1158/1078-0432.CCR-13-0485 [DOI] [PubMed] [Google Scholar]
  • 74.Seiler M, Yoshimi A, Darman R, et al. H3B-8800, an orally available small-molecule splicing modulator, induces lethality in spliceosome-mutant cancers. Nature Medicine. 2018;24(4):497–504. doi: 10.1038/nm.4493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Steensma DP, Maris MB, Yang J, et al. H3B-8800-G0001-101: A first in human phase I study of a splicing modulator in patients with advanced myeloid malignancies. JCO. 2017;35(15_suppl):TPS7075–TPS7075. doi: 10.1200/JCO.2017.35.15_suppl.TPS7075 [DOI] [Google Scholar]
  • 76.Steensma D, Klimek V, Yang J, et al. Phase I Dose Escalation Clinical Trial of H3B-8800, a Splicing Modulator, in Patients with Advanced Myeloid Malignancies. EHA Library. 2019;266651:PS1034. [Google Scholar]
  • 77.Shi Y, Joyner AS, Shadrick W, et al. Pharmacodynamic assays to facilitate preclinical and clinical development of pre-mRNA splicing modulatory drug candidates. Pharmacol Res Perspect. 2015;3(4). doi: 10.1002/prp2.158 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kashyap MK, Kumar D, Villa R, et al. Targeting the spliceosome in chronic lymphocytic leukemia with the macrolides FD-895 and pladienolide-B. Haematologica. 2015;100(7):945–954. doi: 10.3324/haematol.2014.122069 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Bezzi M, Teo SX, Muller J, et al. Regulation of constitutive and alternative splicing by PRMT5 reveals a role for Mdm4 pre-mRNA in sensing defects in the spliceosomal machinery. Genes Dev. 2013;27(17):1903–1916. doi: 10.1101/gad.219899.113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hamard P-J, Santiago GE, Liu F, et al. PRMT5 Regulates DNA Repair by Controlling the Alternative Splicing of Histone-Modifying Enzymes. Cell Reports. 2018;24(10):2643–2657. doi: 10.1016/j.celrep.2018.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Gerhart SV, Kellner WA, Thompson C, et al. Activation of the p53-MDM4 regulatory axis defines the anti-tumour response to PRMT5 inhibition through its role in regulating cellular splicing. Sci Rep. 2018;8. doi: 10.1038/s41598-018-28002-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Rasco D, Tolcher A, Siu LL, et al. Abstract CT038: A phase I, open-label, dose-escalation study to investigate the safety, pharmacokinetics, pharmacodynamics, and clinical activity of GSK3326595 in subjects with solid tumors and non-Hodgkin’s lymphoma. Cancer Res. 2017;77(13 Supplement):CT038–CT038. doi: 10.1158/1538-7445.AM2017-CT038 [DOI] [Google Scholar]
  • 83.Millar HJ, Brehmer D, Verhulst T, et al. Abstract 950: In vivo efficacy and pharmacodynamic modulation of JNJ-64619178, a selective PRMT5 inhibitor, in human lung and hematologic preclinical models. Cancer Res. 2019;79(13 Supplement):950–950. doi: 10.1158/1538-7445.SABCS18-950 [DOI] [Google Scholar]
  • 84.Garcia-Manero G, Daver NG, Montalban-Bravo G, et al. A Phase II Study Evaluating the Combination of Nivolumab (Nivo) or Ipilimumab (Ipi) with Azacitidine in Pts with Previously Treated or Untreated Myelodysplastic Syndromes (MDS). Blood. 2016;128(22):344–344. [Google Scholar]
  • 85.Gerds AT, Scott BL, Greenberg PL, et al. PD-L1 Blockade with Atezolizumab in Higher-Risk Myelodysplastic Syndrome: An Initial Safety and Efficacy Analysis. Blood. 2018;132(Suppl 1):466–466. doi: 10.1182/blood-2018-99-118577 [DOI] [Google Scholar]
  • 86.Dillon MT, Boylan Z, Smith D, et al. PATRIOT: A phase I study to assess the tolerability, safety and biological effects of a specific ataxia telangiectasia and Rad3-related (ATR) inhibitor (AZD6738) as a single agent and in combination with palliative radiation therapy in patients with solid tumours. Clin Transl Radiat Oncol. 2018;12:16–20. doi: 10.1016/j.ctro.2018.06.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Foote KM, Nissink JWM, McGuire T, et al. Discovery and Characterization of AZD6738, a Potent Inhibitor of Ataxia Telangiectasia Mutated and Rad3 Related (ATR) Kinase with Application as an Anticancer Agent. J Med Chem. 2018;61 (22) :9889–9907. doi: 10.1021/acs.jmedchem.8b01187 [DOI] [PubMed] [Google Scholar]
  • 88.Lloyd R, Falenta K, Wijnhoven PW, et al. Abstract 337: The PARP inhibitor olaparib is synergistic with the ATR inhibitor AZD6738 in ATM deficient cancer cells. Cancer Res. 2018;78(13 Supplement):337–337. doi: 10.1158/1538-7445.AM2018-337 [DOI] [Google Scholar]

RESOURCES