Abstract
Myelodysplastic syndrome (MDS) is a group of hematopoietic disorders with limited treatment options. Anemia is a common symptom in MDS, and although erythropoiesis-stimulating agents such as erythropoietin, lenalidomide, and luspatercept are available to treat anemia, many MDS patients do not respond to these first-line therapies. Therefore, alternative drug development strategies are needed to improve therapeutic efficacy. Splicing modulators to correct splicing-related defects have shown promising results in clinical trials. Targeting differentiation of early erythroid progenitors to increase the erythroid output in MDS is another novel approach, which has shown encouraging results at the pre-clinical stage. Together, these therapeutic strategies provide new avenues to target MDS symptoms untreatable previously.
Keywords: Myelodysplastic syndrome, erythropoiesis-stimulating agents, splicing modulation, burst forming unit-erythroid, cholinergic receptor muscarinic 4, hematopoietic arc
MDS: prevalence and classification
MDS is characterized by multilineage cytopenias arising from ineffective hematopoiesis, increased apoptosis of diseased cells, and disruption of hematopoietic cell differentiation (Figure 1A) [1–4]. More than 10000 new cases of MDS are reported every year in the United States and close to 87000 worldwide. There are major clinical consequences associated with MDS that lead to morbidities, mainly caused by cytopenias and the potential for MDS to evolve into acute myeloid leukemia (AML) [5–7]. Progression to AML is mostly associated with high-risk MDS (HR-MDS) which accounts for 30% of the total MDS patients with survival <2 years [1, 8]. The early MDS initiating mutations at stem cell level together with secondary acquired mutations associated with AML give rise to leukemic stem cells that cause the transformation of MDS to AML [5]. As ~75% of people affected by MDS are >60 years old, these patients cannot tolerate intensive therapeutic approaches such as chemotherapies or benefit from allogeneic hematopoietic stem cell transplantation (allo-HSCT) (see Glossary), and often these treatments are too harsh for elderly patients [9–11]. Therefore, treatments for MDS are risk-adapted, involving the definition of different therapy goals according to the patient’s risk status [9, 12].
The International Prognostic Scoring System (IPSS) has been an essential standard for assessing the prognosis of MDS patients, along with a comprehensive diagnostic workup and testing for molecular abnormalities [13]. A revised version of IPSS (IPSS-R) is being used in clinics nowadays. IPSS-R uses criteria including the percentage of abnormal blasts in bone marrow, cytogenetics and detailed blood cell parameters to calculate risk scores which provide important prognostic and predictive information for a subsequent response to a given therapy [8]. Patients are classified based on IPSS-R risk score into low-risk MDS (LR-MDS) and high-risk MDS (HR-MDS) populations. LR-MDS patients exhibit anemia-like symptoms; therefore, the first lines of treatment are erythropoiesis-stimulating agents (ESAs) and red blood cell (RBC) transfusion. HR-MDS patients exhibit AML-like symptoms and are offered more intensive treatments, including chemotherapy with hypomethylating agents (HMAs) and allo-HSCT [14].
Constantly evolving and advancing technologies, particularly bulk and single-cell sequencing methodologies, have improved our understanding of MDS by pinpointing somatic mutations and their roles in the pathogenesis of the disease [15–18]. Several studies examining large numbers of MDS samples have identified ~40 recurrently mutated genes in various categories with almost all of the patients, including those with normal cytogenetics [19], harboring at least one mutation [19, 20]. The most frequently mutated genes are splicing factors, epigenetic regulators, and transcription factors [15, 17, 18, 21–28]. This mutational information enables clinicians potentially to tailor a therapeutic regimen for each patient, ultimately giving a better outcome. This review specifically addresses LR-MDS treatments. We present currently available options, and discuss newly advancing therapeutic strategies for this patient group.
Current LR-MDS treatments
Erythropoiesis-stimulating agents (ESAs)
ESAs work through the erythropoietin (EPO) receptor (EpoR), a cell surface receptor that belongs to the cytokine receptor family [29, 30]. Structural studies of this receptor have revealed that it lacks a cytoplasmic kinase domain and, relies on Janus kinase 2 (JAK2) protein tyrosine kinase to exert Epo-responsive signal transduction [31, 32]. Upon activation of EpoR, JAK2 tyrosine kinase residues are phosphorylated and serve as docking sites for intracellular signaling molecules. Phosphorylated JAK2 directly interacts with signal transducer and activation of transcription 5 (STAT5), which serves as a scaffold to induce EpoR signal transduction. STAT5A and STAT5B are predominant signal transducers of EpoR which upon activation, accumulate in the nucleus to induce the transcription of EpoR responsive genes (Figure 1B) [33, 34].
Treatment for LR-MDS patients without RBC transfusion-dependence (less than 2 RBC units per month) often starts with ESAs, such as recombinant EPO or darbepoetin (DAR, which is re-engineered from EPO to increase molecular mass for greater half-life and potency), as the standard of care [35–37]. Of note, MDS patients differ in their serum EPO levels and that determines the response to ESA treatment. Patients with <200 IU/L EPO level are more likely to respond whereas patients with >500 IU/L serum EPO level do not qualify to receive ESA treatment [38, 39]. ESA has been a first-line option in the LR-MDS patients with symptomatic anemia or low RBC transfusion burden. However, most MDS patients already have excessive EPO levels, which correlates with a lack of response to ESAs.
Patients who are non-responsive to ESAs and have <10 g/dL hemoglobin (Hb) concentration are recommended for RBC transfusion. The dose of RBCs and frequency of transfusion is determined by the severity of anemia and varies case by case [40]. In some cases, management of anemia together with RBC transfusion and ESAs helps patients to achieve transfusion independence (TI) [36, 41]. However, frequent RBC transfusions come with their own set of problems. Notable among these, is iron overload toxicity, which arises as excess iron from each transfusion accumulates in vital organs. Without effective iron clearance, there is a high risk of multiple vital organ failures [42, 43]. Iron chelation therapy (ICT) is often used to treat iron overload in LR-MDS patients but has received mixed response in clinics. The primary reason is that the majority of the MDS patients are elderly with other complications, which confounds the safety and tolerance of this therapy. However, there are published research studies and a clinical trial (NCT00940602i) which suggest that ICT can improve event free and overall survival of low-risk MDS patients and it should be seriously considered for these patients [44, 45].
Lenalidomide
A subset of LR-MDS patients harbor a deletion (Del (5q)) in the long (q) arm of chromosome 5. The commonly deleted region (CDR) known as 5q31 and 5q33 [46], is the location of many MDS-related genes including a core component gene of the 40S ribosomal subunit, ribosomal protein S14 (RPS14) [46–50], cell division cycle 25C (CDC25C) [49, 50], early growth response 1 (EGR1), and several critical micro-RNAs (miRNA) associated with elevated innate immune signaling such as miR-145 and miR-146a [51]. Del (5q) MDS is characterized by distinct cytomorphological abnormalities [52]. It shows relatively high sensitivity to lenalidomide, which exerts its effects through multiple mechanisms. Lenalidomide inhibits the proliferation of MDS cells by targeting cell division cycle 25C (CDC25C) and protein phosphatase 2A (PP2A), which in turn leads to G2/M cell cycle arrest and apoptosis [53, 54]. It has also been reported to bind to cereblon (CRBN), an adapter for the E3 ubiquitin ligase CRL4CRBN; modulation of the activity of CRL4CRBN leads to the degradation of disease-specific substrates. An increase in ubiquitination of casein kinase1 alpha (CK1α) and ikaros family zinc finger1 (IKZF1) followed by proteasome-mediated degradation leads to an increase in apoptosis in MDS cells with Del (5q) (Figure 1B) [55, 56].
Del (5q) LR-MDS patients often present with high EPO levels and become RBC transfusion-dependent in their disease course [57]. In multiple clinical trials, lenalidomide treatment has led to hematological improvements (HI) and even transfusion independence in Del (5q) MDS patients [41, 58, 59]. In a phase-3 trial (NCT00179621ii), lenalidomide treatment led to TI for ≥26 weeks in 56.1% of the Del (5q) MDS patients and helped to achieve complete cytogenetic response, clearance of chromosomal abnormalities, in 50% of the patients. Moreover, lenalidomide treatment improved overall survival (56.5%) and lowered the risk of AML transformation (25.1%).
Novel LR-MDS therapeutic agents and strategies
Activin receptor IIB ligand trap - Luspatercept
Luspatercept is a recombinant fusion protein which acts via the transforming growth factor-beta (TGF-β)/SMAD signaling pathway. TGF-β signaling has a well-established role in erythropoiesis and hematopoietic stem cell function and survival [60], and it has a growth inhibitory effect on erythropoiesis at the erythroblast stage [61]. Luspatercept contains a modified extracellular domain of the human activin receptor type IIB (ActRIIB) linked to a human immunoglobulin G1 (IgG1) [61]. In many pre-clinical studies, it has been shown to bind specifically to endogenous transforming growth factor (TGF)-β superfamily ligands [62, 63]. Upon binding it functions as a trap for TGF-β ligands which causes inhibition of abnormally elevated SMAD2/3 signaling (Figure 1B). This leads to an increase in erythroid cell maturation by promoting differentiation of erythroblasts [61].
RBC transfusion-dependent patients, who do not have 5q deletion and have an EPO level >200 IU/L, are less likely to respond to recombinant EPO treatment and become RBC transfusion-dependent [64, 65]. In those cases, luspatercept has been shown to increase the differentiation of erythroblasts to boost erythroid output [66]. In a phase-3 clinical trial (NCT02631070iii), luspatercept has shown hematological improvement and RBC transfusion independence in 63% of patients who otherwise require 2 units of RBCs every two weeks, although its effect on MDS patients with high EPO levels is limited [62].
Splicing mutations and modulators
Splicing factor mutations are prevalent in MDS patients [67]. Serine/arginine-rich splicing factor 2 (SRSF2), splicing factor 3b subunit 1 (SF3B1), U2 small nuclear RNA auxiliary factor 1 (U2AF1) and zinc finger CCCH-type RNA binding motif and serine/arginine-rich 2 (ZRSR2) are the most frequently mutated splicing factors, and these occur in a mutually exclusive manner [21]. SF3B1, SRSF2, and U2AF1 are subject to heterozygous, change-of-function [68–70] missense mutations affecting specific residues [16, 19, 21]. In contrast, the X chromosome-encoded ZRSR2 is enriched in nonsense and frameshift mutations mainly in male patients, consistent with loss of function [16, 19, 21]. Studies in MDS mouse models have shown that point mutations in splicing factors blocked hematopoiesis and induced apoptosis of hematopoietic stem and progenitor cells (HSPCs) [27, 68, 71, 72]. Each point mutation has a distinct effect on gene expression and splicing. While SRSF2 generally affects cassette exon recognition and SF3B1 alters 3′ splice site recognition, U2AF1 mutation causes exon inclusion and alternative 3′ splice site recognition [68, 71]. Although each mutant protein targets different pre-mRNA splicing events, they produce convergent adverse effects on hematopoiesis, for example, via indirect activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) pathway [72]. On the other hand, impaired minor (U12) intron excision via ZRSR2 loss enhances hematopoietic stem cell self-renewal [73].
SRSF2 is involved in splicing of pre-mRNAs; SRSF2P95H mutation alters SRSF2 recognition of its normal exonic splicing enhancer (ESE) and causes mis-splicing of key hematopoietic regulator genes. SRSF2WT/WT normally binds to CCNG or GGNG consensus sequences with similar affinity, whereas SRSF2P95H/WT mutation leads to an increase in CCNG binding, which leads to degradation of many essential hematopoietic regulator genes (Figure 2A) such as enhancer of zeste homolog 2 (EZH2) [68]. SF3B1K700E mutation causes excessive usage of an alternative 3′ splice site and aberrant splicing events; ~50% of murine and human aberrantly spliced genes undergo nonsense-mediated decay (NMD). Mis-splicing and NMD of mitogen-activated protein kinase kinase kinase 7 (MAP3K7) due to SF3B1K700E mutation leads to hyperactivation of NF-kβ signaling (Figure 2B) [71]. SF3B1K700E mutations are also commonly found in various cancers. Bromodomain-containing protein 9 (BRD9) - a non-canonical BAF complex subunit is highly mis-spliced and degraded in tumors and restoring BRD9 expression reduces tumor growth in vivo [74]. These tumors can also be selectively targeted by splicing modulators. MDS patients carry U2AF1 splicing factor mutations at S34 and Q157 positions. These mutations lead to exon inclusion splicing events and give rise to alternatively spliced versions of many inflammatory and immunomodulatory genes. MDS patients with U2AF1 mutations express a longer version of interleukin-1 receptor-associated kinase 4 (IRAK4-L) gene. IRAK4-L expression gets assembled with myddosome, a receptor-proximal complex which controls innate immune signal transduction, and hyperactivates NF-κβ signaling [75] (Figure 2C). Although the critical targets of ZRSR2 mutation have not been fully elucidated, leucine zipper like transcription regulator 1 (LZTR1), a regulator of Ras-related guanosine triphosphate hydrolyzing enzymes (GTPases), was identified as NMD events of minor intron-containing mRNAs in ZRSR2-mutated MDS cells [73, 76–78].
Following the discovery that natural products and their derivatives, such as pladienolides, FR901464, and spliceostatin A, physically bind to the SF3B complex and inhibit pre-mRNA splicing at an early step in spliceosome assembly [79, 80], pharmacological modulation of splicing has been developed as a potential therapeutic strategy for MDS patients [79–81]. E7107, a pladienolide derivative, was developed as an anti-tumor agent to bind to the SF3B complex to inhibit pre-mRNA splicing [80, 82]. The main challenge is to make these modulators more specific to spliceosome mutant cells and more bioavailable. In recent years, a novel medicinal chemistry approach was successfully used to synthesize small molecules to target mutant spliceosomal machinery, resulting in the discovery of the small chemical compound H3B-8800 (an orally bioavailable analogue of E7107) [83]. H3B-8800 binds to both wild-type and mutant spliceosomes but preferentially kills spliceosomal mutant cells. Sequencing data has shown that treatment with H3B-8800 retains short GC-rich introns which codes for important spliceosomal components. Moreover, spliceosomal mutant cells heavily rely on remaining wild-type spliceosome components and are more prone to the effects of splicing perturbation, providing an important mechanism for specificity towards spliceosomal mutant cells [72, 83]. A Phase 1 clinical trial (NCT02841540iv) of H3B-8800 completed successfully in 2019 (see Clinician’s Corner). In the clinic, H3B-8800 has exhibited good spliceosomal component binding in a dose-dependent manner, and it has been safe even with prolonged dosing [84]. Ongoing clinical trials will further verify its therapeutic efficacy.
Early erythroid progenitor and CHRM4 antagonist
HSPCs are capable of undergoing differentiation to generate mature hematopoietic cells. In the erythroid lineage, the burst-forming unit erythroid (BFU-E) is the first lineage determinant progenitor cell. BFU-E possesses enormous capacity for differentiation, which can generate thousands of erythrocytes. BFU-E differentiates into a late erythroid progenitor, colony-forming unit erythroid (CFU-E) [85]; CFU-E further differentiates into erythroblasts, which can form mature RBCs [61]. CFU-E expresses EpoR and EPO regulates its survival and differentiation [86]. However, specific regulators of BFU-E differentiation are unclear. Studies with human MDS patients have uncovered significantly lower numbers of BFU-E in bone marrow, compared to healthy individuals [87, 88]. These studies have also reported increased EPO levels in MDS patients due to anemic stress. Similarly, low levels of BFU-E, high levels of EPO and anemia symptoms have been reported in splicing factor mutant MDS mouse models [68, 71]. Together, these findings suggest that insufficient BFU-Es generate significantly fewer CFU-Es, which causes the unresponsiveness of MDS patients to EPO, and ultimately reduced RBC production and anemia. Therefore, targeting BFU-E differentiation could be a beneficial and novel approach in treating MDS.
Recently, the G protein-coupled receptor (GPCR), cholinergic receptor muscarinic 4 (CHRM4) was found to be expressed on early erythroid progenitors and to regulate BFU-E differentiation [89]. Blocking CHRM4 results in activation of cyclic adenosine monophosphate (cAMP) and increased cAMP response element-binding protein (CREB) transcription factor activity. CREB regulates many vital genes responsible for transient maintenance of BFU-E progenitor status to allow more RBCs to be generated from each BFU-E (Figure 3). Both genetic and pharmacological inhibition of CHRM4 with selective antagonists resulted in increased erythrocyte production and corrected anemias in mouse models of MDS, aging, and hemolysis [89]. Importantly, pharmacological inhibition of CHRM4 also rescued BFU-E and reduced abnormal EPO to levels comparable to wild-type mice, indicating that pharmacological inhibition of CHRM4 overcame EPO resistance in MDS [89]. These promising pre-clinical efficacies encourage the ongoing downstream pharmacological development of this novel therapy into clinical trials for MDS.
Hematopoietic organs including spleen and bone marrow are highly innervated by cholinergic nerves and nerve terminals which relay stimuli by means of neurotransmitters [90–92]. The identification of a functionally important neurotransmitter receptor CHRM4 involved in regulating BFU-E differentiation is the first example of how the nervous system can directly modulate HSPC differentiation and self-renewal [89]. We coined the term “Hematopoietic Arc (HematopoArc)” for this novel means of regulating hematopoiesis [89]. Further research dissecting each component of this regulation will provide important mechanistic insights.
Concluding remarks
Anemia and anemia-related symptoms are the first signs of MDS, and therefore, treatments with ESAs are the first choice of therapeutics. Conventionally, recombinant erythropoietin and its analogs are used. However, LR-MDS patients often have high endogenous EPO and, thus, they do not qualify to receive these ESAs. In these cases, the recent FDA approved drugs luspatercept and lenalidomide, if patients have Del (5q), could be the drug of choice. Although these agents drive the differentiation of erythroid cells and produce more mature RBCs to boost erythroid output, 80% of patients do not respond due to high EPO level [93]. This has prompted the development of alternative therapeutic strategies (see Outstanding Questions). As with previous developments, these are underpinned by improved mechanistic understanding of MDS and its pathogenesis.
Localizing MDS erythroid differentiation blockage to the erythroblast stage steered development of luspatercept. Similarly, understanding the significance of splicing factors in MDS pathogenesis has driven the development of new therapeutics. Mutually exclusive heterozygous splicing factor point mutations are a frequent class of mutation in MDS patients. Each mutation has a distinct effect on splicing, but they could converge to produce adverse effects on hematopoiesis. Identifying and characterizing these mutant splicing factors enlightened the invention of various splicing modulators, such as pladienolides, FR901464, spliceostatin A, and mostly recently H3B-8800 which can selectively target spliceosomal mutant cells. Clinical evaluations of the specificity and efficacy of these modulators are ongoing and will be vital for LR-MDS treatment in the future.
Focusing on the causal relationship between BFU-E insufficiency and EPO resistance has led to the discovery of CHRM4 antagonists as potential agents to correct anemia and overcome EPO resistance in MDS. A number of clinical studies have reported that MDS patients exhibit significantly reduced levels of early erythroid progenitors or BFU-Es. As a consequence, erythropoiesis is compromised, and therapeutic benefit of conventional treatments is limited. A recent study has shown that CHRM4 is a regulator of BFU-E differentiation; inhibition of CHRM4 was found to drive BFU-E differentiation and increase the erythroid output in pre-clinical models. Efforts to gain a deeper understanding of CHRM4 regulation of erythropoiesis and clinical development of CHRM4 inhibitors are ongoing and potentially will open a new chapter in LR-MDS therapeutics.
Clinician’s Corner.
A phase 1 multicenter clinical trial (NCT02841540iv) of the splicing modulator H3B-8800 was completed recently and it was shown to be well-tolerated with escalated doses for prolonged time. H3B-8800 has also exhibited dose-dependent target engagement in MDS patients. 42% of MDS and other myeloid neoplasm transfusion dependent patients did not require RBC transfusion for ⩾8 weeks while participating in the study.
Antisense oligonucleotide (ASO)-based treatments have been approved by the FDA recently for splicing related disorders such as spinal muscular atrophy and Duchenne muscular dystrophy. An ASO targeting STAT3 has been in phase 2 clinical trial (NCT02983578v) for lung and colorectal cancers and also, it has shown efficacy in pre-clinical models of MDS [94].
ESAs, such as erythropoietin and darbepoetin, are the first choice of drugs for LR-MDS patients. LR-MDS patients having endogenous EPO level <200 IU/L show beneficial responses to ESA treatment while patients with >500 IU/L EPO cannot receive ESAs and become RBC transfusion dependent.
Enhancing the differentiation of BFU-Es with CHRM4 inhibitors provides a means of overcoming the limitation of ESAs by boosting erythropoiesis at an earlier stage in the process and independently of EPO. Pharmacological inhibition of CHRM4 has exhibited promising efficacy in pre-clinical models of MDS, correcting anemic phenotype, early erythroid progenitor insufficiency, abnormally elevated EPO level, and extending survival of splicing factor mutant MDS models. Overall, treatment with CHRM4 inhibitor was well tolerated and exhibited no side-effects on kidney and liver function [89]. Future development of CHRM4 inhibitors has the potential to target the vast majority of MDS patients.
Outstanding Questions.
The splicing factor mutant cells rely heavily on remaining wild-type splicing activity. Can new and improved methods of splicing perturbation be developed?
Are other GPCRs, besides CHRM4, functionally important in erythropoiesis?
Can dysregulated GPCR signaling inform prognosis and efficacy of GPCR modulators?
As a receptor for neurotransmitter acetylcholine (ACh), CHRM4 is involved in cholinergic signaling in the peripheral and central nervous system. What is the precise physiological and molecular mechanism of neural regulation of BFU-E differentiation?
Various biological consequences converge to produce adverse effects on erythropoiesis in MDS. Ultimately will combinatorial regimens be effective in treating MDS?
Highlights.
MDS is characterized by ineffective hematopoiesis arising from mutations in hematopoietic stem cells.
Erythropoiesis stimulating agents are the first choice of drugs for low risk MDS patients but are often ineffective.
Splicing factor mutations are the major class of mutation in MDS and recently developed splicing modulators can specifically target these mutations.
Enhancing differentiation of early erythroid progenitors via inhibition of CHRM4, a newly identified negative regulator of BFU-E differentiation, has shown promising results in pre-clinical testing.
Detailed investigation of nervous system regulation of HSPC differentiation could unravel novel regulatory mechanisms of hematopoiesis.
Acknowledgement
This work is supported by grants to L. Zhang including CSHL President’s Council, NIH/NHLBI grant (U01 HL127522), Northwell Cancer Translational Research Award, and Edward P. Evans Foundation MDS Young Investigator Award.
Glossary
- Allogeneic hematopoietic stem cell transplantation (allo-HSCT)
a clinical procedure, used to treat a variety of blood cancers, in which healthy stem cells from a donor are transferred to a patient to replace their own stem cells.
- International Prognostic Scoring System (IPSS)
a scoring system for staging MDS, which considers various risk factors, such as bone marrow blasts, chromosomal karyotype, number of cytopenias etc., for the prognosis of MDS.
- Cytogenetics
a genetic study of the number and morphology of chromosomes inside the cell.
- Erythropoietin (EPO)
a hormone produced by kidney that plays a crucial role in formation and maintenance of red blood cells.
- International Unit (IU)
an internationally accepted unit of measurement for fat soluble substances such as hormones, vitamins, vaccines and drugs. of 8.4 micrograms of EPO is equivalent to 1000 IU.
- Transfusion Independence (TI)
a state in which a patient’s body produces enough RBCs and does not rely on the exogenous supply of RBCs by transfusion.
- Splicing modulators
small molecules that can selectively target and kill spliceosomal mutant cells, thereby inhibiting abnormal splicing.
- Exonic splicing enhancer (ESE)
RNA sequences within exons that promote splicing of pre-mRNA to mature mRNA.
- G protein-coupled receptor (GPCR)
the largest and most diverse group of transmembrane receptors which serve as the mediator between extracellular stimuli and intracellular signaling cascades.
- Cholinergic receptor muscarinic 4 (CHRM4)
a GPCR of the central and peripheral nervous system that is activated by acetylcholine to bring about a variety of downstream functions.
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.
Resources:
Conflicts of Interest
A patent on targeting early erythroid progenitor to treat myelodysplastic syndrome has been filed related to L. Zhang.
References
- 1.Ades L et al. (2014) Myelodysplastic syndromes. Lancet 383 (9936), 2239–52. [DOI] [PubMed] [Google Scholar]
- 2.Fenaux P et al. (2020) Luspatercept in Patients with Lower-Risk Myelodysplastic Syndromes. N Engl J Med 382 (2), 140–151. [DOI] [PubMed] [Google Scholar]
- 3.Cazzola M (2020) Myelodysplastic Syndromes. N Engl J Med 383 (14), 1358–1374. [DOI] [PubMed] [Google Scholar]
- 4.Pang WW et al. (2013) Hematopoietic stem cell and progenitor cell mechanisms in myelodysplastic syndromes. Proc Natl Acad Sci U S A 110 (8), 3011–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Chen J et al. (2019) Myelodysplastic syndrome progression to acute myeloid leukemia at the stem cell level. Nat Med 25 (1), 103–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Inoue D et al. (2015) SETBP1 mutations drive leukemic transformation in ASXL1-mutated MDS. Leukemia 29 (4), 847–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Saika M et al. (2018) ASXL1 and SETBP1 mutations promote leukaemogenesis by repressing TGFbeta pathway genes through histone deacetylation. Sci Rep 8 (1), 15873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Greenberg PL et al. (2012) Revised international prognostic scoring system for myelodysplastic syndromes. Blood 120 (12), 2454–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Platzbecker U (2019) Treatment of MDS. Blood 133 (10), 1096–1107. [DOI] [PubMed] [Google Scholar]
- 10.Bewersdorf JP and Zeidan AM (2020) Evolving therapies for lower-risk myelodysplastic syndromes. Ann Hematol 99 (4), 677–692. [DOI] [PubMed] [Google Scholar]
- 11.Greenberg PL et al. (2017) Myelodysplastic Syndromes, Version 2.2017, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 15 (1), 60–87. [DOI] [PubMed] [Google Scholar]
- 12.Malcovati L et al. (2013) Diagnosis and treatment of primary myelodysplastic syndromes in adults: recommendations from the European LeukemiaNet. Blood 122 (17), 2943–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Greenberg P et al. (1997) International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 89 (6), 2079–88. [PubMed] [Google Scholar]
- 14.Parker JE et al. (2000) The role of apoptosis, proliferation, and the Bcl-2-related proteins in the myelodysplastic syndromes and acute myeloid leukemia secondary to MDS. Blood 96 (12), 3932–8. [PubMed] [Google Scholar]
- 15.Bejar R et al. (2011) Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med 364 (26), 2496–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Haferlach T et al. (2014) Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 28 (2), 241–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Delhommeau F et al. (2009) Mutation in TET2 in myeloid cancers. N Engl J Med 360 (22), 2289–301. [DOI] [PubMed] [Google Scholar]
- 18.Walter MJ et al. (2011) Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia 25 (7), 1153–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Papaemmanuil E et al. (2013) Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 122 (22), 3616–27; quiz 3699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Malcovati L et al. (2017) Clinical significance of somatic mutation in unexplained blood cytopenia. Blood 129 (25), 3371–3378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yoshida K et al. (2011) Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 478 (7367), 64–9. [DOI] [PubMed] [Google Scholar]
- 22.Graubert TA et al. (2011) Recurrent mutations in the U2AF1 splicing factor in myelodysplastic syndromes. Nat Genet 44 (1), 53–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Langemeijer SM et al. (2009) Acquired mutations in TET2 are common in myelodysplastic syndromes. Nat Genet 41 (7), 838–42. [DOI] [PubMed] [Google Scholar]
- 24.Ernst T et al. (2010) Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet 42 (8), 722–6. [DOI] [PubMed] [Google Scholar]
- 25.Abdel-Wahab O et al. (2011) Concomitant analysis of EZH2 and ASXL1 mutations in myelofibrosis, chronic myelomonocytic leukemia and blast-phase myeloproliferative neoplasms. Leukemia 25 (7), 1200–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bernard E et al. (2020) Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes. Nat Med 26 (10), 1549–1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Abdel-Wahab O et al. (2013) Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J Exp Med 210 (12), 2641–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Inoue D et al. (2013) Myelodysplastic syndromes are induced by histone methylation-altering ASXL1 mutations. J Clin Invest 123 (11), 4627–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Miura O et al. (1993) Inactivation of erythropoietin receptor function by point mutations in a region having homology with other cytokine receptors. Mol Cell Biol 13 (3), 1788–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.D’Andrea AD et al. (1989) Expression cloning of the murine erythropoietin receptor. Cell 57 (2), 277–85. [DOI] [PubMed] [Google Scholar]
- 31.Miura O et al. (1991) Induction of tyrosine phosphorylation by the erythropoietin receptor correlates with mitogenesis. Mol Cell Biol 11 (10), 4895–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Yoshimura A and Lodish HF (1992) In vitro phosphorylation of the erythropoietin receptor and an associated protein, pp130. Mol Cell Biol 12 (2), 706–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Klingmuller U et al. (1996) Multiple tyrosine residues in the cytosolic domain of the erythropoietin receptor promote activation of STAT5. Proc Natl Acad Sci U S A 93 (16), 8324–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fujitani Y et al. (1997) An alternative pathway for STAT activation that is mediated by the direct interaction between JAK and STAT. Oncogene 14 (7), 751–61. [DOI] [PubMed] [Google Scholar]
- 35.Fenaux P et al. (2018) A phase 3 randomized, placebo-controlled study assessing the efficacy and safety of epoetin-alpha in anemic patients with low-risk MDS. Leukemia 32 (12), 2648–2658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Platzbecker U et al. (2017) A phase 3 randomized placebo-controlled trial of darbepoetin alfa in patients with anemia and lower-risk myelodysplastic syndromes. Leukemia 31 (9), 1944–1950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Smith RE Jr. et al. (2001) Novel erythropoiesis stimulating protein (NESP) for the treatment of anaemia of chronic disease associated with cancer. Br J Cancer 84 Suppl 1, 24–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Buckstein R et al. (2017) ITACA: A new validated international erythropoietic stimulating agent-response score that further refines the predictive power of previous scoring systems. Am J Hematol 92 (10), 1037–1046. [DOI] [PubMed] [Google Scholar]
- 39.Hellstrom-Lindberg E et al. (2003) A validated decision model for treating the anaemia of myelodysplastic syndromes with erythropoietin + granulocyte colony-stimulating factor: significant effects on quality of life. Br J Haematol 120 (6), 1037–46. [DOI] [PubMed] [Google Scholar]
- 40.de Swart L et al. (2020) Impact of red blood cell transfusion dose density on progression-free survival in patients with lower-risk myelodysplastic syndromes. Haematologica 105 (3), 632–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.List A et al. (2005) Efficacy of lenalidomide in myelodysplastic syndromes. N Engl J Med 352 (6), 549–57. [DOI] [PubMed] [Google Scholar]
- 42.Malcovati L et al. (2011) Impact of the degree of anemia on the outcome of patients with myelodysplastic syndrome and its integration into the WHO classification-based Prognostic Scoring System (WPSS). Haematologica 96 (10), 1433–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Roy NB et al. (2011) Cardiac iron overload in transfusion-dependent patients with myelodysplastic syndromes. Br J Haematol 154 (4), 521–4. [DOI] [PubMed] [Google Scholar]
- 44.Angelucci E et al. (2020) Iron Chelation in Transfusion-Dependent Patients With Low- to Intermediate-1-Risk Myelodysplastic Syndromes. Ann Intern Med 173 (7), 595–596. [DOI] [PubMed] [Google Scholar]
- 45.Hoeks M et al. (2020) Impact of treatment with iron chelation therapy in patients with lower-risk myelodysplastic syndromes participating in the European MDS registry. Haematologica 105 (3), 640–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhao N et al. (1997) Molecular delineation of the smallest commonly deleted region of chromosome 5 in malignant myeloid diseases to 1–1.5 Mb and preparation of a PAC-based physical map. Proc Natl Acad Sci U S A 94 (13), 6948–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ebert BL et al. (2008) Identification of RPS14 as a 5q- syndrome gene by RNA interference screen. Nature 451 (7176), 335–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Valencia A et al. (2008) Lack of RPS14 promoter aberrant methylation supports the haploinsufficiency model for the 5q- syndrome. Blood 112 (3), 918. [DOI] [PubMed] [Google Scholar]
- 49.Horrigan SK et al. (2000) Delineation of a minimal interval and identification of 9 candidates for a tumor suppressor gene in malignant myeloid disorders on 5q31. Blood 95 (7), 2372–7. [PubMed] [Google Scholar]
- 50.Horrigan SK et al. (1996) Polymerase chain reaction-based diagnosis of del (5q) in acute myeloid leukemia and myelodysplastic syndrome identifies a minimal deletion interval. Blood 88 (7), 2665–70. [PubMed] [Google Scholar]
- 51.Starczynowski DT et al. (2010) Identification of miR-145 and miR-146a as mediators of the 5q- syndrome phenotype. Nat Med 16 (1), 49–58. [DOI] [PubMed] [Google Scholar]
- 52.Oelschlaegel U et al. (2016) Clonal architecture of del(5q) myelodysplastic syndromes: aberrant CD5 or CD7 expression within the myeloid progenitor compartment defines a subset with high clonal burden. Leukemia 30 (2), 517–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Margolis SS et al. (2006) Role for the PP2A/B56delta phosphatase in regulating 14-3-3 release from Cdc25 to control mitosis. Cell 127 (4), 759–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wei S et al. (2009) A critical role for phosphatase haplodeficiency in the selective suppression of deletion 5q MDS by lenalidomide. Proc Natl Acad Sci U S A 106 (31), 12974–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Gandhi AK et al. (2014) Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN.). Br J Haematol 164 (6), 811–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kronke J et al. (2015) Lenalidomide induces ubiquitination and degradation of CK1alpha in del(5q) MDS. Nature 523 (7559), 183–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Giagounidis AA et al. (2004) Clinical, morphological, cytogenetic, and prognostic features of patients with myelodysplastic syndromes and del(5q) including band q31. Leukemia 18 (1), 113–9. [DOI] [PubMed] [Google Scholar]
- 58.List A et al. (2006) Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. N Engl J Med 355 (14), 1456–65. [DOI] [PubMed] [Google Scholar]
- 59.List AF et al. (2014) Extended survival and reduced risk of AML progression in erythroid-responsive lenalidomide-treated patients with lower-risk del(5q) MDS. Leukemia 28 (5), 1033–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Muench DE et al. (2018) SKI controls MDS-associated chronic TGF-beta signaling, aberrant splicing, and stem cell fitness. Blood 132 (21), e24–e34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Suragani RN et al. (2014) Transforming growth factor-beta superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat Med 20 (4), 408–14. [DOI] [PubMed] [Google Scholar]
- 62.Platzbecker U et al. (2017) Luspatercept for the treatment of anaemia in patients with lower-risk myelodysplastic syndromes (PACE-MDS): a multicentre, open-label phase 2 dose-finding study with long-term extension study. Lancet Oncol 18 (10), 1338–1347. [DOI] [PubMed] [Google Scholar]
- 63.Mies A and Platzbecker U (2017) Increasing the effectiveness of hematopoiesis in myelodysplastic syndromes: erythropoiesis-stimulating agents and transforming growth factor-beta superfamily inhibitors. Semin Hematol 54 (3), 141–146. [DOI] [PubMed] [Google Scholar]
- 64.Park S et al. (2017) Outcome of Lower-Risk Patients With Myelodysplastic Syndromes Without 5q Deletion After Failure of Erythropoiesis-Stimulating Agents. J Clin Oncol 35 (14), 1591–1597. [DOI] [PubMed] [Google Scholar]
- 65.Park S et al. (2008) Predictive factors of response and survival in myelodysplastic syndrome treated with erythropoietin and G-CSF: the GFM experience. Blood 111 (2), 574–82. [DOI] [PubMed] [Google Scholar]
- 66.Suragani RN et al. (2014) Modified activin receptor IIB ligand trap mitigates ineffective erythropoiesis and disease complications in murine beta-thalassemia. Blood 123 (25), 3864–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Papaemmanuil E et al. (2011) Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. N Engl J Med 365 (15), 1384–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kim E et al. (2015) SRSF2 Mutations Contribute to Myelodysplasia by Mutant-Specific Effects on Exon Recognition. Cancer Cell 27 (5), 617–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Darman RB et al. (2015) Cancer-Associated SF3B1 Hotspot Mutations Induce Cryptic 3? Splice Site Selection through Use of a Different Branch Point. Cell Rep 13 (5), 1033–45. [DOI] [PubMed] [Google Scholar]
- 70.Ilagan JO et al. (2015) U2AF1 mutations alter splice site recognition in hematological malignancies. Genome Res 25 (1), 14–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Obeng EA et al. (2016) Physiologic Expression of Sf3b1(K700E) Causes Impaired Erythropoiesis, Aberrant Splicing, and Sensitivity to Therapeutic Spliceosome Modulation. Cancer Cell 30 (3), 404–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lee SC et al. (2018) Synthetic Lethal and Convergent Biological Effects of Cancer-Associated Spliceosomal Gene Mutations. Cancer Cell 34 (2), 225–241 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Inoue D et al. (2021) Minor intron retention drives clonal hematopoietic disorders and diverse cancer predisposition. Nat Genet 53 (5), 707–718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Inoue D et al. (2019) Spliceosomal disruption of the non-canonical BAF complex in cancer. Nature 574 (7778), 432–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Smith MA et al. (2019) U2AF1 mutations induce oncogenic IRAK4 isoforms and activate innate immune pathways in myeloid malignancies. Nat Cell Biol 21 (5), 640–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Bigenzahn JW et al. (2018) LZTR1 is a regulator of RAS ubiquitination and signaling. Science 362 (6419), 1171–1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Castel P et al. (2019) RIT1 oncoproteins escape LZTR1-mediated proteolysis. Science 363 (6432), 1226–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Steklov M et al. (2018) Mutations in LZTR1 drive human disease by dysregulating RAS ubiquitination. Science 362 (6419), 1177–1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kaida D et al. (2007) Spliceostatin A targets SF3b and inhibits both splicing and nuclear retention of pre-mRNA. Nat Chem Biol 3 (9), 576–83. [DOI] [PubMed] [Google Scholar]
- 80.Kotake Y et al. (2007) Splicing factor SF3b as a target of the antitumor natural product pladienolide. Nat Chem Biol 3 (9), 570–5. [DOI] [PubMed] [Google Scholar]
- 81.Fan L et al. (2011) Sudemycins, novel small molecule analogues of FR901464, induce alternative gene splicing. ACS Chem Biol 6 (6), 582–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Folco EG et al. (2011) The anti-tumor drug E7107 reveals an essential role for SF3b in remodeling U2 snRNP to expose the branch point-binding region. Genes Dev 25 (5), 440–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Seiler M et al. (2018) H3B-8800, an orally available small-molecule splicing modulator, induces lethality in spliceosome-mutant cancers. Nat Med 24 (4), 497–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Steensma DP et al. (2019) Results of a Clinical Trial of H3B-8800, a Splicing Modulator, in Patients with Myelodysplastic Syndromes (MDS), Acute Myeloid Leukemia (AML) or Chronic Myelomonocytic Leukemia (CMML). Blood 134 (Supplement_1), 673–673. [Google Scholar]
- 85.Li H et al. (2019) Rate of Progression through a Continuum of Transit-Amplifying Progenitor Cell States Regulates Blood Cell Production. Dev Cell 49 (1), 118–129 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lin CS et al. (1996) Differential effects of an erythropoietin receptor gene disruption on primitive and definitive erythropoiesis. Genes Dev 10 (2), 154–64. [DOI] [PubMed] [Google Scholar]
- 87.Nathan DG et al. (1978) Erythroid precursors in congenital hypoplastic (Diamond-Blackfan) anemia. J Clin Invest 61 (2), 489–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Claessens YE et al. (2002) In vitro proliferation and differentiation of erythroid progenitors from patients with myelodysplastic syndromes: evidence for Fas-dependent apoptosis. Blood 99 (5), 1594–601. [DOI] [PubMed] [Google Scholar]
- 89.Trivedi G et al. (2019) Muscarinic acetylcholine receptor regulates self-renewal of early erythroid progenitors. Sci Transl Med 11 (511). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Rosas-Ballina M et al. (2008) Splenic nerve is required for cholinergic antiinflammatory pathway control of TNF in endotoxemia. Proc Natl Acad Sci U S A 105 (31), 11008–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Carnevale D et al. (2016) A cholinergic-sympathetic pathway primes immunity in hypertension and mediates brain-to-spleen communication. Nat Commun 7, 13035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Maryanovich M et al. (2018) Adrenergic nerve degeneration in bone marrow drives aging of the hematopoietic stem cell niche. Nat Med 24 (6), 782–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Wallvik J et al. (2002) Serum erythropoietin (EPO) levels correlate with survival and independently predict response to EPO treatment in patients with myelodysplastic syndromes. Eur J Haematol 68 (3), 180–5. [DOI] [PubMed] [Google Scholar]
- 94.Shastri A et al. (2018) Antisense STAT3 inhibitor decreases viability of myelodysplastic and leukemic stem cells. J Clin Invest 128 (12), 5479–5488. [DOI] [PMC free article] [PubMed] [Google Scholar]