Abstract
Chronic myelomonocytic leukemia (CMML) is an aggressive clonal stem cell disorder categorized among myelodysplastic/ myeloproliferative overlap neoplasms. While sharing features with both myelodysplastic syndromes and myeloproliferative neoplasms, CMML has distinct molecular and clinical profiles. The presence of CMML-specific prognostic models, response criteria, and dedicated clinical trials underscores a unique and complex biology. Age-related changes affecting the bone marrow microenvironment, immune responses, and the intricate balance between epigenetic deregulation and proinflammatory signaling are characteristic of this disease, collectively posing significant scientific and clinical challenges in its management. CMML is an aging-related, clinically heterogeneous neoplasm with limited approved therapeutic options, representing an area of unmet medical need. This review offers a comprehensive analysis of the current understanding of the molecular mechanisms driving CMML evolution and its clinical manifestations within the ever-evolving landscape of precision medicine. In light of the most recent molecular discoveries, we highlight the shortcomings of existing therapies and underscore promising investigational agents. Many of the biological findings discussed are shared across a spectrum of acute and chronic myeloid neoplasms, as well as clonal hematopoiesis, broadening the scope of this review.
Aging and myeloid malignancies
In addition to sustained peripheral blood monocytosis and bone marrow dysplasia1 (Figure 1), CMML is associated with an inherent risk of transformation to secondary acute myeloid leukemia (AML) of about 15-20% over 3 to 5 years.2 Like patients with AML, patients with CMML have about 10-15 mutations per kilobase of coding DNA. Although this is several-fold lower than in other malignancies (melanoma ~1,000, lung cancer ~150),3,4 CMML has a very heterogeneous clinical course.4 While only A S X L1 mutations reproducibly predict inferior outcomes in CMML,5 higher levels of interleukin (IL)-10 (human cytokine synthesis inhibitory factor) have also been shown to improve prognosis,6 emphasizing a complex pathophysiology that extends beyond somatic alterations, including cytokine diversity and epigenetic deregulation.6
Among all hematologic neoplasms, CMML displays the most striking skewing towards older age with a median age at presentation of >70 years.1,2 Aging hematopoietic stem cells (HSC) steadily accumulate mutations (mean of 17 per year after birth).7 While most are inconsequential, between one in 34 and one in 12 non-synonymous mutations provide a selective advantage and drive clonal expansion by cell-intrinsic and extrinsic selection pressures.7,8 Driver mutations commonly occur in leukemia genes, and lead to clonal hematopoiesis of indeterminate potential (CHIP) when such mutations can be detected at a variant allele fraction of ≥2%, in the absence of blood count abnormalities. There is significant overlap between clonal hematopoiesis mutations and CMML driver mutations, with the majority of CMML patients showing mutations in ≥2 clonal hematopoiesis-associated genes (e.g., ASXL1, TET2, SRSF2). This suggests that in most cases, CMML develops on the background of age-related clonal hematopoiesis, with subsequent mutations shaping the CMML phenotype and AML transformation rates.9
Oxidative stress, telomere shortening, and activation of tumor suppressor genes all contribute to an abrupt reduction in the diversity of the HSC pool, such that by the age of 70 years, most peripheral blood cells are derived from only 10-20 HSC clones.7 Interestingly, only 20% of the HSC clones remaining in people aged >70 years have identifiable mutations in known driver genes.7 While CHIP is associated with an increased risk of developing hematologic malignancies,10-12 the more prevalent, presumed “driverless” clonal expansions noted in the elderly, might underlie other blood- and immune-related signs of aging.13-15 Notably, while CHIP is present in >90% of individuals ≥85 years,8 hematologic malignancies continue to be a rare entity, supporting the hypothesis that extrinsic non-cell-autonomous mechanisms are pivotal in shaping the natural history of CHIP.
Epigenetic/splicing dysregulation in chronic myelomonocytic leukemia
Somatic mosaicisms form the structural background in >90% of CMML patients.16,17 Recurrently mutated genes include epigenetic regulators (TET2 ~50% and ASXL1 ~40%), spliceosome components (SRSF2 ~50%) and cell signaling pathways (RAS ~30% and CBL ~15%)18-21 (Figure 2A). While these mutations are not exclusive to CMML and can be found across the spectrum of acute and chronic myeloid neoplasms as well as CHIP, specific mutations can define a dysplastic or a more aggressive proliferative phenotype (Figure 2B-D). In addition, studies in larger cohorts have highlighted that a pattern of TET2-SRSF2 (~46%) co-mutations,22 as well as biallelic TET2 mutations/alterations (biTET2, ~45%), are commonly identified in CMML, secondary to granulocyte-monocyte progenitor (GMP)-biased hematopoesis.23 These events occur either when a secondary TET2 variant is subclonal to an ancestral TET2 variant or following loss of heterozygosity at 4q24.23 Notably, truncating TET2 variants are more likely to occur in the context of biTET2,23 highlighting that clonal selection for a complete loss of TET2 plays an important role in the evolution of CMML. At the protein level, TET2 plays a pleiotropic role in hematopoiesis, with mutations found in both myeloid and lymphoid malignancies. TET2 initiates DNA demethylation and regulates chromatin modifications at regions critical for lineage commitment and differentiation of HSC and progenitor cells.24 In murine models, loss of Te t 2 (in particular, the catalytic domain25) induces a strong myeloid bias26 (Figure 3), with increased accessibility at enhancers of pro-myeloid differentiation genes thought to contribute to this phenotype.27 The strong myeloid bias induced by TET2 variants is also recapitulated in patients, as suggested by the observation that, at single-cell resolution, TET2 mutations mostly expand in the myeloid lineage.28
Variants in SRSF2, a component of the spliceosome machinery, are the second most common ancestral variants in CMML, and also promote a myeloid bias (Figure 3). Mutations at the hotspot proline residue 95 (P95) alter messenger RNA (mRNA) splicing by changing the RNA binding affinity of SRSF2, leading to mis-regulation of exon inclusion.29 These variants, however, lead to only modest changes in global mRNA splicing. Even if a subset of mis-spliced transcripts has been proposed to be relevant for myelodysplastic syndromes,29-31 the molecular mechanisms whereby SRSF2 variants are implicated in MDS and CMML remain elusive. Using a conditional murine model of Srsf2P95H, it was shown that Srsf2P95H/+ native chimeras showed clonal expansion at the expense of wild-type HSC only when transplanted into lethally irradiated recipients without an external competitor, suggesting that the specific characteristics of a microenvironment that is aged matched to the HSC plays a crucial role in allowing this variant to establish clonal dominance.32 In addition, Srsf2P95H/+ mice were observed to develop monocytosis and dysplastic neutrophils on aging (~12 months after induction of the recombinant allele), and eventually succumbed to a myeloproliferative disorder characterized by the presence of additional somatic mutations observed in CMML, including Ras pathway mutations.32
Among all the epigenetic drivers, truncating ASXL1 variants are associated with adverse outcomes in CMML, proliferative disease features (Figure 2B), and resistance to epigenetic therapies.5 Most ASXL1 variants are frameshift or nonsense mutations in exon 12 (last exon) resulting in truncation of the protein at the C-terminus and loss of the plant homeodomain (PHD).33 ASXL1 mutations have been shown to cause both loss of polycomb repressive complex 2 (PRC2)-mediated histone methylation (H3K27me3) at HOXA cluster genes,34 and loss of polycomb repressive complex 1 (PRC1)-mediated histone deubiquitination (H2AK119Ub).35,36
Given the complex ASXL1 interactions, our group attempted to further define the role of ASXL1 in CMML by interrogating the genome, transcriptome, and epigenome of wild-type ASXL1 versus truncating ASX L1-mutated primary CMML samples. We found that ASXL1-mutated patients gained accessibility at several enhancers enriched in ETS and BRD4 complex motifs, with upregulation in genes involved in cell cycle progression, DNA replication, and leukemogenesis.37
Signaling pathway mutations in chronic myelomonocytic leukemia
Signaling mutations are frequent in CMML and are usually associated with myeloproliferative features (so-called proliferative CMML).38 These are largely dominated by oncogenic RAS pathway mutations (>70%; NRAS, CBL, PTPN11, KRAS, NF1, BRAF) but also include JAK2V617F, FLT3 and CSF3R. The latter two are very infrequent, with the presence of an FLT3-internal tandem duplication usually heralding transformation to AML.39
RAS pathway mutations (NRAS, CBL, PTPN11, KRAS, NF1)
Mutations in the epigenetic machinery are not specific to CMML, but are generally considered ancestral to signaling pathway mutations.38,40,41 Germline and somatic RAS pathway mutations are associated with juvenile myelomonocytic leukemia, an aggressive pediatric myeloproliferative neoplasm that resembles proliferative CMML.42 Features of proliferative CMML include constitutional symptoms, extramedullary hematopoiesis and myeloproliferation. This contrasts with dysplastic CMML, which is instead characterized by cytopenia(s) and a more indolent course (Figure 2B-D). Leveraging a large cohort of >1,000 CMML patients, our group demonstrated that oncogenic RAS mutations are more prevalent in, and occur at higher variant allele fractions in proliferative CMML than in dysplastic CMML.38 Interestingly, in murine models, NrasG12D has been shown to have a bimodal effect on HSC, both increasing and decreasing the rate at which some HSC divide; besides, NrasG12D can also increase reconstitution and self-renewal potential of HSC on serial transplantation.43
In mice, during emergency and leukemic myelopoiesis, GMP aggregate in self-renewing GMP clusters. These are transcriptionally defined by the activation of an inducible Irf8 and β-catenin self-renewal network.44 Novel insights into CMML-related phenotypes come from the identification, in a subset of CMML patients carrying RAS mutations and high-risk disease features, of a GMP-like inflammatory population, transcriptionally similar to the cluster of self-renewing GMP described above45 (Figure 3). Besides its canonical RAS/MEK/ ERK oncogenic signaling (Figure 4), mutant RAS induces the generation of reactive oxygen species, which in turn, promote the activation of the NLRP3 inflammasome.46 The latter has a key role in the development of several clinical manifestations associated with KRAS-mutant myeloproliferative neoplasms.46 In response to various signals from pathogens and internal damage, activation of NLRP3 results in release of proinflammatory cytokines (e.g., IL-1β and IL-18)47 through gasdermin D-mediated permeabilization of the plasma membrane. If gasdermin D pores cannot be repaired, cells undergo pyroptosis48 with the potential to amplify inflammatory responses (Figure 3).49,50 In addition to the classical mitogen-activated protein kinase (MAPK) pathway, GTP-bound RAS also binds to p110 to activate the PI3K-AKT-mTOR signaling cascade51 (Figure 4). Thus, inhibition of the phosphoinositol-3 kinase (PI3K) pathway has been explored in multiple RAS-mutated tumor types, both as an initial strategy and in subsequent efforts to overcome resistance to RAS inhibition.52 RAS mutations might also have roles in modulating adaptive immune responses to tumor cells. Using an Asxl1–/– NrasG12D/+ Vav-Cre mouse model, we have shown that Nras and Asxl1 cooperate to accelerate progression of CMML to AML, with AML cells overexpressing all the inhibitory immune checkpoint ligands (PD-L1/PD-L2, CD155, and CD80/CD86), highlighting that a suppressive microenvironment could play an important role in transformation to secondary AML.53 We have also shown accumulation of clonal RAS mutant CD123/CD303+ plasmacytoid dendritic cells in CMML patients (Figures 1C, D and 3), and have documented their association with transformation to AML.54 Further work by our group has shown that these plasmacytoid dendritic cell clusters are positive for indoleamine 2,3-dioxygenase 1/2 (IDO1/2),55 with IDO being an immune-checkpoint enzyme that induces systemic immune tolerance through multiple mechanisms, including regulatory T-cell expansion and tryptophan catabolism56,57 (Figure 3).
JAK2V617F mutations in chronic myelomonocytic leukemia
JAK2V617F mutations are encountered in ~10% CMML patients. By studying a large cohort of CMML patients, we found that JAK2-mutated CMML is associated with higher hemoglobin/ hematocrit levels and platelet counts, and frequently co-occurs with TET2 mutations. However, we did not identify an increased risk of thrombosis, acute leukemia transformation, or impact on overall survival.58 JAK2V617F mutant CMML can at times be difficult to distinguish from JAK2-mutant myeloproliferative neoplasms with monocytosis, although the use of monocyte partitioning flow cytometry (M01/classical monocytes >94% in CMML) and focus on megakaryocytic morphology can help to resolve this dilemma in several cases.59 Irrespective of signaling pathway mutations of signaling pathway mutations, CMML patients demonstrate granulocyte-macrophage colony-stimulating factor (GM-CSF)-dependent hypersensitivity in colony formation assays and by phospho-STAT5 (pSTAT5) flow cytometry, as compared to healthy controls, an effect more pronounced in patients with RAS mutations.60 Lenzilumab (KB003) is a novel engineered human immunoglobulin G1K monoclonal antibody with high affinity for human GM-CSF (Figure 5) and has activity in preclinical models of CMML.60 In a phase I study of lenzilumab in CMML, we documented that the drug was well tolerated and had a durable clinical benefit in 33% of the patients.61 An interim analysis of the PREACH-M trial (ACTRN12621000223831) evaluating the combination of lenzilumab and 5-azacitidine in CMML patients with RAS pathway mutations has shown encouraging results, with complete remissions achieved within three cycles of treatment in 55% of subjects.62 JAK2 is a primary kinase regulating all the known activities of GM-CSF.63 This, along with its constitutive activation in CMML, provides the rationale for the use of JAK inhibitors, with ruxolitinib having completed early phase testing in CMML (Table 1, Figure 5).64
The role of inflammatory monocytes in chronic myelomonocytic leukemia
In CMML, classical monocytes (CD14+/CD16-) represent the predominant monocyte subset (>94% of total) (Figure 1B).65 These have highly inflammatory transcriptional signatures66 and, as a result, CMML patients have substantially different cytokine expression levels compared to healthy controls.6 Accordingly, CMML patients have a >2-fold increased risk of cardiovascular events,67 and ~20% have an associated systemic inflammatory and autoimmune disease (Figure 3).68 The effects of clonal monocytes go beyond the role of these cells in inflammation and organ infiltration. CMML commonly co-occurs with histiocytic malignancies that share ancestral mutations with CMML.69-71 While this suggests a common cell of origin, it also raises the possibility that neoplastic monocyte-derived macrophages contribute to the phenotypic origins of histiocytic neoplasms.
Beyond clinical manifestations, inflammation is also implicated in disease progression (Figure 3). Chronic inflammation can fuel clonal expansion of mutated HSC while inhibiting the function of wild-type HSC.72 In a murine model of Tet2-/-, increased IL-6 levels, due to blood dissemination of gut bacteria from a dysfunctional small-intestinal barrier, were associated with a CMML-like disease.73 In addition, when inflammation is active, TET2-mediated regulation of active chromatin facilitates histone deacetylation and suppresses IL6 and IL-1|3 expression, leading to resolution of inflammation in innate myeloid cells74 and macrophages.75 As a result, in the presence of TET2 loss-of-function variants, myeloid cells show a reduced capacity to resolve inflammation. In addition, Tet2-deficient murine and TET2-mutant human HSC, when exposed to high levels of pro-inflammatory tumor necrosis factor-a in vitro, have a strong proliferative advantage compared with wild-type cells.76 Equally, in vivo, under inflammatory “stress”, murine Tet2-deficient HSC expand rapidly, which results in enhanced production of inflammatory cytokines, including IL-6, and resistance to apoptosis.77
Likewise, mutations affecting pre-mRNA splicing, including SRSF2 mutations, have also been shown to result in the hyperactivation of nuclear factor-KB signaling pathways.78,79 The presence of clonal myeloid cells is therefore likely to sustain a “vicious circle” of inflammation and clonal expansion in which an inflammatory milieu confers a selective advantage to the mutant clone, while at the same time, the mutant clone is able to perpetuate inflammation. Further underscoring this concept, autocrine or paracrine activation of MAPK though C-C chemokine receptor type 2 (CCR2) has been implicated in defective apoptosis of circulating classical monocytes in CMML.80
The progression of chronic myelomonocytic leukemia to acute myeloid leukemia
Rates of progression to secondary AML remain high,2 with current survival outcomes after blast transformation remaining dismal (median overall survival, <9 months).2 Acquisition of additional somatic driver mutations in the coding DNA, or increments in variant allele fraction of existing driver mutations, explain transformation in only 40-60% of patients.38,81 Mutations in the non-coding genome, somatic copy number alterations, and epigenetic mechanisms, likely in the context of reduced immune surveillance, emerge as plausible mechanisms that need to be explored. Understanding steps that lead to AML transformation is a much-needed area of research, and a critical one to improve outcomes in patients.
Contemporary therapeutic approaches for chronic myelomonocytic leukemia
Allogeneic hematopoietic stem cell transplant is the only potential cure for CMML; however, due to older age and comorbidities, this approach is unfeasible for the majority of patients. DNA methyltransferase (DNMT) inhibitors, also referred to as hypomethylating agents, remain the only Food and Drug Administration-approved treatment options for CMML, but overall response rates are <50%, with true remissions being achieved in <20% patients.82,83 In a large multicenter study, we showed that the best predictor of response to DNMT inhibitors is the presence of TET2 mutations in the absence of ASXL1 mutations.84 Although use of DNMT inhibitors can lead to demethylation at promoters and CpG islands,83 response to these inhibitors occurs in the absence of significant variation in the clonal structure.85 Several studies have shown a lack of correlation between differences in promoter methylation and transcriptional changes in CMML,37,38 a phenomenon that might explain why, despite an epigenetic effect, DNMT inhibitors do not affect the mutant allelic burdens, nor alter the natural history of the disease.86 Ascorbic acid is an important cofactor for TET dioxygenase activity, prompting the use of parenteral ascorbic acid in combination with DNMT inhibitors in TET2-mutant CMML. In this setting, ascorbic acid has been postulated to enhance TET2 activity generated from the unmutated allele and/or exploit functional redundancies with TET3 in the hematopoietic system.87
The synergistic effect of venetoclax and 5-azacitidine is much less pronounced in CMML than in AML.88,89 Some mechanistic insights come from studies in AML patients. Monocytic and/or RAS-mutated AML is more resistant to BCL2 inhibition, and thus venetoclax can favor the outgrowth of monocytic subpopulations that arise from low variant allele fraction NRAS-mutated and KRAS-mutated clones. These clones activate an MLL-specific leukemia stem cell signature and show dependency on the anti-apoptotic protein MCL-1.90 CMML monocytes too show dependency on MCL-1, resulting in defective apoptosis, with a combination of MCL1 and MEK inhibitors showing early promise in xenografts models.80 Although, traditionally, the RAS/RAF/MEK/ERK pathway has been difficult to target in hematologic malignancies due to the lack of effective drugs and the late and subclonal nature of its mutations, CMML is an exception, given that in proliferative CMML, RAS mutations are often early and dominant clonal events.38 Novel RAS-directed therapies including the on and off KRAS G12C and G12D inhibitors and the pan-RAS GTPase inhibitors can play an important role in the management of proliferative CMML,91,92 and can inform safety and dosing in myeloid neoplasms, in which RAS mutations play a role in disease progression and contribute to resistance to FLT3, IDH1, IDH2, and BCL2 inhibitors.
Inhibition of the RAS-activated PI3K pathway is an enticing yet underexplored approach in CMML. As dual MAPK/PI3K pathway inhibition resulted in dose-limiting toxicities in solid tumors,52 other combinatorial strategies are needed. For example, the combination of the PI3Kd inhibitor, umbralisib, and the JAK1/2 inhibitor, ruxolitinib, was synergistic in pre-clinical colony-forming assays using primary CMML samples.93 This combination has since entered early phase clinical trials (NCT02493530). Moreover, in addition to their impact on methylation, DNMT inhibitors form covalent DNA-DNMT1 adducts that invoke an ATR-CHK1-mediated DNA damage response.94-96 Thus, combinations of DNMT inhibitors with inhibitors of cell cycle checkpoints are of particular interest. Specifically, polo-like kinase 1 (PLK1) is upregulated in RAS-mutated proliferative CMML and PLK1 inhibition was efficacious in patient-derived xenograft models of proliferative CMML.38 Accordingly, the PLK1 inhibitor, onvansertib, has entered early phase clinical trials (NCT05549661). Combinations with other cell cycle checkpoint and DNA damage repair inhibitors remain to be explored. Additional targets of interest are outlined in Figures 4 and 5, with reported results in CMML cohorts summarized in Table 1.
Conclusion
Epigenetic dysregulation in CMML leads to myeloid bias and clonal monocytosis. Subsequent acquisition of epigenetic/ transcription factor mutations typically results in dysplastic CMML, whereas signaling mutations are more commonly associated with the more aggressive proliferative CMML.86 Increased activation of pro-inflammatory pathways in clonal monocytes as well as accumulation of leukemia-derived plasmacytoid dendritic cells, causing suppression of the adaptive immune system, drive more severe clinical manifestations with inferior outcomes. Despite active research, there remains an unmet clinical need to improve outcomes for CMML patients. Single-agent therapy fails to alter disease biology and the complex pathophysiology of CMML highlights the need to explore combination strategies, with several clinical trials currently underway.
Footnotes
- Received June 12, 2024
- Accepted October 4, 2024
Correspondence
Disclosures
MMP has received research funding from Kura Oncology, Stem Line, Epigenetix, Polaris, and Solutherapeutics.
Funding
This work was supported in part by funding from the NIH National Cancer Institute (R01CA272496) to MMP.
References
- Arber DA, Orazi A, Hasserjian RP. International Consensus Classification of myeloid neoplasms and acute leukemias: integrating morphologic, clinical, and genomic data. Blood. 2022; 140(11):1200-1228. Google Scholar
- Patnaik MM, Tefferi A. Atypical chronic myeloid leukemia and myelodysplastic/myeloproliferative neoplasm, not otherwise specified: 2023 update on diagnosis, risk stratification, and management. Am J Hematol. 2023; 98(4):681-689. Google Scholar
- Merlevede J, Droin N, Qin T. Mutation allele burden remains unchanged in chronic myelomonocytic leukaemia responding to hypomethylating agents. Nat Commun. 2016; 7:10767. Google Scholar
- Ball M, List AF, Padron E. When clinical heterogeneity exceeds genetic heterogeneity: thinking outside the genomic box in chronic myelomonocytic leukemia. Blood. 2016; 128(20):2381-2387. Google Scholar
- Gelsi-Boyer V, Trouplin V, Roquain J. ASXL1 mutation is associated with poor prognosis and acute transformation in chronic myelomonocytic leukaemia. Br J Haematol. 2010; 151(4):365-375. Google Scholar
- Niyongere S, Lucas N, Zhou JM. Heterogeneous expression of cytokines accounts for clinical diversity and refines prognostication in CMML. Leukemia. 2019; 33(1):205-216. Google Scholar
- Mitchell E, Spencer Chapman M, Williams N. Clonal dynamics of haematopoiesis across the human lifespan. Nature. 2022; 606(7913):343-350. Google Scholar
- Fabre MA, de Almeida JG, Fiorillo E. The longitudinal dynamics and natural history of clonal haematopoiesis. Nature. 2022; 606(7913):335-342. Google Scholar
- Mason CC, Khorashad JS, Tantravahi SK. Age-related mutations and chronic myelomonocytic leukemia. Leukemia. 2016; 30(4):906-913. Google Scholar
- Genovese G, Kähler AK, Handsaker RE. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014; 371(26):2477-2487. Google Scholar
- Jaiswal S, Fontanillas P, Flannick J. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014; 371(26):2488-2498. Google Scholar
- Abelson S, Collord G, Ng SWK. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature. 2018; 559(7714):400-404. Google Scholar
- Kuranda K, Vargaftig J, de la Rochere P. Age-related changes in human hematopoietic stem/progenitor cells. Aging Cell. 2011; 10(3):542-546. Google Scholar
- Plowden J, Renshaw-Hoelscher M, Engleman C, Katz J, Sambhara S. Innate immunity in aging: impact on macrophage function. Aging Cell. 2004; 3(4):161-167. Google Scholar
- Chambers SM, Shaw CA, Gatza C, Fisk CJ, Donehower LA, Goodell MA. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. 2007; 5(8):e201. Google Scholar
- Patnaik MM, Lasho TL, Vijayvargiya P. Prognostic interaction between ASXL1 and TET2 mutations in chronic myelomonocytic leukemia. Blood Cancer J. 2016; 6(1)Google Scholar
- Mei M, Pillai R, Kim S. The mutational landscape in chronic myelomonocytic leukemia and its impact on allogeneic hematopoietic cell transplantation outcomes: a Center for Blood and Marrow Transplantation Research (CIBMTR) analysis. Haematologica. 2023; 108(1):150-160. Google Scholar
- Itzykson R, Kosmider O, Renneville A. Prognostic score including gene mutations in chronic myelomonocytic leukemia. J Clin Oncol. 2013; 31(19):2428-2436. Google Scholar
- Patnaik MM, Itzykson R, Lasho TL. ASXL1 and SETBP1 mutations and their prognostic contribution in chronic myelomonocytic leukemia: a two-center study of 466 patients. Leukemia. 2014; 28(11):2206-2212. Google Scholar
- Patnaik MM, Parikh SA, Hanson CA, Tefferi A. Chronic myelomonocytic leukaemia: a concise clinical and pathophysiological review. Br J Haematol. 2014; 165(3):273-286. Google Scholar
- Papaemmanuil E, Gerstung M, Malcovati L. Chronic Myeloid Disorders Working Group of the International Cancer Genome Consortium. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013; 122(22):3616-3627. Google Scholar
- Palomo L, Meggendorfer M, Hutter S. Molecular landscape and clonal architecture of adult myelodysplastic/ myeloproliferative neoplasms. Blood. 2020; 136(16):1851-1862. Google Scholar
- Awada H, Nagata Y, Goyal A. Invariant phenotype and molecular association of biallelic TET2 mutant myeloid neoplasia. Blood Adv. 2019; 3(3):339-349. Google Scholar
- Rasmussen KD, Jia G, Johansen JV. Loss of TET2 in hematopoietic cells leads to DNA hypermethylation of active enhancers and induction of leukemogenesis. Genes Dev. 2015; 29(9):910-922. Google Scholar
- Ito K, Lee J, Chrysanthou S. Non-catalytic roles of Tet2 are essential to regulate hematopoietic stem and progenitor cell homeostasis. Cell Rep. 2019; 28(10):2480-2490.e4. Google Scholar
- Li Z, Cai X, Cai CL. Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. Blood. 2011; 118(17):4509-4518. Google Scholar
- Ostrander EL, Kramer AC, Mallaney C. Divergent effects of Dnmt3a and Tet2 mutations on hematopoietic progenitor cell fitness. Stem Cell Rep. 2020; 14(4):551-560. Google Scholar
- Jakobsen NA, Turkalj S, Zeng AGX. Selective advantage of mutant stem cells in human clonal hematopoiesis is associated with attenuated response to inflammation and aging. Cell Stem Cell. 2024; 31(8):1127-1144. Google Scholar
- Zhang J, Lieu YK, Ali AM. Disease-associated mutation in SRSF2 misregulates splicing by altering RNA-binding affinities. Proc Natl Acad Sci U S A. 2015; 112(34):E4726-4734. Google Scholar
- Kim E, Ilagan JO, Liang Y. SRSF2 mutations contribute to myelodysplasia by mutant-specific effects on exon recognition. Cancer Cell. 2015; 27(5):617-630. Google Scholar
- Kon A, Yamazaki S, Nannya Y. Physiological Srsf2 P95H expression causes impaired hematopoietic stem cell functions and aberrant RNA splicing in mice. Blood. 2018; 131(6):621-635. Google Scholar
- Smeets MF, Tan SY Xu JJ. Srsf2P95H initiates myeloid bias and myelodysplastic/myeloproliferative syndrome from hemopoietic stem cells. Blood. 2018; 132(6):608-621. Google Scholar
- Gelsi-Boyer V, Trouplin V, Adélaïde J. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009; 145(6):788-800. Google Scholar
- Abdel-Wahab O, Adli M, LaFave LM. ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell. 2012; 22(2):180-193. Google Scholar
- Asada S, Goyama S, Inoue D. Mutant ASXL1 cooperates with BAP1 to promote myeloid leukaemogenesis. Nat Commun. 2018; 9(1):2733. Google Scholar
- Campagne A, Lee MK, Zielinski D. BAP1 complex promotes transcription by opposing PRC1-mediated H2A ubiquitylation. Nat Commun. 2019; 10(1):348. Google Scholar
- Binder M, Carr RM, Lasho TL. Oncogenic gene expression and epigenetic remodeling of cis-regulatory elements in ASXL1-mutant chronic myelomonocytic leukemia. Nat Commun. 2022; 13(1):1434. Google Scholar
- Carr RM, Vorobyev D, Lasho T. RAS mutations drive proliferative chronic myelomonocytic leukemia via a KMT2A-PLK1 axis. Nat Commun. 2021; 12(1):2901. Google Scholar
- Bezerra ED, Lasho TL, Finke CM. CSF3R T618I mutant chronic myelomonocytic leukemia (CMML) defines a proliferative CMML subtype enriched in ASXL1 mutations with adverse outcomes. Blood Cancer J. 2021; 11(3):54. Google Scholar
- Corces-Zimmerman MR, Hong WJ, Weissman IL, Medeiros BC, Majeti R. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Natl Acad Sci U S A. 2014; 111(7):2548-2553. Google Scholar
- Shlush LI, Zandi S, Mitchell A. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature. 2014; 506(7488):328-333. Google Scholar
- Lasho T, Patnaik MM. Juvenile myelomonocytic leukemia - a bona fide RASopathy syndrome. Best Pract Res Clin Haematol. 2020; 33(2):101171. Google Scholar
- Li Q, Bohin N, Wen T. Oncogenic Nras has bimodal effects on stem cells that sustainably increase competitiveness. Nature. 2013; 504(7478):143-147. Google Scholar
- Hérault A, Binnewies M, Leong S. Myeloid progenitor cluster formation drives emergency and leukaemic myelopoiesis. Nature. 2017; 544(7648):53-58. Google Scholar
- Ferrall-Fairbanks MC, Dhawan A, Johnson B. Progenitor hierarchy of chronic myelomonocytic leukemia identifies inflammatory monocytic-biased trajectory linked to worse outcomes. Blood Cancer Discov. 2022; 3(6):536-553. Google Scholar
- Hamarsheh S, Osswald L, Saller BS. Oncogenic KrasG12D causes myeloproliferation via NLRP3 inflammasome activation. Nat Commun. 2020; 11(1):1659. Google Scholar
- Hurtado-Navarro L, Cuenca-Zamora EJ, Zamora L. NLRP3 inflammasome activation and symptom burden in KRAS-mutated CMML patients is reverted by IL-1 blocking therapy. Cell Rep Med. 2023; 4(12):101329. Google Scholar
- Broz P, Pelegrín P, Shao F. The gasdermins, a protein family executing cell death and inflammation. Nat Rev Immunol. 2020; 20(3):143-157. Google Scholar
- Baroja-Mazo A, Martín-Sánchez F, Gomez AI. The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014; 15(8):738-748. Google Scholar
- Franklin BS, Bossaller L, De Nardo D. The adaptor ASC has extracellular and ‘prionoid’ activities that propagate inflammation. Nat Immunol. 2014; 15(8):727-737. Google Scholar
- Schmid MC, Avraamides CJ, Dippold HC. Receptor tyrosine kinases and TLR/IL1Rs unexpectedly activate myeloid cell PI3kg, a single convergent point promoting tumor inflammation and progression. Cancer Cell. 2011; 19(6):715-727. Google Scholar
- Moore AR, Rosenberg SC, McCormick F, Malek S. RAS-targeted therapies: is the undruggable drugged?. Nat Rev Drug Discov. 2020; 19(8):533-552. Google Scholar
- You X, Liu F, Binder M. Asxl1 loss cooperates with oncogenic Nras in mice to reprogram the immune microenvironment and drive leukemic transformation. Blood. 2022; 139(7):1066-1079. Google Scholar
- Lucas N, Duchmann M, Rameau P. Biology and prognostic impact of clonal plasmacytoid dendritic cells in chronic myelomonocytic leukemia. Leukemia. 2019; 33(10):2466-2480. Google Scholar
- Mangaonkar AA, Reichard KK, Binder M. Bone marrow dendritic cell aggregates associate with systemic immune dysregulation in chronic myelomonocytic leukemia. Blood Adv. 2020; 4(21):5425-5430. Google Scholar
- Munn DH, Mellor AL. IDO in the tumor microenvironment: inflammation, counter-regulation, and tolerance. Trends Immunol. 2016; 37(3):193-207. Google Scholar
- Munn DH, Zhou M, Attwood JT. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998; 281(5380):1191-1193. Google Scholar
- Patnaik MM, Pophali PA, Lasho TL. Clinical correlates, prognostic impact and survival outcomes in chronic myelomonocytic leukemia patients with the JAK2V617F mutation. Haematologica. 2019; 104(6):e236-e239. Google Scholar
- Patnaik MM, Timm MM, Vallapureddy R. Flow cytometry based monocyte subset analysis accurately distinguishes chronic myelomonocytic leukemia from myeloproliferative neoplasms with associated monocytosis. Blood Cancer J. 2017; 7(7):e584. Google Scholar
- Padron E, Painter JS, Kunigal S. GM-CSF-dependent pSTAT5 sensitivity is a feature with therapeutic potential in chronic myelomonocytic leukemia. Blood. 2013; 121(25):5068-5077. Google Scholar
- Patnaik MM, Sallman DA, Mangaonkar AA. Phase 1 study of lenzilumab, a recombinant anti-human GM-CSF antibody, for chronic myelomonocytic leukemia. Blood. 2020; 136(7):909-913. Google Scholar
- Hiwase D, Ross DM, Steven W. Lenzilumab in addition to azacitidine improves complete response rates in chronic myelomonocytic leukemia. Blood. 2023; 142(Supplement 1):1847. Google Scholar
- Watanabe S, Itoh T, Arai K. Roles of JAK kinases in human GM-CSF receptor signal transduction. J Allergy Clin Immunol. 1996; 98(6 Pt 2):S183-191. Google Scholar
- Padron E, Dezern A, Andrade-Campos M. Myelodysplastic Syndrome Clinical Research Consortium. A multi-institution phase I trial of ruxolitinib in patients with chronic myelomonocytic leukemia (CMML). Clin Cancer Res. 2016; 22(15):3746-3754. Google Scholar
- Selimoglu-Buet D, Wagner-Ballon O, Saada V. Characteristic repartition of monocyte subsets as a diagnostic signature of chronic myelomonocytic leukemia. Blood. 2015; 125(23):3618-3626. Google Scholar
- Franzini A, Pomicter AD, Yan D. The transcriptome of CMML monocytes is highly inflammatory and reflects leukemiaspecific and age-related alterations. Blood Adv. 2019; 3(20):2949-2961. Google Scholar
- Elbæk MV, Sørensen AL, Hasselbalch HC. Cardiovascular disease in chronic myelomonocytic leukemia: do monocytosis and chronic inflammation predispose to accelerated atherosclerosis?. Ann Hematol. 2019; 98(1):101-109. Google Scholar
- Mekinian A, Grignano E, Braun T. Systemic inflammatory and autoimmune manifestations associated with myelodysplastic syndromes and chronic myelomonocytic leukaemia: a French multicentre retrospective study. Rheumatology (Oxford). 2016; 55(2):291-300. Google Scholar
- Papo M, Diamond EL, Cohen-Aubart F. High prevalence of myeloid neoplasms in adults with non-Langerhans cell histiocytosis. Blood. 2017; 130(8):1007-1013. Google Scholar
- Bonnet P, Chasset F, Moguelet P. Erdheim-Chester disease associated with chronic myelomonocytic leukemia harboring the same clonal mutation. Haematologica. 2019; 104(11):e530-e533. Google Scholar
- Ghobadi A, Miller CA, Li T. Shared cell of origin in a patient with Erdheim-Chester disease and acute myeloid leukemia. Haematologica. 2019; 104(8):e373-e375. Google Scholar
- Pietras EM, Mirantes-Barbeito C, Fong S. Chronic interleukin-1 exposure drives haematopoietic stem cells towards precocious myeloid differentiation at the expense of self-renewal. Nat Cell Biol. 2016; 18(6):607-618. Google Scholar
- Meisel M, Hinterleitner R, Pacis A. Microbial signals drive pre-leukaemic myeloproliferation in a Tet2-deficient host. Nature. 2018; 557(7706):580-584. Google Scholar
- Zhang Q, Zhao K, Shen Q. Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6. Nature. 2015; 525(7569):389-393. Google Scholar
- Cull AH, Snetsinger B, Buckstein R, Wells RA, Rauh MJ. Tet2 restrains inflammatory gene expression in macrophages. Exp Hematol. 2017; 55:56-70. Google Scholar
- Abegunde SO, Buckstein R, Wells RA, Rauh MJ. An inflammatory environment containing TNFa favors Tet2-mutant clonal hematopoiesis. Exp Hematol. 2018; 59:60-65. Google Scholar
- Cai Z, Kotzin JJ, Ramdas B. Inhibition of inflammatory signaling in Tet2 mutant preleukemic cells mitigates stress-induced abnormalities and clonal hematopoiesis. Cell Stem Cell. 2018; 23(6):833-849. Google Scholar
- Lee SC, North K, Kim E. Synthetic lethal and convergent biological effects of cancer-associated spliceosomal gene mutations. Cancer Cell. 2018; 34(2):225-241. Google Scholar
- Smith MA, Choudhary GS, Pellagatti A. U2AF1 mutations induce oncogenic IRAK4 isoforms and activate innate immune pathways in myeloid malignancies. Nat Cell Biol. 2019; 21(5):640-650. Google Scholar
- Sevin M, Debeurme F, Laplane L. Cytokine-like protein 1-induced survival of monocytes suggests a combined strategy targeting MCL1 and MAPK in CMML. Blood. 2021; 137(24):3390-3402. Google Scholar
- Castaño-Díez S, López-Guerra M, Bosch-Castañeda C. Real-world data on chronic myelomonocytic leukemia: clinical and molecular characteristics, treatment, emerging drugs, and patient outcomes. Cancers (Basel). 2022; 14(17):4107. Google Scholar
- Santini V, Allione B, Zini G. A phase II, multicentre trial of decitabine in higher-risk chronic myelomonocytic leukemia. Leukemia. 2018; 32(2):413-418. Google Scholar
- Coston T, Pophali P, Vallapureddy R. Suboptimal response rates to hypomethylating agent therapy in chronic myelomonocytic leukemia; a single institutional study of 121 patients. Am J Hematol. 2019; 94(7):767-779. Google Scholar
- Coltro G, Mangaonkar AA, Lasho TL. Clinical, molecular, and prognostic correlates of number, type, and functional localization of TET2 mutations in chronic myelomonocytic leukemia (CMML) - a study of 1084 patients. Leukemia. 2020; 34(5):1407-1421. Google Scholar
- Schnegg-Kaufmann AS, Thoms JAI, Bhuyan GS. Contribution of mutant HSC clones to immature and mature cells in MDS and CMML, and variations with AZA therapy. Blood. 2023; 141(11):1316-1321. Google Scholar
- Itzykson R, Solary E. An evolutionary perspective on chronic myelomonocytic leukemia. Leukemia. 2013; 27(7):1441-1450. Google Scholar
- Guan Y, Greenberg EF, Hasipek M. Context dependent effects of ascorbic acid treatment in TET2 mutant myeloid neoplasia. Commun Biol. 2020; 3(1):493. Google Scholar
- DiNardo CD, Jonas BA, Pullarkat V. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020; 383(7):617-662. Google Scholar
- Tremblay D, Csizmar CM, DiNardo CD. Venetoclax (VEN) improves response rates but not survival in patients with chronic myelomonocytic leukemia (CMML) treated with hypomethylating agents (HMA): a multicenter, propensity score analysis. Blood. 2023; 142(Supplement 1):321. Google Scholar
- Pei S, Pollyea DA, Gustafson A. monocytic subclones confer resistance to venetoclax-based therapy in patients with acute myeloid leukemia. Cancer Discov. 2020; 10(4):536-551. Google Scholar
- Punekar SR, Velcheti V, Neel BG, Wong KK. The current state of the art and future trends in RAS-targeted cancer therapies. Nat Rev Clin Oncol. 2022; 19(10):637-655. Google Scholar
- Holderfield M, Lee BJ, Jiang J. Concurrent inhibition of oncogenic and wild-type RAS-GTP for cancer therapy. Nature. 2024; 629(8013):919-926. Google Scholar
- Villaume MT, Arrate MP, Ramsey HE. The delta isoform of phosphatidylinositol-3-kinase predominates in chronic myelomonocytic leukemia and can be targeted effectively with umbralisib and ruxolitinib. Exp Hematol. 2021; 97:57-65. Google Scholar
- Palii SS, Van Emburgh BO, Sankpal UT, Brown KD, Robertson KD. DNA methylation inhibitor 5-Aza-2’-deoxycytidine induces reversible genome-wide DNA damage that is distinctly influenced by DNA methyltransferases 1 and 3B. Mol Cell Biol. 2008; 28(2):752-771. Google Scholar
- Thépot S, Lainey E, Cluzeau T. Hypomethylating agents reactivate FOXO3A in acute myeloid leukemia. Cell Cycle. 2011; 10(14):2323-2330. Google Scholar
- Yabushita T, Chinen T, Nishiyama A, Asada S, Shimura R, Isobe T. Mitotic perturbation is a key mechanism of action of decitabine in myeloid tumor treatment. Cell Rep. 2023; 42(9):113098. Google Scholar
- Itzykson R, Santini V, Thepot S. Decitabine versus hydroxyurea for advanced proliferative chronic myelomonocytic leukemia: results of a randomized phase III trial within the EMSCO network. J Clin Oncol. 2023; 41(10):1888-1897. Google Scholar
- Kobayashi Y, Munakata W, Ogura M. Phase I study of panobinostat and 5-azacitidine in Japanese patients with myelodysplastic syndrome or chronic myelomonocytic leukemia. Int J Hematol. 2018; 107(1):83-91. Google Scholar
- Gillberg L, Ørskov AD, Nasif A. Oral vitamin C supplementation to patients with myeloid cancer on azacitidine treatment: normalization of plasma vitamin C induces epigenetic changes. Clin Epigenetics. 2019; 11(1):143. Google Scholar
- Xie Z, McCullough K, Lasho TL. Phase II study assessing safety and preliminary efficacy of high dose intravenous ascorbic acid in patients with TET2 mutant clonal cytopenias. Blood. 2023; 142(Supplement 1):4611. Google Scholar
- Yee KW, Chen HW, Hedley DW. A phase I trial of the aurora kinase inhibitor, ENMD-2076, in patients with relapsed or refractory acute myeloid leukemia or chronic myelomonocytic leukemia. Invest New Drugs. 2016; 34(5):614-624. Google Scholar
- Navada SC, Garcia-Manero G, OdchimarReissig R. Rigosertib in combination with azacitidine in patients with myelodysplastic syndromes or acute myeloid leukemia: results of a phase 1 study. Leuk Res. 2020; 94:106369. Google Scholar
- Short NJ, Jabbour E, Naqvi K. A phase II study of omacetaxine mepesuccinate for patients with higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia after failure of hypomethylating agents. Am J Hematol. 2019; 94(1):74-79. Google Scholar
- Steensma DP, Wermke M, Klimek VM. Phase I first-inhuman dose escalation study of the oral SF3B1 modulator H3B-8800 in myeloid neoplasms. Leukemia. 2021; 35(12):3542-3550. Google Scholar
- Assi R, Kantarjian HM, Garcia-Manero G. A phase II trial of ruxolitinib in combination with azacytidine in myelodysplastic syndrome/myeloproliferative neoplasms. Am J Hematol. 2018; 93(2):277-285. Google Scholar
- Hunter AM, Newman H, Dezern AE. Integrated human and murine clinical study establishes clinical efficacy of ruxolitinib in chronic myelomonocytic leukemia. Clin Cancer Res. 2021; 27(22):6095-6105. Google Scholar
- Padron E, Tinsley-Vance SM, DeZern AE. Efficacy and safety of ruxolitinib for treatment of symptomatic chronic myelomonocytic leukemia (CMML): results of a multicenter phase II clinical trial. Blood. 2022; 140(Supplement 1):1101-1103. Google Scholar
- Borthakur G, Popplewell L, Boyiadzis M. Activity of the oral mitogen-activated protein kinase kinase Inhibitor trametinib in RAS-mutant relapsed or refractory myeloid malignancies. Cancer. 2016; 122(12):1871-1879. Google Scholar
- Feldman EJ, Cortes J, DeAngelo DJ. On the use of lonafarnib in myelodysplastic syndrome and chronic myelomonocytic leukemia. Leukemia. 2008; 22(9):1707-1711. Google Scholar
- Ravoet C, Mineur P, Robin V. Farnesyl transferase inhibitor (lonafarnib) in patients with myelodysplastic syndrome or secondary acute myeloid leukaemia: a phase II study. Ann Hematol. 2008; 87(11):881-885. Google Scholar
- Buckstein R, Kerbel R, Cheung M. Lenalidomide and metronomic melphalan for CMML and higher risk MDS: a phase 2 clinical study with biomarkers of angiogenesis. Leuk Res. 2014; 38(7):756-763. Google Scholar
- Kenealy M, Patton N, Filshie R. Results of a phase II study of thalidomide and azacitidine in patients with clinically advanced myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia (CMML) and low blast count acute myeloid leukemia (AML). Leuk Lymphoma. 2017; 58(2):298-307. Google Scholar
- Burgstaller S, Stauder R, Kuehr T. A phase I study of lenalidomide in patients with chronic myelomonocytic leukemia (CMML) - AGMT_CMML-1. Leuk Lymphoma. 2018; 59(5):1121-1126. Google Scholar
- Sekeres MA, Othus M, List AF. Randomized phase II study of azacitidine alone or in combination with lenalidomide or with vorinostat in higher-risk myelodysplastic syndromes and chronic myelomonocytic leukemia: North American Intergroup Study SWOG S1117. J Clin Oncol. 2017; 35(24):2745-2753. Google Scholar
- Adès L, Duployez N, Guerci-Bresler A. A randomised phase II study of azacitidine (AZA) alone or with lenalidomide (LEN), valproic acid (VPA) or idarubicin (IDA) in higher-risk MDS or low blast AML: GFM’s “pick a winner” trial, with the impact of somatic mutations. Br J Haematol. 2022; 198(3):535-544. Google Scholar
- Attar EC, Amrein PC, Fraser JW. Phase I dose escalation study of bortezomib in combination with lenalidomide in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Leuk Res. 2013; 37(9):1016-1020. Google Scholar
- Sekeres MA, Watts J, Radinoff A. Randomized phase 2 trial of pevonedistat plus azacitidine versus azacitidine for higher-r.isk MDS/CMML or low-blast AML. Leukemia. 2021; 35(7):2119-2124. Google Scholar
- Short NJ, Muftuoglu M, Ong F. A phase 1/2 study of azacitidine, venetoclax and pevonedistat in newly diagnosed secondary AML and in MDS or CMML after failure of hypomethylating agents. J Hematol Oncol. 2023; 16(1):73. Google Scholar
- Patnaik MM, Ali H, Wang ES. Tagraxofusp (SL-401) in patients with chronic myelomonocytic leukemia (CMML): updated results of an ongoing phase 1/2 trial. Blood. 2021; 138(Supplement 1):538. Google Scholar
- Patnaik MM, Sallman DA, Mangaonkar AA. Phase 1 study of lenzilumab, a recombinant anti-human GM-CSF antibody, for chronic myelomonocytic leukemia. Blood. 2020; 136(7):909-913. Google Scholar
- Hiwase D, Ross DM, Lane SW. Lenzilumab in addition to azacitidine improves complete response rates in chronic myelomonocytic leukemia. Blood. 2023; 142(Supplement 1):1847. Google Scholar
- Brunner AM, Esteve J, Porkka K. Phase Ib study of sabatolimab (MBG453), a novel immunotherapy targeting TIM-3 antibody, in combination with decitabine or azacitidine in high- or very high-risk myelodysplastic syndromes. Am J Hematol. 2024; 99(2):E32-E36. Google Scholar
- Saliba AN, Litzow MR, Gangat N. Outcomes of venetoclaxbased therapy in chronic phase and blast transformed chronic myelomonocytic leukemia. Am J Hematol. 2021; 96(11):E433-E436. Google Scholar
- Montalban-Bravo G, Hammond D, DiNardo CD. Activity of venetoclax-based therapy in chronic myelomonocytic leukemia. Leukemia. 2021; 35(5):1494-1499. Google Scholar
- Tremblay D, Csizmar CM, DiNardo CD. Venetoclax (VEN) improves response rates but not survival in patients with chronic myelomonocytic leukemia (CMML) treated with hypomethylating agents (HMA): a multicenter, propensity score analysis. Blood. 2023; 142(Supplement 1):321. Google Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.