Next Article in Journal
Strategies for Offline Adaptive Biology-Guided Radiotherapy (BgRT) on a PET-Linac Platform
Previous Article in Journal
Procedural Physician-Scientists as Catalysts for Innovation in Team Science and Clinical Care
Previous Article in Special Issue
Molecular Pathways and Targeted Therapies in Relapsed/Refractory Diffuse Large B-Cell Lymphoma (DLBCL)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances and Challenges in the Management of Myelodysplastic Syndromes

Section of Medical Oncology and Hematology, Department of Internal Medicine, Yale School of Medicine, Yale Comprehensive Cancer Center, New Haven, CT 06510, USA
*
Authors to whom correspondence should be addressed.
Cancers 2025, 17(15), 2469; https://doi.org/10.3390/cancers17152469
Submission received: 26 June 2025 / Revised: 21 July 2025 / Accepted: 21 July 2025 / Published: 25 July 2025
(This article belongs to the Special Issue New Insights of Hematology in Cancer)

Simple Summary

Myelodysplastic syndromes/neoplasms (MDS) represent a group of diverse hematological malignancies that lead to bone marrow failure, low blood counts, and a variable risk of progression to acute myeloid leukemia. Over the last two decades, significant insights have been gained into the underlying biology of MDS, including the contribution of genetic mutations, immune dysregulation, and the bone marrow microenvironment to disease pathogenesis. By summarizing recent studies, therapeutic developments, and evolving treatment algorithms through the lens of precision medicine, this review aims to guide clinicians in delivering personalized, risk-adapted care for individuals with lower- and higher-risk MDS, to discuss the unmet needs and main challenges and to inform future research directions in this rapidly advancing field.

Abstract

Myelodysplastic syndromes/neoplasms (MDS) represent a biologically and clinically diverse group of myeloid malignancies marked by cytopenias, morphological dysplasia, and an inherent risk of progression to acute myeloid leukemia. Over the past two decades, the field has made significant advances in characterizing the molecular landscape of MDS, leading to refined classification systems to reflect the underlying genetic and biological diversity. In 2025, the treatment of MDS is increasingly individualized, guided by integrated clinical, cytogenetic, and molecular risk stratification tools. For lower-risk MDS, the treatment paradigm has evolved beyond erythropoiesis-stimulating agents (ESAs) with the introduction of novel effective agents such as luspatercept and imetelstat, as well as shortened schedules of hypomethylating agents (HMAs). For higher-risk disease, monotherapy with HMAs continue to be the standard of care as combination therapies of HMAs with novel agents have, to date, failed to redefine treatment paradigms. The recognition of precursor states like clonal hematopoiesis of indeterminate potential (CHIP) and the increasing use of molecular monitoring will hopefully enable earlier intervention/prevention strategies. This review provides a comprehensive overview of the current treatment approach for MDS, highlighting new classifications, prognostic tools, evolving therapeutic options, and ongoing challenges. We discuss evidence-based recommendations, treatment sequencing, and emerging clinical trials, with a focus on translating biological insights into improved outcomes for patients with MDS.

1. Introduction

Myelodysplastic syndromes/neoplasms (MDS) represent a heterogeneous group of clonal myeloid malignancies characterized by ineffective hematopoiesis, cytopenias, and an inherent risk of progression to acute myeloid leukemia (AML). MDS predominantly affects the older population, with an incidence rate of 26.1 per 100,000 in individuals aged 65 and older and a median age of diagnosis of 77 years [1,2]. Clonal cytopenias of undetermined significance (CCUS), MDS, and AML exist along a continuum defined by diverse morphological, cytogenetic, and molecular features and varying risks of morbidity and mortality. Over the past several years, interest in MDS has grown exponentially, driven mainly by a deeper understanding of the role of the bone marrow microenvironment and immune dysregulation in its pathogenesis [3,4], as well as by advancements in molecular characterization and its impact on prognostication. These insights have fueled the pursuit of novel therapeutic strategies and innovative drug combinations, addressing a critical need for improved treatment options in this population. This progress builds on several decades of the evolution of MDS management from its initial morphological classification in the 1980s to today’s personalized approaches, reflecting substantial advances in diagnosis, risk stratification, and treatment. We aim to comprehensively review the current management and future directions and challenges in treating patients with MDS.

1.1. Challenges in Classification and Risk-Assessment: The Evolving System(s) for MDS

The classification of MDS has evolved from exclusive reliance on morphology-based criteria to incorporating molecular data that better correlate with prognosis [5,6,7]. In 2022, two working groups revised and published novel classifications for MDS: the World Health Organization (WHO) Classification [6] and the International Consensus Classification (ICC) [5]. Both systems aimed to capture the heterogeneity and complexity of MDS and reflect recent advances in understanding the biology of the disease. However, the coexistence of these two parallel systems leads to ambiguity among pathologists, clinicians, and patients, complicating clinical trial design and affecting the interpretation and applicability of research findings. A notable divergence is the introduction by the ICC of an “AML/MDS” category, defined by 10–19% circulating and/or bone marrow blasts (excluding cases with AML-defining cytogenetics and ≥10% blasts) [5]. Additionally, both classification systems now recognize TP53-mutated MDS as a distinct, higher-risk entity (‘MDS with biallelic TP53 inactivation’ in WHO and ‘myeloid neoplasms with TP53’ in ICC) [5,6]. Supporting the emphasis on molecular classification, Bernard et al. demonstrated that blast percentage alone did not universally predict the outcome among molecular subgroups, suggesting that genetically-defined subtypes should drive future MDS classification approaches [8,9]. Furthermore, recent findings from the International Consortium for MDS (icMDS) demonstrated significant molecular heterogeneity within morphologically-defined MDS cases, with specific mutations correlating distinctly with clinical phenotype and prognosis [10]. Collectively, these advancements advocate for a greater reliance on molecular signatures to enhance diagnostic precision, prognostic accuracy, and therapeutic decision making, including the expansion of more intensive therapies and clinical trials that mirror the aggressiveness of the disease phenotype [11,12].
The field would greatly benefit from the unification of these systems into a single, consensus-based classification framework. Such an alliance should integrate emerging knowledge of the biology, prognosis, and novel technological advances. For an in-depth review of the classification systems and proposed synchronization strategies, the reader is directed to an expert report by the icMDS [13].

1.2. Risk-Assessment in the Era of Precision Medicine

The prognosis of MDS varies significantly depending on the patient’s age, degree of cytopenias, percentage of bone marrow blasts, and cytogenetic abnormalities. The International Prognostic Scoring System (IPSS) [14] and revised IPSS (IPSS-R) [15] are well established and validated tools widely used in clinical practice and clinical trials to assess disease risk, inform treatment decisions, and assess trial eligibility. However, these prognostic tools fall short of accounting for the increasingly relevant genetic landscape. The newly developed molecular IPSS (IPSS-M) incorporates key genetic alterations and offers a more comprehensive risk stratification reflecting the robust association of gene mutations on prognosis [16]. The IPSS-M has been validated in multiple studies, including in a higher risk cohort of patients treated with hypomethylating agents (HMA) [17], and performs well in patients with LR-MDS and those treated with an allogeneic hematopoietic stem cell transplant (allo-HCT) [18]. As the prognostic significance of additional, rarer, and currently less well-defined genetic alterations becomes clearer, the scoring systems will likely continue to evolve and become more precise and individualized.
Lower risk MDS (LR-MDS) is commonly defined as having low or intermediate-1 risk groups according to IPSS (score ≤ 1); very low, low, or some patients with intermediate risk groups by IPSS-R (score ≤ 3.5); and very low, low, and moderate low under IPSS-M (score ≤ 0). In contrast, higher risk MDS (HR-MDS) includes individuals with scores exceeding these thresholds. We will adhere to these definitions throughout the review.

1.3. Treatment of Lower-Risk MDS

The primary treatment goal for individuals with LR-MDS is to improve and/or preserve quality of life by reducing transfusion burden and complications associated with cytopenias. Arguably, treatment also should aim to cure the disease and prolong survival as patient outcomes within the “lower risk” category can be highly variable. The median overall survival (OS) for IPSS-R very low, low, and intermediate risk is 8.8, 5.3, and 3 years, respectively [15], with similar duration for low and moderate low IPSS-M (4.6–6 years) [16].
Erythropoiesis-stimulating agents (ESAs) remain a mainstay of therapy for LR-MDS and are discussed in detail in later sections. Novel therapies for LR-MDS have primarily benefited patients with anemia as the predominant cytopenia. In contrast, treatment options for those with clinically significant isolated thrombocytopenia and/or neutropenia remain limited and rely most on supportive care measures or hypomethylating agents (HMA). Immunosuppressive-based therapy can be considered for selected patients with hypoplastic MDS [19], HLA-DR15 histocompatibility type, or a paroxysmal nocturnal hemoglobinuria clone, similar to the management of acquired aplastic anemia [20]. Thrombopoietin agonists, such as eltrombopag [21] and romiplostim [22], have been investigated in this population given their ability to stimulate not only platelet count recovery but also trilineage hematopoiesis, as observed in patients with aplastic anemia [23]. While they have been shown to reduce bleeding events, they carry a higher incidence of grade 3 and 4 adverse events. A recent study showed no difference in clonal evolution with eltrombopag compared to placebo in patients with LR-MDS; however, a theoretical risk of accelerated progression to AML remains a concern when these agents are used in HR-MDS [21,24]. Asymptomatic patients who do not require platelet or red blood cell (RBC) transfusions and have an absolute neutrophil count > 500 cell/microL may be managed with a “watch and wait” approach, with laboratory monitoring at regular intervals based on the degree of cytopenias and risk of progression. Supportive care is essential for all patients with MDS, especially those with profound cytopenias which impact quality of life and progression-free survival (PFS) [25,26]. An extensive review on supportive care for patients with MDS is beyond the scope of this paper [27].
Allo-HCT is generally not recommended as the initial treatment strategy for LR-MDS as delayed transplant at disease progression or clonal evolution is associated with improved gain in survival [28,29]. Some exceptions could include patients who have profound and/or refractory cytopenias or who have an underlying germline predisposition to myeloid malignancies [30].
LR-MDS and CCUS, defined as persistent (>4 months) and unexplained cytopenias with evidence of clonal hematopoiesis in the absence of morphological dysplasia, share some overlapping clinical and molecular features [5]. Xie et al. demonstrated that high-risk CCUS is associated with clinical outcomes comparable to those of LR-MDS and reported that both PFS and OS are similar to those observed in LR-MDS [31]. Recent efforts have led to the development of clinical calculators such as the Clonal Hematopoiesis Risk Score (CHRS) [32] and the Clonal Cytopenia Risk Score (CCRS) [31]. These models incorporate molecular features, including number of mutations and variant allele frequency, to estimate the risk for progression to myeloid neoplasms. The CHRS, derived from large population datasets, stratifies clonal hematopoiesis of indeterminate potential and CCUS into low-, intermediate-, and high-risk categories, with the high-risk group demonstrating a markedly elevated 10-year cumulative incidence for myeloid neoplasm of 52.2%. In contrast, the CCRS was developed exclusively in CCUS cohorts utilizing three adverse factors: presence of splicing factor mutations, number of mutations (≥2), and thrombocytopenia (<100 × 109/L), to predict the 2-year risk of progression to AML/MDS (Table 1). Both scoring systems offer practical frameworks for identifying high-risk individuals with CCUS that may behave biologically akin to LR-MDS who may benefit from closer surveillance, early intervention, or inclusion in clinical trials typically reserved for patients with LR-MDS. These risk models exemplify the shift toward precision risk assessment in the evaluation of clonal cytopenias.

1.4. MDS with Chromosome 5 Deletion

The management of MDS with chromosome 5 deletion (del[5q]) is distinct from other subtypes of MDS as it is highly responsive to lenalidomide [33]. A multicenter phase II study demonstrated that 76% (95% confidence interval [CI], 68–82) of patients had a reduction in the number of transfusions while 67% (95% CI, 59–74) became transfusion independent (median duration of response not reached after 2.2 years of follow-up) [33]. A subsequent phase III study showed a significant improvement in the RBC transfusion independence (RBC-TI) [34] rate among patients treated with lenalidomide 10 mg (61%) and 5 mg (51.1%), compared to the placebo cohort (7.8%) (p < 0.001), as well as an AML-free survival advantage [35]. Among patients who are RBC-TI, Díez-Campelo et al. reported that an early intervention with low dose lenalidomide (5 mg daily) significantly reduced the risk for RBC transfusion dependence by nearly 70% (hazard ratio [HR] = 0.203, 95% CI: 0.132–0.692, p = 0.0046) [36]. The short and long-term benefits of lenalidomide support maximizing the treatment duration in this subgroup of MDS. Nevertheless, the response often is ultimately lost and alternate therapies such as ESAs if erythropoietin [EPO] ≤ 500 mU/mL, HMAs, imetelstat, luspatercept, or clinical trials if EPO > 500 mU/mL can be considered [20].

1.5. Management of Anemia in LR-MDS

The majority of patients with MDS will develop anemia during the course of their disease, and approximately 50% will require RBC transfusions. Traditionally, ESAs have been considered the first-line treatment for patients with LR-MDS and clinically significant anemia (hemoglobin < 10 g/dL or requiring RBC transfusions). ESAs effectively reduce RBC transfusion needs, particularly in patients with low endogenous EPO levels and lower transfusion requirements [37,38]. However, the quality of response is variable, and most patients will relapse to their transfusion-dependent state approximately 2 years after starting treatment [39]. Patients that relapse early experience an increased risk for AML progression and a shorter OS. These implications have driven the exploration of newer agents such as luspatercept and imetelstat (Figure 1).

1.6. Luspatercept

Luspatercept is a transforming growth factor-beta superfamily ligand trap which enhances late-stage erythropoiesis and ameliorates MDS-related anemia [40]. Luspatercept is currently approved for the treatment of anemia in patients with LR-MDS who are ESA-naïve and may require RBC transfusion [41]. Luspatercept is also approved for the treatment of LR-MDS and MDS/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T) with RBC transfusion-dependent (RBC-TD) anemia who have relapse/refractory disease [41].
The phase II PACE-MDS trial included patients with LR-MDS who were RBC-TD and previously treated with ESAs [42]. The study demonstrated erythroid hematological improvement (HI-E) in two thirds of patients, with 38% achieving RBC-TI, regardless of baseline EPO level or prior ESA use [42]. Patients with the lowest transfusion requirements had earlier and more sustained improvement in hemoglobin levels (median increase, 2 g/dL). Additionally, a robust response was seen in patients with MDS with ≥15% ringed sideroblasts (MDS-RS; RBC-TI 42%, HI-E 69%) and MDS with an SF3B1 mutation (RBC-TI 44%, HI-E 77%). Long term results of the PACE-MDS support the use of luspatercept for non-RS MDS which have a nearly 36% HI-E rate [43]. The phase 3 MEDALIST trial, conducted exclusively in patients with MDS with RS ≥ 15% and RS ≥ 5% with an SF3B1 mutation who were RBC-TD and refractory or ineligible to ESAs, showed that 38% of patients on luspatercept achieved RBC-TI for at least 8 weeks over a 24-week period, compared to 13% in the placebo group (p < 0.001) [44].
Frontline luspatercept was compared to epoetin alpha in ESA-naïve and RBC-TD patients in the phase 3 randomized COMMANDS trial [45]. The RBC-TI rate (TI for ≥12 weeks and hemoglobin increase ≥ 1.5 g/dL) was significantly greater with luspatercept compared to ESA (59 vs. 31%, p < 0.001), and correlated with significant improvement in health-related quality of life metrics [46]. Long-term results further confirmed the superiority of luspatercept, with a longer median duration of RBC-TI (35.6 months versus 20.9 months with ESA; p = 0.0016) and a positive OS trend [47], bringing luspatercept to the frontline of RBC-TD LR-MDS [48]. The benefit was irrespective of RS status, although only 26% were RS-negative in this trial. The observed trend towards improved OS with luspatercept with longer follow up of the COMMANDS trial raises the possibility that its role may extend beyond supportive care and warrants a consideration of potential natural history modifying effects. Luspatercept is being compared to ESA in the randomized ELEMENT-MDS trial in patients with LR-MDS who have symptomatic anemia (hemoglobin ≤ 9.5 g/dL) but are RBC-TI and ESA-naïve (NCT05949684) [49].
Real-world data on the use of luspatercept are also emerging to allow treatment patterns in the clinical setting to be better understood. A recent analysis shows that the recommended dose-escalation schedule of luspatercept is underutilized which may limit its full therapeutic potential and reduce the proportion of patients achieving optimal responses [50]. Additional studies exploring initiating luspatercept at the maximum dose (1.75 mg/kg) have shown favorable results without new safety signals [51,52].

1.7. Imetelstat

Imetelstat is a first-in-class direct and competitive telomerase inhibitor approved for the treatment of patients with RBC-TD LR-MDS who have relapsed after, are refractory to, or are ineligible for ESAs [53]. Approval was based on results from the randomized phase III IMerge trial which showed significantly higher rates of durable RBC-TI at multiple time points, including 8 weeks, 24 weeks and 1 year, in the imetelstat group compared to placebo [54]. The benefit was observed across RS status and baseline transfusion requirements. Notably, results suggested that imetelstat carried disease-modifying activity, evidenced by cytogenetic responses, the reduction in malignant clonal populations, and correlation with longer durations of response. The impact of the perceived disease modification on survival will need to be seen with longer follow-up. More recent data shows that imetelstat retains clinical activity regardless of prior treatments for LR-MDS (including luspatercept) and prior response to ESA [55]. Although this analysis was limited by a small sample size, the findings inform drug sequencing practices in the evolving treatment landscape of LR-MDS. Imetelstat offers an alternative for patients who do not respond to or are ineligible for ESAs (EPO > 500 mU/mL) with the potential to delay progression and next-line of therapy. This mechanism sets it apart from other agents that primarily target erythropoiesis without addressing the underlying clonal dynamics of MDS.
RBD-TD is associated with reduced quality of life metrics and inferior clinical outcomes, particularly in those who develop secondary iron overload [26,56,57]. Iron overload can lead to end-organ damage if left untreated, including hepatic siderosis and iron-overload cardiomyopathy, which can further complicate prognosis and management. Iron chelation therapy plays an important role in mitigating these risks. Current treatment guidelines recommend starting iron chelation therapy for patients who have received more than 20 units of RBC transfusions and a ferritin level exceeding 2500 ng/mL, aiming for levels < 1000 ng/mL [20,27,58].

1.8. HMAs in LR-MDS

Treatment with HMAs earlier in the disease’s course has garnered growing interest due the potential to modify the natural history of MDS. Unlike traditional supportive care, HMAs such as azacitidine (AZA) and decitabine (DEC) target epigenetic dysregulation and promote hematological improvement. Prospective studies have shown that attenuated HMA dosing can lead to meaningful hematological responses, including improvement in anemia and transfusion requirements, and a favorable toxicity profile [59]. Patients with relapsed or refractory MDS, with ongoing RBC transfusion requirements, and with elevated EPO levels should be considered for a clinical trial or more intensive HMA-based treatments [20].
The availability of these newer therapies for LR-MDS introduces a welcome complexity in determining the optimal selection, sequencing, and combination of therapies. While ESAs remain an important option for patients with low EPO and the absence of RS, luspatercept in our opinion is the preferred first-line treatment for all patients with non-del5q LR-MDS with clinically significant isolated anemia (Figure 1). Other novel agents with different mechanisms of action that target anemia in LR-MDS are listed in Table 2. As the therapeutic landscape continues to evolve, ongoing studies and real-world evidence are critical to clarifying the most effective strategies for integrating these novel agents into the management of LR-MDS in clinical practice.

1.9. Treatment of Higher-Risk MDS

Despite the rapid expansion of treatment options for LR-MDS, HR-MDS continues to represent a significant unmet clinical need. The median OS for patients with IPSS-R high and very high risk is 1.6 and 0.8 years [15], respectively, with similar predictions under the IPSS-M (1.7 and 1.0 years, respectively) [16], and therefore the main goal of treatment in HR-MDS is to delay leukemic transformation and extend survival. Allo-HCT remains the only potentially curative treatment option; however, the associated morbidity and mortality risks limit patient eligibility, necessitating separate treatment approaches for transplant-eligible and -ineligible individuals (Figure 2).

1.10. Transplant Eligible Patients

HMA-based therapy is the standard of care in HR-MDS, ideally used as a bridging tool to allo-HCT. Individuals with significant frailty, comorbidities, poor performance status, and advanced age (>80 years) are generally considered ineligible for transplant [71]. Studies have demonstrated that patients with HR-MDS and a readily available suitable donor for allo-HCT experience better outcomes, supporting its strong consideration for all eligible patients regardless of age [72,73,74]. Detailed discussion regarding donor selection, conditioning, and transplant logistics are increasingly complex and beyond the scope of this review, and the reader is referred to excellent recent reviews on this subject [75]. For example, there are increasing data that haploidentical donors and mismatched unrelated donors can be used successfully in allogeneic transplantation for MDS patients when combined with post-transplantation cyclophosphamide [76,77]. We do, however, emphasize the need for the early referral of any potentially eligible patient with higher-risk MDS for transplantation consideration/consultation.
Intensive induction therapy as a bridge to transplant should be considered for patients with an elevated bone marrow blast count (≥10%) or lacking a suitable donor [71]. The recommendation for cytoreduction for ≥10% blasts is based on the positive correlation between higher blast count at the time of transplant and relapse risk [71,78,79]. Guidance on intensive induction for blasts between 5 and 9% is less clear. In practice, these patients are considered to have HR-MDS and are candidates for early transplant, but whether there is added benefit of intensive induction or HMA therapy to achieve morphological CR before allo-HCT is still debated. Schroeder et al. reported that patients who underwent immediate allo-HCT had a significantly higher 5-year OS probability compared to those who received pre-transplant therapy (68.5% vs. 37.4%, p = 0.023), with similar results for those with 5–9% blasts [80]. A large systematic review found that achieving post-induction CR before transplant did not correlate with longer OS [81]. Furthermore, a large cohort dataset of HMA-treated patients reported that achieving a composite CR, using the new International Working Group 2023 criteria [82], correlated with a superior OS (HR = 0.602, p < 0.001) only in patients who did not undergo transplantation [83]. These findings highlight the uncertainly regarding the role of bridging, and prospective trials are needed to determine which patients and disease features would benefit most from cytoreduction.
Post-transplant maintenance in MDS remains an area of active investigation. There is particular interest in patients with TP53 mutations who stand to gain the greatest benefit from novel approaches given their poor outcomes even after allo-HCT. In a large trial for patients with AML and MDS, there was no difference in relapse free survival (RFS) between maintenance subcutaneous AZA compared to placebo [84]. Low-dose DEC (5 mg/m2 days 1–5, 28-day cycle) plus recombinant human G-CSF was shown to reduce the incidence of AML relapse post-transplant in a randomized phase II trial of 204 patients with AML [85]. Notably, the study included only a small number of patients harboring TP53 mutations (n = 4). The randomized phase III AMADEUS trial (NCT04173533) [86] of oral AZA vs. placebo maintenance is currently underway after showing early safety and efficacy results [87]. Other maintenance approaches are being explored and are reported in Table 3.
Measurable residual disease (MRD) monitoring may be a more sensitive gauge of disease burden than morphology alone and should be routinely assessed and validated in prospective clinical trials [96]. Monitoring patient-specific MRD after transplant in MDS is challenging but has the potential to identify early relapse, predict transplant response, and select patients for post-transplant maintenance or pre-emptive therapies [97,98,99,100].

1.11. Transplant-Ineligible

HMAs are the cornerstone of front-line therapy for transplant ineligible patients since the approval of AZA in 2004 [20,101]. AZA, DEC, and oral decitabine-cedazuridine (C-DEC) are the only FDA-approved first-line agents for HR-MDS and form the foundation of many emerging combination strategies (Table 4). Despite their widespread use, HMAs offer limited OS benefit, highlighting the need for more effective treatment options.
The pivotal phase III AZA-001 trial demonstrated a superior OS of 24.5 months with AZA (75 mg/m2) compared to 15 months with best available therapy [115]. The results of this study likely overestimate survival outcomes, as real-world data have not consistently replicated such a robust response with a median OS up to around 18 months [116,117]. DEC was evaluated in a randomized phase III trial which showed a significantly superior overall response rate (ORR) and statistically non-significant trends towards longer time to AML progression and OS [118]. Approximately 20% of patients who receive HMA and reach a stable disease response can accomplish a better response at a later timepoint during treatment [119]. In practice, patients should continue therapy until disease progression or unacceptable toxicity or they are on the path to allo-HCT. Recently, oral C-DEC showed equivalent pharmacological features and efficacy as intravenous decitabine, and a median OS of 31.8 months [120,121]. This study lacked a comparator arm, limiting our ability to assess clinical superiority to intravenous DEC.
Although most patients respond within the initial 4–6 cycles of HMA (efficacy assessment), unfortunately most will ultimately relapse; therefore, newly diagnosed patients with HR-MDS should be strongly considered for a clinical trial whenever possible.

1.12. Early Promise and Later Disappointments: HMA Combinations in HR-MDS

Multiple novel HMA combination strategies have failed to yield successful results in large clinical trials despite promising early results.
The combination of AZA with the histone deacetylase inhibitor vorinostat suggested high response rates in early-phase studies [122]. However, the randomized phase II trial (SWOG S1117) found no OS benefit and a higher drug discontinuation rate in the vorinostat plus AZA arm [123]. Similarly, AZA and pevonedistat, a NEDD8-activating enzyme inhibitor, showed synergy with AZA in preclinical models, and early studies showed favorable outcomes [124]. The subsequent phase III PANTHER trial showed no significant improvement in event-free survival (EFS) or OS with the combination [106]. Magrolimab, an anti-CD47 macrophage checkpoint inhibitor, also showed encouraging results and high response rates [125] The following phase III ENHANCE trial was terminated early as an interim analysis showed no OS advantage [126]. Lastly, the later phase trials of AZA plus sabatolimab (STIMULUS-MDS1 and 2), an anti-T-cell receptor T-cell immunoglobulin and mucin domain-3 (TIM-3) antibody, failed to meet their primary endpoint despite early promise [108,127,128]. These results underscore the heterogeneity of HR-MDS and the challenges of improving survival in this population. Novel combination trials are ongoing and anticipated to help address the significant unmet need in this highly heterogenous and complex patient population.
Venetoclax, a potent small molecule BCL-2 inhibitor, reshaped the treatment paradigm for older or unfit patients with AML. The landmark VIALE-A trial demonstrated a significant improvement in median OS, nearly doubling survival with the addition of venetoclax to AZA when compared to AZA alone (14.7 versus 9.6 months) [129]. Stemming from the success of this combination in AML and the observed synergy in myeloid malignancies [130], several studies have emerged in MDS [131,132].
In the front-line setting, real-world data showed the efficacy of the AZA/venetoclax combination for patients with HR-MDS, with an emphasis on better outcomes for patients who are able to reach transplant [133]. One recent study demonstrated superior CR rates with HMA/venetoclax compared to HMA alone (33% vs. 12%, p < 0.001) but found no difference in OS [134]. A phase 1b study published in 2025 by Garcia et al. reported that 30% of patients with treatment-naïve HR-MDS reached a CR as best response with AZA/venetoclax and had a median OS of 26 months, which was similar among patients who achieved CR and mCR with HI [135]. These results are encouraging and superior to historical responses with AZA monotherapy. Additionally, nearly 40% were able to proceed to allo-HCT and had a median OS not reached (NR, 95% CI: 40.1-NR). Unsurprisingly, survival was shorter in the TP53-mutated population (11.2 months). Longer follow-up is needed to confirm if patients have experienced prolonged responses and a survival benefit. The highly anticipated global phase III randomized VERONA trial aimed to assess the efficacy and safety of AZA/venetoclax (AZA 75 mg/m2 IV or SC; venetoclax 400 mg daily for 14 days; 28-day cycles) versus AZA monotherapy in treatment-naïve HR-MDS [136]. The trial did not meet its primary endpoint of OS (HR 0.908, p = 0.3772) according to a recent press release, and further details are awaited to fully interpret these findings and their clinical implications [102]. Clinical and molecular data, rates of allo-HCT, and subsequent therapies will be important for interpreting the study outcomes and assessing the full clinical impact of the AZA/venetoclax combination. The combination of AZA/venetoclax is increasingly used in clinical practice for transplant-ineligible individuals; however, key questions remain regarding patient selection and optimal dosing and the scheduling of therapy. These uncertainties highlight the need for additional studies to better define effective consolidation and maintenance strategies in this setting.
In addition, the combination of C-DEC and venetoclax was also recently explored, and after a median follow-up of 10.8 (IQR 5.6–16.4) months, 37 of 39 patients responded, and nearly half underwent allo-HCT, highlighting the potential role of these regimens as a bridge to transplant [137].
It is important to note that venetoclax is associated with significant and prolonged myelosuppression, which in clinical practice can lead to treatment delays, dose reductions, and life-threatening bleeding and infectious complications. Patients who are on the path to transplant could be considered for this intensive approach to reduce blast count pre-transplant given the reported rapid responses observed with this combination [131].

1.13. IDH1/2 Inhibitors

IDH1/2 mutations are present in approximately 5% of patients with MDS and are associated with a higher progression to AML [138].
The IDH1 inhibitor, ivosidenib (IVO), is currently approved for relapsed/refractory IDH1-mutated MDS based on the AG120-C-001 MDS sub-study results which showed CR rates of nearly 40% and a median OS of 35.7 months [113]. The IDIOME study, a single arm phase II by the GFM, demonstrated favorable results with IVO [139,140]. Among patients with previous HMA failure, the study reported an ORR of 63.6%, and a median OS of 8.9 months. In treatment-naïve HR-MDS, the addition of AZA to IVO after three cycles led to an ORR of 78.3%, with a median OS not reached after 25.2 months of follow-up. The global PyramIDH phase III randomized trial comparing single-agent HMA and single-agent IVO in the front-line setting is currently underway (NCT06465953) [141].
The IDH1 inhibitor olutasidenib is currently approved for the treatment of relapsed/refractory IDH1-mutated AML [142]. Olutasidenib led to durable remissions in patients with HR-MDS (n = 22) in a multicenter, open-label, phase I/II trial [143] and continues to be explored in combination with HMAs in an ongoing phase II trial (NCT06597734) [144].
Enasidenib (ENA), an oral selective IDH2 inhibitor, is approved for the treatment of relapsed or refractory IDH2-mutated AML and has demonstrated promising results in MDS. An open-label phase I trial conducted by Stein et al. reported an ORR of 53% (nine of 17 patients). A favorable response was also seen in six of 13 patients who were previously treated with HMA (ORR 46%) [145]. A subsequent phase II trial, which included 50 patients with MDS, evaluated the safety and efficacy of AZA plus ENA combination in a treatment-naïve cohort and ENA monotherapy for patients pre-treated with HMA. The majority of patients treated with the combination achieved a response (ORR 74%) with a median time to best response of 1.3 (0.9–3.8) months, whereas 52% reached stable disease with ENA monotherapy (ORR 35%) and experienced a slower median time to best response of 4.6 (2.7–7.6) months [146].

1.14. TP53-Mutated MDS

Mutations in TP53 leads to a loss of the normal tumor suppressive functions and promotes genomic instability and disease progression. In MDS, these mutations identify a very high-risk subgroup within HR-MDS characterized by an aggressive disease biology and resistance to conventional therapies. Patients with MDS with TP53 mutations, particularly those with bi-allelic inactivation, have a notably poor prognosis and are at high risk for AML transformation [147]. Responses to conventional HMA and HMA-based therapies are often brief, making allo-HCT the preferred strategy when feasible.
Despite its urgent clinical need, TP53-mutated MDS has remained largely refractory to therapeutic advances. APR-246 (eprenetapopt), a first-in class small molecule designed to restore wild-type p53 function, initially generated excitement based on early-phase data suggesting responses when combined with AZA [148,149]. However, enthusiasm was tempered by the results of the pivotal phase III trial which failed to meet its primary endpoint of CR when comparing AZA plus APR-246 to AZA monotherapy [150]. Similarly, the phase Ib magrolimab plus AZA study included 25 patients (26%) with a TP53 mutation, of whom 10 achieved CR and a median OS of 16.3 months (95% CI, 1.8-NR) [125]. Nevertheless, subsequent data failed to show a clinical benefit in this high-risk population [126]. The phase Ib sabatolimab plus HMA trial reported an ORR of 71.4% (95% CI, 41.9–91.6) among 10 of 14 TP53-mutated HR-MDS patients (four of whom reached CR, 28.6%) [128]. However, the phase III STIMULUS-MDS2 trial failed to meet its primary endpoint of OS [127].
Effective and durable therapies for TP53-mutated MDS remain an urgent unmet need. Emergent treatment approaches and clinical trial enrollment should be strongly recommended for this subgroup of individuals with high-risk MDS biology.

2. Conclusions

The current management of MDS is dynamic and rapidly evolving, driven by advances in molecular diagnostics, refined classification systems, and the promise of targeted therapies. The approach to treatment is risk-adapted and biologically informed, aiming to match therapeutic intensity with disease severity, molecular profile, and individual patient goals. Given the complexity of MDS and its treatment landscape, a multidisciplinary and collaborative approach, ideally involving specialized pathologists and hematologists at centers with expertise in myeloid malignancies, is essential to deliver optimal care, provide access to emerging therapies, and facilitate participation in clinical trials. Continued collaborative research remains critical to translating the biological complexity of MDS into durable and meaningful clinical benefit.

Funding

This research received no external funding.

Acknowledgments

J.M.S. is supported by the National Cancer Institute of the National Institutes of Health under Award Number T32CA233414. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Conflicts of Interest

J.M.S.: participated in advisory board for Sobi. T.K. has no conflicts to disclose. A.M.Z. participated in advisory boards, consulted, participated in clinical trial committees, and/or received honoraria from AbbVie, Akesobio, Agios, Amgen, Astellas, BioCryst, Beigene, Boehringer-Ingelheim, Celgene/BMS, Chiesi/Cornerstone biopharma, Daiichi Sankyo, Dr Reddy, Epizyme, Faron, Fibrogen, GSK, Glycomimetics, Genentech, Gilead, Geron, Janssen, Jasper, Karyopharm, Kyowa Kirin, Keros, Kura, Novartis, Notable, Orum, Otsuka, Pfizer, Regeneron, Rigel, Seattle Genetics, Shattuck labs, Schrodinger, Syros, Syndax, Servier, Takeda, Treadwell, Taiho, Vincerx, and Zentalis.

References

  1. SEER*Explorer: An Interactive Website for SEER Cancer Statistics [Internet]. Surveillance Research Program, National Cancer Institute. 17 April 2024. [Updated: 5 November 2024]. Available online: https://seer.cancer.gov/statistics-network/explorer/ (accessed on 19 February 2025).
  2. Ma, X. Epidemiology of Myelodysplastic Syndromes. Am. J. Med. 2012, 125, S2–S5. [Google Scholar] [CrossRef] [PubMed]
  3. Kouroukli, O.; Symeonidis, A.; Foukas, P.; Maragkou, M.-K.; Kourea, E.P. Bone Marrow Immune Microenvironment in Myelodysplastic Syndromes. Cancers 2022, 14, 5656. [Google Scholar] [CrossRef] [PubMed]
  4. Li, X.; Zou, C.; Xiang, X.; Zhao, L.; Chen, M.; Yang, C.; Wu, Y. Myelodysplastic Neoplasms (MDS): Pathogenesis and Therapeutic Prospects. Biomolecules 2025, 15, 761. [Google Scholar] [CrossRef] [PubMed]
  5. Arber, D.A.; Orazi, A.; Hasserjian, R.P.; Borowitz, M.J.; Calvo, K.R.; Kvasnicka, H.-M.; Wang, S.A.; Bagg, A.; Barbui, T.; Branford, S.; et al. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: Integrating morphologic, clinical, and genomic data. Blood 2022, 140, 1200–1228. [Google Scholar] [CrossRef] [PubMed]
  6. Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F.; Bejar, R.; Berti, E.; Busque, L.; Chan, J.K.C.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719. [Google Scholar] [CrossRef] [PubMed]
  7. Bennett, J.M.; Catovsky, D.; Daniel, M.T.; Flandrin, G.; Galton, D.A.; Gralnick, H.R.; Sultan, C. Proposals for the classification of the myelodysplastic syndromes. Br. J. Haematol. 1982, 51, 189–199. [Google Scholar] [CrossRef] [PubMed]
  8. Bernard, E.; Hasserjian, R.P.; Greenberg, P.L.; Arango Ossa, J.E.; Creignou, M.; Tuechler, H.; Gutierrez-Abril, J.; Domenico, D.; Medina-Martinez, J.S.; Levine, M.; et al. Molecular taxonomy of myelodysplastic syndromes and its clinical implications. Blood 2024, 144, 1617–1632. [Google Scholar] [CrossRef] [PubMed]
  9. Kewan, T.; Durmaz, A.; Bahaj, W.; Gurnari, C.; Terkawi, L.; Awada, H.; Ogbue, O.D.; Ahmed, R.; Pagliuca, S.; Awada, H.; et al. Molecular patterns identify distinct subclasses of myeloid neoplasia. Nat. Commun. 2023, 14, 3136. [Google Scholar] [CrossRef] [PubMed]
  10. Komrokji, R.S.; Lanino, L.; Ball, S.; Bewersdorf, J.P.; Marchetti, M.; Maggioni, G.; Travaglino, E.; Al Ali, N.H.; Fenaux, P.; Platzbecker, U.; et al. Data-driven, harmonised classification system for myelodysplastic syndromes: A consensus paper from the International Consortium for Myelodysplastic Syndromes. Lancet Haematol. 2024, 11, e862–e872. [Google Scholar] [CrossRef] [PubMed]
  11. Döhner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Büchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [PubMed]
  12. Dohner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022, 140, 1345–1377. [Google Scholar] [CrossRef] [PubMed]
  13. Stahl, M.; Bewersdorf, J.P.; Xie, Z.; Porta, M.G.D.; Komrokji, R.; Xu, M.L.; Abdel-Wahab, O.; Taylor, J.; Steensma, D.P.; Starczynowski, D.T.; et al. Classification, risk stratification and response assessment in myelodysplastic syndromes/neoplasms (MDS): A state-of-the-art report on behalf of the International Consortium for MDS (icMDS). Blood Rev. 2023, 62, 101128. [Google Scholar] [CrossRef] [PubMed]
  14. Greenberg, P.; Cox, C.; LeBeau, M.M.; Fenaux, P.; Morel, P.; Sanz, G.; Sanz, M.; Vallespi, T.; Hamblin, T.; Oscier, D.; et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 1997, 89, 2079–2088. [Google Scholar] [CrossRef] [PubMed]
  15. Greenberg, P.L.; Tuechler, H.; Schanz, J.; Sanz, G.; Garcia-Manero, G.; Solé, F.; Bennett, J.M.; Bowen, D.; Fenaux, P.; Dreyfus, F.; et al. Revised International Prognostic Scoring System for Myelodysplastic Syndromes. Blood 2012, 120, 2454–2465. [Google Scholar] [CrossRef] [PubMed]
  16. Bernard, E.; Tuechler, H.; Greenberg, P.L.; Hasserjian, R.P.; Arango Ossa, J.E.; Nannya, Y.; Devlin, S.M.; Creignou, M.; Pinel, P.; Monnier, L.; et al. Molecular International Prognostic Scoring System for Myelodysplastic Syndromes. NEJM Evid. 2022, 1, EVIDoa2200008. [Google Scholar] [CrossRef] [PubMed]
  17. Kewan, T.; Bewersdorf, J.P.; Blaha, O.; Stahl, M.; Al Ali, N.H.; DeZern, A.E.; Sekeres, M.A.; Carraway, H.E. Validation of the Molecular International Prognostic Scoring System (IPSS-M) in Patients (Pts) with Myelodysplastic Syndromes/Neoplasms (MDS) Who Were Treated with Hypomethylating Agents (HMA). Blood 2023, 142 (Suppl. S1), 4980. [Google Scholar] [CrossRef]
  18. Gurnari, C.; Gagelmann, N.; Badbaran, A.; Awada, H.; Dima, D.; Pagliuca, S.; D’Aveni-Piney, M.; Attardi, E.; Voso, M.T.; Cerretti, R.; et al. Outcome prediction in myelodysplastic neoplasm undergoing hematopoietic cell transplant in the molecular era of IPSS-M. Leukemia 2023, 37, 717–719. [Google Scholar] [CrossRef] [PubMed]
  19. Stahl, M.; Deveaux, M.; De Witte, T.; Neukirchen, J.; Sekeres, M.A.; Brunner, A.M.; Roboz, G.J.; Steensma, D.P.; Bhatt, V.R.; Platzbecker, U.; et al. The use of immunosuppressive therapy in MDS: Clinical outcomes and their predictors in a large international patient cohort. Blood Adv. 2018, 2, 1765–1772. [Google Scholar] [CrossRef] [PubMed]
  20. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines. Myelodysplastic Syndromes (Version 1.2025). Available online: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1446 (accessed on 10 January 2025).
  21. Oliva, E.N.; Riva, M.; Niscola, P.; Santini, V.; Breccia, M.; Giai, V.; Poloni, A.; Patriarca, A.; Crisà, E.; Capodanno, I.; et al. Eltrombopag for Low-Risk Myelodysplastic Syndromes with Thrombocytopenia: Interim Results of a Phase II, Randomized, Placebo-Controlled Clinical Trial (EQOL-MDS). J. Clin. Oncol. 2023, 41, 4486–4496. [Google Scholar] [CrossRef] [PubMed]
  22. Fenaux, P.; Muus, P.; Kantarjian, H.; Lyons, R.M.; Larson, R.A.; Sekeres, M.A.; Becker, P.S.; Orejudos, A.; Franklin, J. Romiplostim monotherapy in thrombocytopenic patients with myelodysplastic syndromes: Long-term safety and efficacy. Br. J. Haematol. 2017, 178, 906–913. [Google Scholar] [CrossRef] [PubMed]
  23. Desmond, R.; Townsley, D.M.; Dumitriu, B.; Olnes, M.J.; Scheinberg, P.; Bevans, M.; Parikh, A.R.; Broder, K.; Calvo, K.R.; Wu, C.O.; et al. Eltrombopag restores trilineage hematopoiesis in refractory severe aplastic anemia that can be sustained on discontinuation of drug. Blood 2014, 123, 1818–1825. [Google Scholar] [CrossRef] [PubMed]
  24. Dickinson, M.; Cherif, H.; Fenaux, P.; Mittelman, M.; Verma, A.; Portella, M.S.O.; Burgess, P.; Ramos, P.M.; Choi, J.; Platzbecker, U. Azacitidine with or without eltrombopag for first-line treatment of intermediate- or high-risk MDS with thrombocytopenia. Blood 2018, 132, 2629–2638. [Google Scholar] [CrossRef] [PubMed]
  25. Abel, G.A.; Hebert, D.; Lee, C.; Rollison, D.; Gillis, N.; Komrokji, R.; Foran, J.M.; Liu, J.J.; Al Baghdadi, T.; Deeg, J.; et al. Health-related quality of life and vulnerability among people with myelodysplastic syndromes: A US national study. Blood Adv. 2023, 7, 3506–3515. [Google Scholar] [CrossRef] [PubMed]
  26. De Swart, L.; Crouch, S.; Hoeks, M.; Smith, A.; Langemeijer, S.; Fenaux, P.; Symeonidis, A.; Cermâk, J.; Hellström-Lindberg, E.; Stauder, R.; et al. Impact of red blood cell transfusion dose density on progression-free survival in patients with lower-risk myelodysplastic syndromes. Haematologica 2020, 105, 632–639. [Google Scholar] [CrossRef] [PubMed]
  27. Stempel, J.M.; Podoltsev, N.A.; Dosani, T. Supportive Care for Patients with Myelodysplastic Syndromes. Cancer J. 2023, 29, 168–178. [Google Scholar] [CrossRef] [PubMed]
  28. DeFilipp, Z.; Ciurea, S.O.; Cutler, C.; Robin, M.; Warlick, E.D.; Nakamura, R.; Brunner, A.M.; Dholaria, B.; Walker, A.R.; Kroger, N.; et al. Hematopoietic Cell Transplantation in the Management of Myelodysplastic Syndrome: An Evidence-Based Review from the American Society for Transplantation and Cellular Therapy Committee on Practice Guidelines. Transplant. Cell. Ther. 2023, 29, 71–81. [Google Scholar] [CrossRef] [PubMed]
  29. Cutler, C.S. A decision analysis of allogeneic bone marrow transplantation for the myelodysplastic syndromes: Delayed transplantation for low-risk myelodysplasia is associated with improved outcome. Blood 2004, 104, 579–585. [Google Scholar] [CrossRef] [PubMed]
  30. Gurnari, C.; Robin, M.; Godley, L.A.; Drozd-Sokolowska, J.; Wlodarski, M.W.; Raj, K.; Onida, F.; Worel, N.; Ciceri, F.; Carbacioglu, S.; et al. Germline predisposition traits in allogeneic hematopoietic stem-cell transplantation for myelodysplastic syndromes: A survey-based study and position paper on behalf of the Chronic Malignancies Working Party of the EBMT. Lancet Haematol. 2023, 10, e994–e1005. [Google Scholar] [CrossRef] [PubMed]
  31. Xie, Z.; Komrokji, R.; Al Ali, N.; Regelson, A.; Geyer, S.; Patel, A.; Saygin, C.; Zeidan, A.M.; Bewersdorf, J.P.; Mendez, L.; et al. Risk prediction for clonal cytopenia: Multicenter real-world evidence. Blood 2024, 144, 2033–2044. [Google Scholar] [CrossRef] [PubMed]
  32. Weeks, L.D.; Niroula, A.; Neuberg, D.; Wong, W.; Lindsley, R.C.; Luskin, M.R.; Berliner, N.; Stone, R.M.; Deangelo, D.J.; Soiffer, R.J.; et al. Prediction of Risk for Myeloid Malignancy in Clonal Hematopoiesis. NEJM Evid. 2023, 2, EVIDoa2200310. [Google Scholar] [CrossRef] [PubMed]
  33. List, A.; Dewald, G.; Bennett, J.; Giagounidis, A.; Raza, A.; Feldman, E.; Powell, B.; Greenberg, P.; Thomas, D.; Stone, R.; et al. Lenalidomide in the Myelodysplastic Syndrome with Chromosome 5q Deletion. N. Engl. J. Med. 2006, 355, 1456–1465. [Google Scholar] [CrossRef] [PubMed]
  34. Cheson, B.D.; Bennett, J.M.; Kantarjian, H.; Pinto, A.; Schiffer, C.A.; Nimer, S.D.; Löwenberg, B.; Beran, M.; de Witte, T.M.; Stone, R.M. Report of an international working group to standardize response criteriafor myelodysplastic syndromes. Blood 2000, 96, 3671–3674. [Google Scholar] [CrossRef] [PubMed]
  35. Fenaux, P.; Giagounidis, A.; Selleslag, D.; Beyne-Rauzy, O.; Mufti, G.; Mittelman, M.; Muus, P.; Te Boekhorst, P.; Sanz, G.; Del Canizo, C.; et al. A randomized phase 3 study of lenalidomide versus placebo in RBC transfusion-dependent patients with Low-/Intermediate-1-risk myelodysplastic syndromes with del5q. Blood 2011, 118, 3765–3776. [Google Scholar] [CrossRef] [PubMed]
  36. Díez-Campelo, M.; López-Cadenas, F.; Xicoy, B.; Lumbreras, E.; González, T.; Del Rey González, M.; Sánchez-García, J.; Coll Jordà, R.; Slama, B.; Hernández-Rivas, J.-Á.; et al. Low dose lenalidomide versus placebo in non-transfusion dependent patients with low risk, del(5q) myelodysplastic syndromes (SintraREV): A randomised, double-blind, phase 3 trial. Lancet Haematol. 2024, 11, e659–e670. [Google Scholar] [CrossRef] [PubMed]
  37. Park, S.; Kelaidi, C.; Meunier, M.; Casadevall, N.; Gerds, A.T.; Platzbecker, U. The prognostic value of serum erythropoietin in patients with lower-risk myelodysplastic syndromes: A review of the literature and expert opinion. Ann. Hematol. 2020, 99, 7–19. [Google Scholar] [CrossRef] [PubMed]
  38. Hellström-Lindberg, E.; Negrin, R.; Stein, R.; Krantz, S.; Lindberg, G.; Vardiman, J.; Öst, Å.; Greenberg, P. Erythroid response to treatment with G-CSF plus erythropoietin for the anaemia of patients with myelodysplastic syndromes: Proposal for a predictive model. Br. J. Haematol. 1997, 99, 344–351. [Google Scholar] [CrossRef] [PubMed]
  39. Park, S.; Grabar, S.; Kelaidi, C.; Beyne-Rauzy, O.; Picard, F.; Bardet, V.; Coiteux, V.; Leroux, G.; Lepelley, P.; Daniel, M.T.; et al. Predictive factors of response and survival in myelodysplastic syndrome treated with erythropoietin and G-CSF: The GFM experience. Blood 2008, 111, 574–582. [Google Scholar] [CrossRef] [PubMed]
  40. Zhou, L.; Nguyen, A.N.; Sohal, D.; Ying Ma, J.; Pahanish, P.; Gundabolu, K.; Hayman, J.; Chubak, A.; Mo, Y.; Bhagat, T.D.; et al. Inhibition of the TGF-β receptor I kinase promotes hematopoiesis in MDS. Blood 2008, 112, 3434–3443. [Google Scholar] [CrossRef] [PubMed]
  41. Celgene Coorporation, Bristol-Myers Squibb Company. REBLOZYL® (Lusparercept-Aamt) for Injection, for Subcutaneous Use; Celgene Coorporation, Bristol-Myers Squibb Company: Summit, NY, USA, 2025. [Google Scholar]
  42. Platzbecker, U.; Germing, U.; Gotze, K.S.; Kiewe, P.; Mayer, K.; Chromik, J.; Radsak, M.; Wolff, T.; Zhang, X.; Laadem, A.; et al. 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. 2017, 18, 1338–1347. [Google Scholar] [CrossRef] [PubMed]
  43. Platzbecker, U.; Gotze, K.S.; Kiewe, P.; Germing, U.; Mayer, K.; Radsak, M.; Wolff, T.; Chromik, J.; Sockel, K.; Oelschlagel, U.; et al. Long-Term Efficacy and Safety of Luspatercept for Anemia Treatment in Patients with Lower-Risk Myelodysplastic Syndromes: The Phase II PACE-MDS Study. J. Clin. Oncol. 2022, 40, 3800–3807. [Google Scholar] [CrossRef] [PubMed]
  44. Fenaux, P.; Platzbecker, U.; Mufti, G.J.; Garcia-Manero, G.; Buckstein, R.; Santini, V.; Díez-Campelo, M.; Finelli, C.; Cazzola, M.; Ilhan, O.; et al. Luspatercept in Patients with Lower-Risk Myelodysplastic Syndromes. N. Engl. J. Med. 2020, 382, 140–151. [Google Scholar] [CrossRef] [PubMed]
  45. Platzbecker, U.; Della Porta, M.G.; Santini, V.; Zeidan, A.M.; Komrokji, R.S.; Shortt, J.; Valcarcel, D.; Jonasova, A.; Dimicoli-Salazar, S.; Tiong, I.S.; et al. Efficacy and safety of luspatercept versus epoetin alfa in erythropoiesis-stimulating agent-naive, transfusion-dependent, lower-risk myelodysplastic syndromes (COMMANDS): Interim analysis of a phase 3, open-label, randomised controlled trial. Lancet 2023, 402, 373–385. [Google Scholar] [CrossRef] [PubMed]
  46. Oliva, E.N.; Platzbecker, U.; Della Porta, M.G.; Garcia-Manero, G.; Santini, V.; Fenaux, P.; Shortt, J.; Komrokji, R.S.; Pelligra, C.G.; Guo, S.; et al. Health-Related Quality of Life of Luspatercept Versus Epoetin Alfa in Red Blood Cell Transfusion-Dependent Lower-Risk Myelodysplastic Syndromes: Results from the Final Datacut of the Phase 3 COMMANDS Study. Blood 2024, 144, 3216. [Google Scholar] [CrossRef]
  47. Garcia-Manero, G.; Della Porta, M.G.; Zeidan, A.M.; Komrokji, R.; Pozharskaya, V.; Rose, S.; Keeperman, K.; Lai, Y.; Kalsekar, S.; Aggarwal, B.; et al. Overall survival (OS) and duration of response for transfusion independence (TI) in erythropoiesis stimulating agent (ESA)–naive patients (pts) with very low-, low-, or intermediate-risk myelodysplastic syndromes (MDS) treated with luspatercept (LUSPA) vs epoetin alfa (EA) in the COMMANDS trial. J. Clin. Oncol. 2025, 43, 6512. [Google Scholar] [CrossRef]
  48. Garcia-Manero, G.; Santini, V.; Zeidan, A.M.; Komrokji, R.S.; Pozharskaya, V.; Rose, S.; Keeperman, K.; Lai, Y.; Kalsekar, S.; Aggarwal, B.; et al. Long-Term Transfusion Independence with Luspatercept Versus Epoetin Alfa in Erythropoiesis-Stimulating Agent-Naive, Lower-Risk Myelodysplastic Syndromes in the COMMANDS Trial. Adv. Ther. 2025, 42, 3576–3589. [Google Scholar] [CrossRef] [PubMed]
  49. Zeidan, A.M.; Komrokji, R.S.; Buckstein, R.; Santini, V.; Rose, S.; Malini, P.; Lew, G.; Aggarwal, D.; Keeperman, K.L.; Jiang, H.; et al. The ELEMENT-MDS Trial: A Phase 3 Randomized Study Evaluating Luspatercept Versus Epoetin Alfa in Erythropoiesis-Stimulating Agent-Naive, Non-Transfusion-Dependent, Lower-Risk Myelodysplastic Syndromes. Blood 2023, 142, 6503. [Google Scholar] [CrossRef]
  50. Mukherjee, S.; Brown-Bickerstaff, C.; Falkenstein, A.; Makinde, A.Y.; Bland, E.; Laney, J.; Garretson, M.; Huggar, D.; McBride, A. Treatment patterns and outcomes with luspatercept in patients with lower-risk myelodysplastic syndromes: A retrospective US cohort analysis. HemaSphere 2024, 8, e38. [Google Scholar] [CrossRef] [PubMed]
  51. Götze, K.S.S.; Hecht, A.; Kubasch, A.S.; Diez-Campelo, M.; Stüssi, G.; Valcarcel, D.; Font Lopez, P.; Mora Castera, E.; Xicoy, B.; Bernal Del Castillo, T.; et al. Preliminary Safety and Efficacy of Luspatercept Initiated at Maximum Approved Dose in Patients with Transfusion Dependent Anemia Due to Very Low, Low and Intermediate Risk MDS with Ring Sideroblasts: First Results of the Lusplus Trial. Blood 2024, 144, 3207. [Google Scholar] [CrossRef]
  52. Della Porta, M.G.; Diez-Campelo, M.; Santini, V.; Buckstein, R.; Ades, L.; Sahagun, L.; Das, G.; Bryant, T.; Zelinsky, T.; Lai, Y.; et al. Preliminary efficacy and safety analysis of the MAXILUS study of luspatercept initiated at the maximum approved dose in transfusion-dependent lower-risk myelodysplastic syndromes. HemaSphere 2025, 9, PF634. [Google Scholar]
  53. RYTELO® (Imetelstat) for Injection, for Intravenous Use; Geron Coorporation: Foster City, CA, USA, 2025.
  54. Platzbecker, U.; Santini, V.; Fenaux, P.; Sekeres, M.A.; Savona, M.R.; Madanat, Y.F.; Díez-Campelo, M.; Valcárcel, D.; Illmer, T.; Jonášová, A.; et al. Imetelstat in patients with lower-risk myelodysplastic syndromes who have relapsed or are refractory to erythropoiesis-stimulating agents (IMerge): A multinational, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2024, 403, 249–260. [Google Scholar] [CrossRef] [PubMed]
  55. Platzbecker, U.; Santini, V.; Zeidan, A.M.; Sekeres, M.A.; Fenaux, P.; Raza, A.; Mittelman, M.; Thépot, S.; Buckstein, R.; Germing, U.; et al. Effect of Prior Treatments on the Clinical Activity of Imetelstat in Transfusion-Dependent Patients with Erythropoiesis-Stimulating Agent, Relapsed or Refractory/Ineligible Lower-Risk Myelodysplastic Syndromes. Blood 2024, 144, 352. [Google Scholar] [CrossRef]
  56. Malcovati, L.; Porta, M.G.D.; Pascutto, C.; Invernizzi, R.; Boni, M.; Travaglino, E.; Passamonti, F.; Arcaini, L.; Maffioli, M.; Bernasconi, P.; et al. Prognostic Factors and Life Expectancy in Myelodysplastic Syndromes Classified According to WHO Criteria: A Basis for Clinical Decision Making. J. Clin. Oncol. 2005, 23, 7594–7603. [Google Scholar] [CrossRef] [PubMed]
  57. Stauder, R.; Yu, G.; Koinig, K.A.; Bagguley, T.; Fenaux, P.; Symeonidis, A.; Sanz, G.; Cermak, J.; Mittelman, M.; Hellström-Lindberg, E.; et al. Health-related quality of life in lower-risk MDS patients compared with age- and sex-matched reference populations: A European LeukemiaNet study. Leukemia 2018, 32, 1380–1392. [Google Scholar] [CrossRef] [PubMed]
  58. Yang, Y.; Tang, Z.; An, T.; Zhao, L. The impact of iron chelation therapy on patients with lower/intermediate IPSS MDS and the prognostic role of elevated serum ferritin in patients with MDS and AML: A meta-analysis. Medicine 2019, 98, e17406. [Google Scholar] [CrossRef] [PubMed]
  59. Sasaki, K.; Jabbour, E.; Montalban-Bravo, G.; Darbaniyan, F.; Do, K.-A.; Class, C.; Short, N.J.; Kanagal-Shamana, R.; Kadia, T.; Borthakur, G.; et al. Low-Dose Decitabine versus Low-Dose Azacitidine in Lower-Risk MDS. NEJM Evid. 2022, 1, EVIDoa2200034. [Google Scholar] [CrossRef] [PubMed]
  60. Diez-Campelo, M.; Ross, D.M.; Giagounidis, A.; Tan, S.; Cluzeau, T.; Chee, L.C.Y.; Valcarcel, D.; Arnan, M.; Graham, C.; McGinty, A.; et al. Durable Clinical Benefit with Ker-050 Treatment: Findings from an Ongoing Phase 2 Study in Participants with Lower-Risk MDS. Blood 2023, 142, 196. [Google Scholar] [CrossRef]
  61. Tan, S.; Arnan, M.; Chee, L.C.Y.; Cluzeau, T.; Diez-Campelo, M.; Giagounidis, A.; Hiwase, D.; Ross, D.M.; Sekeres, M.A.; Valcarcel, D.; et al. Hematologic Improvement and Fatigue Reduction with Elritercept (KER-050) in Participants with Lower-Risk (LR) Myelodysplastic Neoplasms (MDS) with Non-Transfusion Dependent Anemia: New Analyses from an Ongoing Phase 2 Trial. Blood 2024, 144, 4591. [Google Scholar] [CrossRef]
  62. RVU120 for Treatment of Anemia in Patients with Lower-Risk Myelodysplastic Neoplasms (MDS), ClinicalTrials.gov ID NCT06243458. Available online: https://www.clinicaltrials.gov/study/NCT06243458?term=NCT06243458&rank=1 (accessed on 11 June 2025).
  63. Rodriguez Sevilla, J.J.; Adema, V.; Chien, K.S.; Ganan-Gomez, I.; Montalban-Bravo, G.; Urrutia, S.; Joseph, J.; Yang, H.; Borthakur, G.; Short, N.J.; et al. A Phase 2 Study of Canakinumab in Patients with Lower-Risk Myelodysplastic Syndromes or Chronic Myelomonocytic Leukemia. Blood 2023, 142, 1866. [Google Scholar] [CrossRef]
  64. Bouligny, I.; Carraway, H.E.; Sekeres, M.; Dezern, A.; Komrokji, R.; Stone, R.M.; Roboz, G.J.; Montalban-Bravo, G.; Bataller, A.; Sasaki, K.; et al. A randomized, multicenter phase II trial of 3-day decitabine, 3-day azacitidine, or 5-day azacitidine in lower-risk myelodysplastic syndrome. HemaSphere 2025, 9, PF629. [Google Scholar]
  65. Ades, L.; Cluzeau, T.; Comont, T.; Aguinaga, L.; Stamatoullas, A.; Meunier, M.; Gyan, E.; Garnier, A.; D’Aveni, M.; Thépot, S.; et al. Combining ESA and Luspatercept in Non-RS MDS Patients Having Failed ESA—Results of the Phase 1-2 Part a of the GFM Combola Study. Blood 2024, 144, 351. [Google Scholar] [CrossRef]
  66. Garcia-Manero, G.; Bachiashvili, K.; Griffiths, E.A.; Zeidan, A.M.; Traer, E.; Saini, L.; Amin, H.; Mohan, S.R.; Lubbert, M.; Maness-Harris, L.; et al. Randomized Phase 1-2 Study to Assess Safety and Efficacy of Low-Dose (LD) Oral Decitabine/Cedazuridine (ASTX727) in Lower-Risk Myelodysplastic Syndromes (LR-MDS) Patients: Interim Safety Analysis. Clin. Lymphoma Myeloma Leuk. 2023, 23 (Suppl. S1), S376–S377. [Google Scholar] [CrossRef]
  67. A Study of LB-100 in Patients with Low or Intermediate-1 Risk Myelodysplastic Syndromes (MDS), ClinicalTrials.gov ID NCT03886662. Available online: https://clinicaltrials.gov/study/NCT03886662?cond=NCT03886662&rank=1 (accessed on 11 June 2025).
  68. Garcia-Manero, G.; Madanat, Y.F.; Sekeres, M.A.; Carraway, H.E.; Cusnir, M.; McCloskey, J.; Naqvi, K.; Schiller, G.J.; Yan, L.; Gordi, T.; et al. R289, a Dual Irak 1/4 Inhibitor, in Patients with Relapsed/Refractory (R/R) Lower-Risk Myelodysplastic Syndrome (LR-MDS): Initial Results from a Phase 1b Study. Blood 2024, 144, 4595. [Google Scholar] [CrossRef]
  69. Garcia-Manero, G.; Abaza, Y.; Greenberg, P.L.; Haque, T.; Velázquez Kennedy, K.; Della Porta, M.G.; Ooi, M.G.; Stahl, M.; Xie, Z.; Mangaonkar, A.A.; et al. Preliminary Safety and Biomarker Results of the NLRP3 Inflammasome Inhibitor DFV890 in Adult Patients with Myeloid Diseases: A Phase 1b Study. Blood 2024, 144, 353. [Google Scholar] [CrossRef]
  70. Study of SX-682 Alone and in Combination with Oral or Intravenous Decitabine in Subjects with Myelodysplastic Syndrome, ClinicalTrials.gov ID NCT04245397. Available online: https://clinicaltrials.gov/study/NCT04245397?cond=NCT04245397&rank=1 (accessed on 11 June 2025).
  71. Gurnari, C.; Robin, M.; Ades, L.; Aljurf, M.; Almeida, A.M.; Duarte, F.B.; Bernard, E.; Cutler, C.S.; Della Porta, M.G.; de Witte, T.M.; et al. Clinical-genomic profiling of MDS to inform allo-HSCT:Recommendations from an international panel on behalf of the EBMT. Blood 2025, 145, 1987–2001. [Google Scholar] [CrossRef] [PubMed]
  72. Nakamura, R.; Saber, W.; Martens, M.J.; Ramirez, A.; Scott, B.; Oran, B.; Leifer, E.; Tamari, R.; Mishra, A.; Maziarz, R.T.; et al. Biologic Assignment Trial of Reduced-Intensity Hematopoietic Cell Transplantation Based on Donor Availability in Patients 50-75 Years of Age with Advanced Myelodysplastic Syndrome. J. Clin. Oncol. 2021, 39, 3328–3339. [Google Scholar] [CrossRef] [PubMed]
  73. Kröger, N.; Sockel, K.; Wolschke, C.; Bethge, W.; Schlenk, R.F.; Wolf, D.; Stadler, M.; Kobbe, G.; Wulf, G.; Bug, G.; et al. Comparison Between 5-Azacytidine Treatment and Allogeneic Stem-Cell Transplantation in Elderly Patients with Advanced MDS According to Donor Availability (VidazaAllo Study). J. Clin. Oncol. 2021, 39, 3318–3327. [Google Scholar] [CrossRef] [PubMed]
  74. Robin, M.; Porcher, R.; Adès, L.; Raffoux, E.; Michallet, M.; François, S.; Cahn, J.Y.; Delmer, A.; Wattel, E.; Vigouroux, S.; et al. HLA-matched allogeneic stem cell transplantation improves outcome of higher risk myelodysplastic syndrome A prospective study on behalf of SFGM-TC and GFM. Leukemia 2015, 29, 1496–1501. [Google Scholar] [CrossRef] [PubMed]
  75. Bischof, L.; Ussmann, J.; Platzbecker, U.; Jentzsch, M.; Franke, G.N. Allogeneic stem cell transplantation for MDS-clinical issues, choosing preparative regimens and outcome. Leuk. Lymphoma 2025, 1–14. [Google Scholar] [CrossRef] [PubMed]
  76. Shaw, B.E.; Jimenez-Jimenez, A.M.; Burns, L.J.; Logan, B.R.; Khimani, F.; Shaffer, B.C.; Shah, N.N.; Mussetter, A.; Tang, X.Y.; McCarty, J.M.; et al. National Marrow Donor Program-Sponsored Multicenter, Phase II Trial of HLA-Mismatched Unrelated Donor Bone Marrow Transplantation Using Post-Transplant Cyclophosphamide. J. Clin. Oncol. 2021, 39, 1971–1982. [Google Scholar] [CrossRef] [PubMed]
  77. Luznik, L.; Pasquini, M.C.; Logan, B.; Soiffer, R.J.; Wu, J.; Devine, S.M.; Geller, N.; Giralt, S.; Heslop, H.E.; Horowitz, M.M.; et al. Randomized Phase III BMT CTN Trial of Calcineurin Inhibitor-Free Chronic Graft-Versus-Host Disease Interventions in Myeloablative Hematopoietic Cell Transplantation for Hematologic Malignancies. J. Clin. Oncol. 2022, 40, 356–368. [Google Scholar] [CrossRef] [PubMed]
  78. Alzahrani, M.; Power, M.; Abou Mourad, Y.; Barnett, M.; Broady, R.; Forrest, D.; Gerrie, A.; Hogge, D.; Nantel, S.; Sanford, D.; et al. Improving Revised International Prognostic Scoring System Pre-Allogeneic Stem Cell Transplantation Does Not Translate Into Better Post-Transplantation Outcomes for Patients with Myelodysplastic Syndromes: A Single-Center Experience. Biol. Blood Marrow Transplant. 2018, 24, 1209–1215. [Google Scholar] [CrossRef] [PubMed]
  79. Yahng, S.A.; Kim, M.; Kim, T.M.; Jeon, Y.W.; Yoon, J.H.; Shin, S.H.; Lee, S.E.; Eom, K.S.; Lee, S.; Min, C.K.; et al. Better transplant outcome with pre-transplant marrow response after hypomethylating treatment in higher-risk MDS with excess blasts. Oncotarget 2017, 8, 12342–12354. [Google Scholar] [CrossRef] [PubMed]
  80. Schroeder, J.C.; Mix, L.; Faustmann, P.; Weller, J.F.; Fehn, A.; Phely, L.; Riedel, A.; Vogel, W.; Faul, C.; Lengerke, C.; et al. Superior outcome of upfront allogeneic hematopoietic cell transplantation versus hypomethylating agent induction in myelodysplastic syndrome. Bone Marrow Transplant. 2024, 59, 1332–1334. [Google Scholar] [CrossRef] [PubMed]
  81. Wang, H.; Li, Y.; Zhou, W.; Wang, R.; Li, Y.; Yu, L. Pre-transplant therapy for patients with myelodysplastic syndromes: A systematic review and meta-analysis. Leuk. Res. 2021, 110, 106645. [Google Scholar] [CrossRef] [PubMed]
  82. Zeidan, A.M.; Platzbecker, U.; Bewersdorf, J.P.; Stahl, M.; Ades, L.; Borate, U.; Bowen, D.T.; Buckstein, R.J.; Brunner, A.M.; Carraway, H.E.; et al. Consensus proposal for revised International Working Group response criteria for higher risk myelodysplastic syndromes. Blood 2023, 141, 2047–2061. [Google Scholar] [CrossRef] [PubMed]
  83. Rolles, B.; Bewersdorf, J.P.; Kewan, T.; Blaha, O.; Stempel, J.M.; Lanino, L.; Al Ali, N.H.; Dezern, A.E.; Sekeres, M.A.; Uy, G.L.; et al. Impact of Response to Hypomethylating Agent-Based Therapy on Survival Outcomes in the Context of Baseline Clinical-Molecular Risk and Transplant Status in Patients with Myelodysplastic Syndromes/Neoplasms (MDS): An Analysis from the International Consorti. Blood 2024, 144, 664. [Google Scholar] [CrossRef]
  84. Oran, B.; De Lima, M.; Garcia-Manero, G.; Thall, P.F.; Lin, R.; Popat, U.; Alousi, A.M.; Hosing, C.; Giralt, S.; Rondon, G.; et al. A phase 3 randomized study of 5-azacitidine maintenance vs observation after transplant in high-risk AML and MDS patients. Blood Adv. 2020, 4, 5580–5588. [Google Scholar] [CrossRef] [PubMed]
  85. Gao, L.; Zhang, Y.; Wang, S.; Kong, P.; Su, Y.; Hu, J.; Jiang, M.; Bai, H.; Lang, T.; Wang, J.; et al. Effect of rhG-CSF Combined with Decitabine Prophylaxis on Relapse of Patients with High-Risk MRD-Negative AML After HSCT: An Open-Label, Multicenter, Randomized Controlled Trial. J. Clin. Oncol. 2020, 38, 4249–4259. [Google Scholar] [CrossRef] [PubMed]
  86. Randomised Study of Oral Azacitidine vs Placebo Maintenance in AML or MDS Patients After Allo-SCT (AMADEUS), ClinicalTrials.gov ID NCT04173533. Available online: https://clinicaltrials.gov/study/NCT04173533 (accessed on 21 February 2025).
  87. De Lima, M.; Oran, B.; Champlin, R.E.; Papadopoulos, E.B.; Giralt, S.A.; Scott, B.L.; William, B.M.; Hetzer, J.; Laille, E.; Hubbell, B.; et al. CC-486 Maintenance after Stem Cell Transplantation in Patients with Acute Myeloid Leukemia or Myelodysplastic Syndromes. Biol. Blood Marrow Transplant. 2018, 24, 2017–2024. [Google Scholar] [CrossRef] [PubMed]
  88. Preemptive CIML NK Cell Therapy After Hematopoietic Stem Cell Transplantation, ClinicalTrials.gov ID NCT06138587. Available online: https://clinicaltrials.gov/study/NCT06138587?cond=NCT06138587&rank=1 (accessed on 11 June 2025).
  89. Pre-Emptive Therapy with DEC-C to Improve Outcomes in MDS Patients with Measurable Residual Disease Post Allogeneic Hematopoietic Cell Transplant, ClinicalTrials.gov ID NCT04742634. Available online: https://clinicaltrials.gov/study/NCT04742634?cond=NCT04742634&rank=1 (accessed on 11 June 2025).
  90. Fathi, A.T.; Kim, H.T.; Soiffer, R.J.; Levis, M.J.; Li, S.; Kim, A.S.; Defilipp, Z.; El-Jawahri, A.; McAfee, S.L.; Brunner, A.M.; et al. Multicenter Phase I Trial of Ivosidenib as Maintenance Treatment Following Allogeneic Hematopoietic Cell Transplantation for IDH1-Mutated Acute Myeloid Leukemia. Clin. Cancer Res. 2023, 29, 2034–2042. [Google Scholar] [CrossRef] [PubMed]
  91. Fathi, A.T.; Kim, H.T.; Soiffer, R.J.; Levis, M.J.; Li, S.; Kim, A.S.; Mims, A.S.; Defilipp, Z.; El-Jawahri, A.; McAfee, S.L.; et al. Enasidenib as maintenance following allogeneic hematopoietic cell transplantation for IDH2-mutated myeloid malignancies. Blood Adv. 2022, 6, 5857–5865. [Google Scholar] [CrossRef] [PubMed]
  92. Mishra, A.; Tamari, R.; Dezern, A.E.; Byrne, M.T.; Gooptu, M.; Chen, Y.-B.; Deeg, H.J.; Sallman, D.; Gallacher, P.; Wennborg, A.; et al. Eprenetapopt Plus Azacitidine After Allogeneic Hematopoietic Stem-Cell Transplantation for TP53-Mutant Acute Myeloid Leukemia and Myelodysplastic Syndromes. J. Clin. Oncol. 2022, 40, 3985–3993. [Google Scholar] [CrossRef] [PubMed]
  93. Guillaume, T.; Thepot, S.; Peterlin, P.; Ceballos, P.; Bourgeois, A.L.; Garnier, A.; Orvain, C.; Giltat, A.; Francois, S.; Bris, Y.L.; et al. Prophylactic or Preemptive Low-Dose Azacitidine and Donor Lymphocyte Infusion to Prevent Disease Relapse following Allogeneic Transplantation in Patients with High-Risk Acute Myelogenous Leukemia or Myelodysplastic Syndrome. Transplant. Cell Ther. 2021, 27, 839.e1-839.e6. [Google Scholar] [CrossRef] [PubMed]
  94. Panobinostat Maintenance After HSCT fo High-Risk AML and MDS, ClinicalTrials.gov ID NCT04326764. Available online: https://clinicaltrials.gov/study/NCT04326764?cond=NCT04326764&rank=1 (accessed on 11 June 2025).
  95. Bug, G.; Burchert, A.; Wagner, E.M.; Kröger, N.; Berg, T.; Güller, S.; Metzelder, S.K.; Wolf, A.; Hünecke, S.; Bader, P.; et al. Phase I/II study of the deacetylase inhibitor panobinostat after allogeneic stem cell transplantation in patients with high-risk MDS or AML (PANOBEST trial). Leukemia 2017, 31, 2523–2525. [Google Scholar] [CrossRef] [PubMed]
  96. Ma, Y.Y.; Wei, Z.L.; Xu, Y.J.; Shi, J.M.; Yi, H.; Lai, Y.R.; Jiang, E.L.; Wang, S.B.; Wu, T.; Gao, L.; et al. Poor pretransplantation minimal residual disease clearance as an independent prognostic risk factor for survival in myelodysplastic syndrome with excess blasts: A multicenter, retrospective cohort study. Cancer 2023, 129, 2013–2022. [Google Scholar] [CrossRef] [PubMed]
  97. Huang, C.; Jia, Y.; Yang, J.; Cai, Y.; Tong, Y.; Qiu, H.; Zhou, K.; Xia, X.; Zhang, Y.; Shen, C.; et al. Azacitidine combined with interferon-α for pre-emptive treatment of AML/MDS after allogeneic peripheral blood stem cell transplantation: A prospective phase II study. Br. J. Haematol. 2024, 205, 1067–1076. [Google Scholar] [CrossRef] [PubMed]
  98. Sallman, D.; McLemore, A.F.; Komrokji, R.S.; Elmariah, H.; Kuykendall, A.T.; Bejanyan, N.; Chan, O.; Faramand, R.; Pidala, J.A.; Yoder, S.J.; et al. Measurable Residual Disease Monitoring By Duplex Sequencing for TP53 in the Post Allogeneic Stem Cell Transplantation Study with Eprenetapopt (APR-246) + Azacitidine Strongly Predicts Outcomes. Blood 2024, 144, 1046. [Google Scholar] [CrossRef]
  99. Schulz, E.; Aplan, P.D.; Freeman, S.D.; Pavletic, S.Z. Moving toward a conceptualization of measurable residual disease in myelodysplastic syndromes. Blood Adv. 2023, 7, 4381–4394. [Google Scholar] [CrossRef] [PubMed]
  100. Tobiasson, M.; Pandzic, T.; Illman, J.; Nilsson, L.; Westrom, S.; Ejerblad, E.; Olesen, G.; Bjorklund, A.; Olsnes Kittang, A.; Werlenius, O.; et al. Patient-Specific Measurable Residual Disease Markers Predict Outcome in Patients with Myelodysplastic Syndrome and Related Diseases After Hematopoietic Stem-Cell Transplantation. J. Clin. Oncol. 2024, 42, 1378–1390. [Google Scholar] [CrossRef] [PubMed]
  101. Kaminskas, E.; Farrell, A.T.; Wang, Y.-C.; Sridhara, R.; Pazdur, R. FDA Drug Approval Summary: Azacitidine (5-azacytidine, Vidaza™) for Injectable Suspension. Oncologist 2005, 10, 176–182. [Google Scholar] [CrossRef] [PubMed]
  102. Abbvie Press Release, June 16, 2025. AbbVie Provides Update on VERONA Trial for Newly Diagnosed Higher-Risk Myelodysplastic Syndromes. Available online: https://news.abbvie.com/2025-06-16-AbbVie-Provides-Update-on-VERONA-Trial-for-Newly-Diagnosed-Higher-Risk-Myelodysplastic-Syndromes (accessed on 16 June 2025).
  103. APR-246 & Azacitidine for the Treatment of TP53 Mutant Myelodysplastic Syndromes (MDS), ClinicalTrials.gov ID NCT03745716. Available online: https://clinicaltrials.gov/study/NCT03745716?term=apr-246&cond=mds&rank=1&tab=results#more-information (accessed on 11 June 2025).
  104. Garcia-Manero, G.; Fenaux, P.; Al-Kali, A.; Baer, M.R.; Sekeres, M.A.; Roboz, G.J.; Gaidano, G.; Scott, B.L.; Greenberg, P.; Platzbecker, U.; et al. Rigosertib versus best supportive care for patients with high-risk myelodysplastic syndromes after failure of hypomethylating drugs (ONTIME): A randomised, controlled, phase 3 trial. Lancet Oncol. 2016, 17, 496–508. [Google Scholar] [CrossRef] [PubMed]
  105. Garcia-Manero, G.; McCloskey, J.; Scott, B.L.; Griffiths, E.A.; Kiner-Strachan, B.; Brunner, A.M.; Zeidan, A.M.; Traer, E.; Madanat, Y.F.; Meyer, J.; et al. Results from a Phase 1 Open-Label Dose Escalation and Expansion Trial of Oral Azacitidine + Cedazuridine (ASTX030) in Patients with Myelodysplastic Syndromes (MDS) and MDS/Myeloproliferative Neoplasms (MPN). Blood 2024, 144, 662. [Google Scholar] [CrossRef]
  106. Adès, L.; Girshova, L.; Doronin, V.A.; Díez-Campelo, M.; Valcárcel, D.; Kambhampati, S.; Viniou, N.-A.; Woszczyk, D.; De Paz Arias, R.; Symeonidis, A.; et al. Pevonedistat plus azacitidine vs azacitidine alone in higher-risk MDS/chronic myelomonocytic leukemia or low-blast-percentage AML. Blood Adv. 2022, 6, 5132–5145. [Google Scholar] [CrossRef] [PubMed]
  107. Magrolimab + Azacitidine Versus Azacitidine + Placebo in Untreated Participants with Myelodysplastic Syndrome (MDS) (ENHANCE), ClinicalTrials.gov ID NCT04313881. Available online: https://clinicaltrials.gov/study/NCT04313881 (accessed on 24 June 2025).
  108. Zeidan, A.M.; Ando, K.; Rauzy, O.; Turgut, M.; Wang, M.C.; Cairoli, R.; Hou, H.A.; Kwong, Y.L.; Arnan, M.; Meers, S.; et al. Sabatolimab plus hypomethylating agents in previously untreated patients with higher-risk myelodysplastic syndromes (STIMULUS-MDS1): A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Haematol. 2024, 11, e38–e50. [Google Scholar] [CrossRef] [PubMed]
  109. Tamibarotene Plus Azacitidine in Participants with Newly Diagnosed RARA-Positive Higher-Risk Myelodysplastic Syndrome, ClinicalTrials.gov ID NCT04797780. Available online: https://clinicaltrials.gov/study/NCT04797780?tab=results (accessed on 24 June 2025).
  110. Lisaftoclax (APG-2575) Combined with Azacytidine (AZA) in the Treatment of Patients with Higher-Risk Myelodysplastic Syndrome (GLORA-4), ClinicalTrials.gov ID NCT06641414. Available online: https://clinicaltrials.gov/study/NCT06641414?term=lisaftoclax%20&rank=3 (accessed on 19 July 2025).
  111. A Trial of AK117 (Anti-CD47) in Patients with Myelodysplastic Syndrome, ClinicalTrials.gov ID NCT04900350. Available online: https://clinicaltrials.gov/study/NCT04900350 (accessed on 24 June 2025).
  112. A Study of BGB-11417 in Participants with Myeloid Malignancies, ClinicalTrials.gov ID NCT04771130. Available online: https://clinicaltrials.gov/study/NCT04771130?cond=mds&term=sonrotoclax%20&rank=1#study-record-dates (accessed on 19 July 2025).
  113. Dinardo, C.D.; Roboz, G.J.; Watts, J.M.; Madanat, Y.F.; Prince, G.T.; Baratam, P.; De Botton, S.; Stein, A.; Foran, J.M.; Arellano, M.L.; et al. Final phase 1 substudy results of ivosidenib for patients with mutant IDH1 relapsed/refractory myelodysplastic syndrome. Blood Adv. 2024, 8, 4209–4220. [Google Scholar] [CrossRef] [PubMed]
  114. Sebert, M.; Chevret, S.; Dimicoli-Salazar, S.; Cluzeau, T.; Rauzy, O.; Stamatoulas Bastard, A.; Laribi, K.; Fossard, G.; Thépot, S.; Gloaguen, S.; et al. Enasidenib (ENA) Monotherapy in Patients with IDH2 mutated Myelodysplastic Syndrome (MDS), the Ideal Phase 2 Study By the GFM and Emsco Groups. Blood 2024, 144, 1839. [Google Scholar] [CrossRef]
  115. Fenaux, P.; Mufti, G.J.; Hellstrom-Lindberg, E.; Santini, V.; Finelli, C.; Giagounidis, A.; Schoch, R.; Gattermann, N.; Sanz, G.; List, A.; et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: A randomised, open-label, phase III study. Lancet Oncol. 2009, 10, 223–232. [Google Scholar] [CrossRef] [PubMed]
  116. Zeidan, A.M.; Salimi, T.; Epstein, R.S. Real-world use and outcomes of hypomethylating agent therapy in higher-risk myelodysplastic syndromes: Why are we not achieving the promise of clinical trials? Future Oncol. 2021, 17, 5163–5175. [Google Scholar] [CrossRef] [PubMed]
  117. Rajakumaraswamy, N.; Gandhi, M.; Wei, A.H.; Sallman, D.A.; Daver, N.G.; Mo, S.; Iqbal, S.; Karalliyadda, R.; Chen, M.; Wang, Y.; et al. Real-world Effectiveness of Azacitidine in Treatment-Naive Patients with Higher-risk Myelodysplastic Syndromes. Clin. Lymphoma Myeloma Leuk. 2024, 24, 260-268.e2. [Google Scholar] [CrossRef] [PubMed]
  118. Kantarjian, H.; Issa, J.-P.J.; Rosenfeld, C.S.; Bennett, J.M.; Albitar, M.; Dipersio, J.; Klimek, V.; Slack, J.; De Castro, C.; Ravandi, F.; et al. Decitabine improves patient outcomes in myelodysplastic syndromes. Cancer 2006, 106, 1794–1803. [Google Scholar] [CrossRef] [PubMed]
  119. Nazha, A.; Sekeres, M.A.; Garcia-Manero, G.; Barnard, J.; Al Ali, N.H.; Roboz, G.J.; Steensma, D.P.; Dezern, A.E.; Zimmerman, C.; Jabbour, E.J.; et al. Outcomes of patients with myelodysplastic syndromes who achieve stable disease after treatment with hypomethylating agents. Leuk. Res. 2016, 41, 43–47. [Google Scholar] [CrossRef] [PubMed]
  120. Garcia-Manero, G.; Griffiths, E.A.; Steensma, D.P.; Roboz, G.J.; Wells, R.; McCloskey, J.; Odenike, O.; DeZern, A.E.; Yee, K.; Busque, L.; et al. Oral cedazuridine/decitabine for MDS and CMML: A phase 2 pharmacokinetic/pharmacodynamic randomized crossover study. Blood 2020, 136, 674–683. [Google Scholar] [CrossRef] [PubMed]
  121. Garcia-Manero, G.; McCloskey, J.; Griffiths, E.A.; Yee, K.W.L.; Zeidan, A.M.; Al-Kali, A.; Deeg, H.J.; Patel, P.A.; Sabloff, M.; Keating, M.M.; et al. Oral decitabine-cedazuridine versus intravenous decitabine for myelodysplastic syndromes and chronic myelomonocytic leukaemia (ASCERTAIN): A registrational, randomised, crossover, pharmacokinetics, phase 3 study. Lancet Haematol. 2024, 11, e15–e26. [Google Scholar] [CrossRef] [PubMed]
  122. Silverman, L.R.; Verma, A.; Odchimar-Reissig, R.; Feldman, E.J.; Navada, S.; Demakos, E.P.; Baer, M.R.; Najfeld, V.; Sparano, J.A.; Piekarz, R. A Phase II Trial Of Epigenetic Modulators Vorinostat In Combination with Azacitidine (azaC) In Patients with The Myelodysplastic Syndrome (MDS): Initial Results Of Study 6898 Of The New York Cancer Consortium. Blood 2013, 122, 383. [Google Scholar] [CrossRef]
  123. Sekeres, M.A.; Othus, M.; List, A.F.; Odenike, O.; Stone, R.M.; Gore, S.D.; Litzow, M.R.; Buckstein, R.; Fang, M.; Roulston, D.; et al. 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, 2745–2753. [Google Scholar] [CrossRef] [PubMed]
  124. Sekeres, M.A.; Watts, J.; Radinoff, A.; Sangerman, M.A.; Cerrano, M.; Lopez, P.F.; Zeidner, J.F.; Campelo, M.D.; Graux, C.; Liesveld, J.; et al. Randomized phase 2 trial of pevonedistat plus azacitidine versus azacitidine for higher-risk MDS/CMML or low-blast AML. Leukemia 2021, 35, 2119–2124. [Google Scholar] [CrossRef] [PubMed]
  125. Sallman, D.A.; Al Malki, M.M.; Asch, A.S.; Wang, E.S.; Jurcic, J.G.; Bradley, T.J.; Flinn, I.W.; Pollyea, D.A.; Kambhampati, S.; Tanaka, T.N.; et al. Magrolimab in Combination with Azacitidine in Patients with Higher-Risk Myelodysplastic Syndromes: Final Results of a Phase Ib Study. J. Clin. Oncol. 2023, 41, 2815–2826. [Google Scholar] [CrossRef] [PubMed]
  126. Gilead Press Release, July 21, 2023. Gilead To Discontinue Phase 3 ENHANCE Study of Magrolimab Plus Azacitidine in Higher-Risk MDS. Available online: https://www.gilead.com/news/news-details/2023/gilead-to-discontinue-phase-3-enhance-study-of-magrolimab-plus-azacitidine-in-higher-risk-mds (accessed on 2 March 2025).
  127. Zeidan, A.M.; Xiao, Z.; Sanz, G.; Giagounidis, A.; Sekeres, M.; Lao, Z.; Deeren, D.; Gao, S.; Riva, M.; Lee, J.; et al. Primary results of the phase III STIMULUS-MDS2 study of sabatolimab + azacitidine vs placebo + azacitidine as frontline therapy for patients with higher-risk MDS or CMML-2. In Proceedings of the European Hematology Association Congress 2024, Madrid, Spain, 13–16 June 2024. Abstract S180. [Google Scholar]
  128. Brunner, A.M.; Esteve, J.; Porkka, K.; Knapper, S.; Traer, E.; Scholl, S.; Garcia-Manero, G.; Vey, N.; Wermke, M.; Janssen, J.J.W.M.; et al. 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. 2023, 99, E32–E36. [Google Scholar] [CrossRef] [PubMed]
  129. Dinardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar] [CrossRef] [PubMed]
  130. Bogenberger, J.M.; Kornblau, S.M.; Pierceall, W.E.; Lena, R.; Chow, D.; Shi, C.X.; Mantei, J.; Ahmann, G.; Gonzales, I.M.; Choudhary, A.; et al. BCL-2 family proteins as 5-Azacytidine-sensitizing targets and determinants of response in myeloid malignancies. Leukemia 2014, 28, 1657–1665. [Google Scholar] [CrossRef] [PubMed]
  131. Bazinet, A.; Darbaniyan, F.; Jabbour, E.; Montalban-Bravo, G.; Ohanian, M.; Chien, K.; Kadia, T.; Takahashi, K.; Masarova, L.; Short, N.; et al. Azacitidine plus venetoclax in patients with high-risk myelodysplastic syndromes or chronic myelomonocytic leukaemia: Phase 1 results of a single-centre, dose-escalation, dose-expansion, phase 1–2 study. Lancet Haematol. 2022, 9, e756–e765. [Google Scholar] [CrossRef] [PubMed]
  132. Zeidan, A.M.; Borate, U.; Pollyea, D.A.; Brunner, A.M.; Roncolato, F.; Garcia, J.S.; Filshie, R.; Odenike, O.; Watson, A.M.; Krishnadasan, R.; et al. A phase 1b study of venetoclax and azacitidine combination in patients with relapsed or refractory myelodysplastic syndromes. Am. J. Hematol. 2023, 98, 272–281. [Google Scholar] [CrossRef] [PubMed]
  133. Apodaca Chavez, E.I.; Crisp, R.; Varela Constantino, A.L.; Enrico, A.I.; Gomez-De Leon, A.; Ovilla-Martínez, R.; Wernicke, P.; Camargo Molano, C.; Boada, M.; Velloso, E.D.R.P.; et al. Azacitidine and Venetoclax in High-Risk Myelodysplastic Syndrome: A Real-World Perspective from the Glam Registry with Long-Term Follow-up. Blood 2024, 144, 6718. [Google Scholar] [CrossRef]
  134. Guru Murthy, G.S.; Ball, S.; Feld, J.; Abboud, R.; Haddadin, M.; Patel, S.A.; Kishtagari, A.; Hawkins, H.; Guerra, V.; Dworkin, E.; et al. Clinical Utilization and Outcomes of Hypomethylating Agents and Venetoclax in Patients with Myelodysplastic Syndrome—A Multicenter Retrospective Analysis. Blood 2024, 144, 3206. [Google Scholar] [CrossRef]
  135. Garcia, J.S.; Platzbecker, U.; Odenike, O.; Fleming, S.; Fong, C.Y.; Borate, U.; Jacoby, M.A.; Nowak, D.; Baer, M.R.; Peterlin, P.; et al. Efficacy and safety of venetoclax plus azacitidine for patients with treatment-naive high-risk myelodysplastic syndromes. Blood 2025, 145, 1126–1135. [Google Scholar] [CrossRef] [PubMed]
  136. Zeidan, A.M.; Garcia, J.S.; Fenaux, P.; Platzbecker, U.; Miyazaki, Y.; Xiao, Z.; Zhou, Y.; Naqvi, K.; Kye, S.; Garcia-Manero, G. Phase 3 VERONA study of venetoclax with azacitidine to assess change in complete remission and overall survival in treatment-naïve higher-risk myelodysplastic syndromes. J. Clin. Oncol. 2021, 39, TPS7054. [Google Scholar] [CrossRef]
  137. Bataller, A.; Montalban-Bravo, G.; Bazinet, A.; Alvarado, Y.; Chien, K.; Venugopal, S.; Ishizawa, J.; Hammond, D.; Swaminathan, M.; Sasaki, K.; et al. Oral decitabine plus cedazuridine and venetoclax in patients with higher-risk myelodysplastic syndromes or chronic myelomonocytic leukaemia: A single-centre, phase 1/2 study. Lancet Haematol. 2024, 11, e186–e195. [Google Scholar] [CrossRef] [PubMed]
  138. Medeiros, B.C.; Fathi, A.T.; Dinardo, C.D.; Pollyea, D.A.; Chan, S.M.; Swords, R. Isocitrate dehydrogenase mutations in myeloid malignancies. Leukemia 2017, 31, 272–281. [Google Scholar] [CrossRef] [PubMed]
  139. Sebert, M.; Cluzeau, T.; Beyne-Rauzy, O.; Stamatoullas-Bastard, A.; Dimicoli-Salazar, S.; Thepot, S.; Peterlin, P.; Park, S.; Gourin, M.-P.; Brehar, O.; et al. Ivosidenib Monotherapy Is Effective in Patients with IDH1 Mutated Myelodysplastic Syndrome (MDS): The Idiome Phase 2 Study By the GFM Group. Blood 2021, 138 (Suppl. S1), 62. [Google Scholar] [CrossRef]
  140. Sebert, M.; Clappier, E.; Cluzeau, T.; Bergugnat, H.; Beyne-Rauzy, O.; Stamatoullas-Bastard, A.; Dimicoli-Salazar, S.; Larcher, L.; Goldwirt, L.; Thepot, S.; et al. Ivosidenib Monotherapy in IDH1 Mutated Myelodysplastic Syndrome, Final Results of the IDIOME Trial, a GFM Study. EHA Library, S182 2024. Available online: https://library.ehaweb.org/eha/2024/eha2024-congress/422286/marie.sbert.ivosidenib.monotherapy.in.idh1.mutated.myelodysplastic.syndrome.html (accessed on 12 May 2025).
  141. Sebert, M.; Platzbecker, U.; Valcárcel Ferreiras, D.; Santini, V.; Borate, U.; Kapsalis, S.M.; Simonot, L.; Yuan, W.; Patel, P.A.; Garcia-Manero, G. Phase 3 Study of Either Ivosidenib (IVO) Monotherapy or Azacitidine (AZA) Monotherapy in Patients with IDH1 Mutant Myelodysplastic Syndromes (MDS) Who Are Hypomethylating Agent (HMA) Naive (PyramIDH). Blood 2024, 114 (Suppl. S1), 1845.1841. [Google Scholar] [CrossRef]
  142. Forma Therapeutics, Inc. REZLIDHIA™ (Olutasidenib) Capsules, for Oral Use; Forma Therapeutics, Inc.: Greenville, NC, USA, 2025. [Google Scholar]
  143. Cortes, J.; Yang, J.; Lee, S.; Dinner, S.; Wang, E.S.; Baer, M.R.; Donnellan, W.; Watts, J.M. Olutasidenib Alone or in Combination with Azacitidine Induces Durable Complete Remissions in Patients with mIDH1 Myelodysplastic Syndromes/Neoplasms (MDS). Blood 2023, 142 (Suppl. S1), 1872. [Google Scholar] [CrossRef]
  144. Olutasidenib in Combination with Hypomethylation Therapy for the Treatment of IDH1-Mutated Higher-Risk Myelodysplastic Syndromes, Chronic Myelomonocytic Leukemia, and Myeloproliferative Neoplasms. ClinicalTrials.ov ID. Available online: https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2024-07758&r=1 (accessed on 12 May 2025).
  145. Stein, E.M.; Fathi, A.T.; DiNardo, C.D.; Pollyea, D.A.; Roboz, G.J.; Collins, R.; Sekeres, M.A.; Stone, R.M.; Attar, E.C.; Frattini, M.G.; et al. Enasidenib in patients with mutant IDH2 myelodysplastic syndromes: A phase 1 subgroup analysis of the multicentre, AG221-C-001 trial. Lancet Haematol. 2020, 7, e309–e319. [Google Scholar] [CrossRef] [PubMed]
  146. Dinardo, C.D.; Venugopal, S.; Lachowiez, C.; Takahashi, K.; Loghavi, S.; Montalban-Bravo, G.; Wang, X.; Carraway, H.; Sekeres, M.; Sukkur, A.; et al. Targeted therapy with the mutant IDH2 inhibitor enasidenib for high-risk IDH2-mutant myelodysplastic syndrome. Blood Adv. 2023, 7, 2378–2387. [Google Scholar] [CrossRef] [PubMed]
  147. Bernard, E.; Nannya, Y.; Hasserjian, R.P.; Devlin, S.M.; Tuechler, H.; Medina-Martinez, J.S.; Yoshizato, T.; Shiozawa, Y.; Saiki, R.; Malcovati, L.; et al. Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes. Nat. Med. 2020, 26, 1549–1556. [Google Scholar] [CrossRef] [PubMed]
  148. Cluzeau, T.; Sebert, M.; Rahmé, R.; Cuzzubbo, S.; Lehmann-Che, J.; Madelaine, I.; Peterlin, P.; Bève, B.; Attalah, H.; Chermat, F.; et al. Eprenetapopt Plus Azacitidine in TP53-Mutated Myelodysplastic Syndromes and Acute Myeloid Leukemia: A Phase II Study by the Groupe Francophone des Myélodysplasies (GFM). J. Clin. Oncol. 2021, 39, 1575–1583. [Google Scholar] [CrossRef] [PubMed]
  149. Sallman, D.A.; Dezern, A.E.; Garcia-Manero, G.; Steensma, D.P.; Roboz, G.J.; Sekeres, M.A.; Cluzeau, T.; Sweet, K.L.; McLemore, A.; McGraw, K.L.; et al. Eprenetapopt (APR-246) and Azacitidine in TP53-Mutant Myelodysplastic Syndromes. J. Clin. Oncol. 2021, 39, 1584–1594. [Google Scholar] [CrossRef] [PubMed]
  150. Aprea Therapeutics, Press Release December 28, 2020. Aprea Therapeutics Announces Results of Primary Endpoint from Phase 3 Trial of Eprenetapopt in TP53 Mutant Myelodysplastic Syndromes (MDS). Available online: https://ir.aprea.com/news-releases/news-release-details/aprea-therapeutics-announces-results-primary-endpoint-phase-3 (accessed on 2 February 2024).
Figure 1. A proposed treatment algorithm of patients with LR-MDS in 2025. Footnotes: ANC: absolute neutrophil count, EPO: erythropoietin, ESA: erythropoietin-stimulating agents, HMA: hypomethylating agents, IST: immunosuppressive therapy, RS: ringed sideroblasts, TPO: thrombopoietin.
Figure 1. A proposed treatment algorithm of patients with LR-MDS in 2025. Footnotes: ANC: absolute neutrophil count, EPO: erythropoietin, ESA: erythropoietin-stimulating agents, HMA: hypomethylating agents, IST: immunosuppressive therapy, RS: ringed sideroblasts, TPO: thrombopoietin.
Cancers 17 02469 g001
Figure 2. A proposed treatment algorithm of patients with HR-MDS in 2025. Footnotes: Allo-HCT: allogeneic stem cell transplant, DLI: donor lymphocyte infusion HMA: hypomethylating agents, IV: intravenous, SC: subcutaneous.
Figure 2. A proposed treatment algorithm of patients with HR-MDS in 2025. Footnotes: Allo-HCT: allogeneic stem cell transplant, DLI: donor lymphocyte infusion HMA: hypomethylating agents, IV: intravenous, SC: subcutaneous.
Cancers 17 02469 g002
Table 1. Comparison of Risk Stratification Models for Clonal Cytopenias: CHRS vs. CCRS.
Table 1. Comparison of Risk Stratification Models for Clonal Cytopenias: CHRS vs. CCRS.
FeatureCHRS (Clonal Hematopoiesis Risk Score)CCRS (Clonal Cytopenia Risk Score)
PublicationWeeks et al., NEJM Evidence, 2023 [32]Xie et al., Blood, 2024 [31]
PurposePredicts risk of progression to myeloid neoplasm malignancy in patients CHIP/CCUSStratifies CCUS patients based on progression risk to MDS or AML
PopulationCHIP and CCUS (including individuals
without cytopenias)
Specifically developed for CCUS
Key Components8 prognostic factors:
Number of mutations, specific genes
(high-risk mutations, DNMT3A), VAF, presence of cytopenia, RDW, MCV, age
3 prognostic factors:
Number of mutations (≥2), specific genes (splicing factor mutations), and platelet count < 100 × 109/L
Risk Scoring Low risk: ≤9.5 points
Intermediate risk: 10–12 points
High risk: ≥12.5 points
Low risk: <2.5 points
Intermediate risk: 2.5–≤5 points
High risk: ≥5 points
Gene weightingHigher risk for splicing factor mutations,
AML-like (IDH1/2, FLT3, RUNX1), JAK2 and TP53 assigned score 2.5; if ≥2 mutations assigned score of 2
Splicing factor mutations assigned score of 2;
if ≥2 mutations assigned score of 3
Risk for MN5-year CI for MN (±SD) (%):
No CHIP/CCUS: 0.0740 (±0.0064)
Low risk: 0.232 (±0.0484)
Intermediate risk: 2.76 (±0.482)
High risk: 24.4 (±4.12)
2-year CI (%, 95% confidence
interval) of MN progression:
Low risk: 6.4 (13–11.4)
Intermediate risk: 14.1 (7.9–22.2)
High risk: 37.2 (19.8–54.7)
StrengthsLarge population cohort; integrates age and VAF; validatedOnly 3 prognostic factors; CCUS-specific; validated
AML: acute myeloid leukemia, CCUS: clonal cytopenias of undetermined significance, CHIP: clonal cytopenias of indeterminate potential, CI: cumulative incidence, MDS: myelodysplastic syndromes, MN: myeloid neoplasms, SD: standard deviation, VAF: variant allele frequency.
Table 2. Selected Ongoing Clinical Trials of LR-MDS.
Table 2. Selected Ongoing Clinical Trials of LR-MDS.
Agent or Drug CombinationMechanism of ActionTrial PhasePopulationClinical Trial IDReferences
Luspatercept vs. ESATGF-β superfamily ligand trapIIILR-MDS (IPSS-R ≤ 3.5), RBC-TINCT05949684Zeidan et al., ASH, 2023 [49]
KER-050
(Elritercept)
Modified activin receptor type IIA ligand trapIILR-MDS (IPSS-R ≤ 3.5)NCT04419649Diez-Campelo et al., ASH, 2023 [60]; Tan et al., ASH, 2024 [61]
RVU120CDK8/19 small molecule inhibitorIILR-MDS (IPSS-R ≤ 3.5), R/R or ineligible for other therapiesNCT06243458 [62]ClinicalTrials.gov
CanakinumabAnti-IL-1β human monoclonal antibodyIILR-MDS (IPSS-R ≤ 3.5), R/R ESA, HMAsNCT04239157Rodriguez Sevilla et al., ASH, 2023 [63]
Attenuated durations of HMAsDNMT inhibitorIILR-MDS (IPSS ≤ 1)NCT02269280Bouligny et al., EHA, 2025 [64]
Luspatercept + ESATGF-β superfamily ligand trapI/IILR-MDS (IPSS ≤ 1), non-RS, R/R ESA2021-000596-37 *Ades et al., ASH, 2024 [65]
Oral decitabine + cedazuridine (ASTX727)DNMT inhibitorI/IILR-MDS (IPSS ≤ 1),
non-RS, R/R
NCT03502668Garcia-Manero et al., SOHO, 2023 [66]
LB-100Protein phosphatase 2A inhibitorIb/IILR-MDS (IPSS ≤ 1), requiring treatmentNCT03886662 [67]ClinicalTrials.gov
R289IRAK1/4 inhibitorIbLR-MDS (IPSS-R ≤ 3.5), R/R, included del(5q)NCT05308264Garcia-Manero et al., ASH, 2024 [68]
DFV890Selective NLRP3 inhibitorIbLR-MDS (IPSS-R ≤ 3.5), previously treatedNCT05552469Garcia-Manero et al., ASH, 2024 [69]
SX-682 +/− DecitabineCXCR1/2 inhibitorILR-MDS (IPSS ≤1), R/RNCT04245397 [70]ClinicalTrials.gov
CKD: cyclin-dependent kinase, CXCR4: CXC chemokine receptor 4, DNMT: DNA methyltransferase, ESA: erythropoiesis stimulating agent, HMAs: hypomethylating agents, IL: interleukin, IPSS: international prognostic scoring system, IPSS-R: IPSS-revised, IRAK: interleukin1 receptor-associated kinase, LR-MDS: lower risk MDS, MDS: myelodysplastic syndromes, NLRP3: nucleotide-binding and leucine-rick repeat P3, R/R: relapsed/refractory, RBC-TI: red blood cell transfusion independent, TGF-ß: transforming growth factor-beta. * European Union clinical trials register identifier.
Table 3. Maintenance therapy trials for patients who remain in remission after transplant.
Table 3. Maintenance therapy trials for patients who remain in remission after transplant.
Agent/Drug CombinationTrial PhaseStatusPopulationResultsClinical Trial IDReference
Oral azacitidine vs. placeboIIIActiveAML, MDSN/ANCT04173533ClinicalTrials.gov [86]
IL-2I/IbActiveAML, MDSN/ANCT06138587ClinicalTrials.gov [88]
Decitabine and cedazuridineI/IIRecruitingMDSN/ANCT04742634ClinicalTrials.gov [89]
IDH1 inhibitorICompletedAML, MDSCIR 19% (95% CI: 4.0–41)
2y-PFS 81% (95% CI: 52–94)
2y-OS 88% (95% CI: 59–97)
NCT03564821Fathi et al., Clin Cancer Res, 2023 [90]
IDH2 inhibitorICompletedAML, MDSCIR 16% (95% CI: 3.7–36)
2y-PFS 69% (95% CI: 39–86)
2y-OS 74% (95% CI: 44–90)
NCT03515512Fathi et al., Blood Adv, 2022 [91]
Eprenetapopt + azacitidineIICompletedTP53-mutated AML, MDS1-y PFS 59.9% (95% CI: 41–74)
1y-OS 78.8% (95% CI: 60.6–89.3)
MRD monitoring after transplant predicts outcome
NCT03931291Mishra et al., JCO, 2022 [92]
Azacitidine + DLIIICompletedAML, MDS2y-PFS 68.3% (95% CI: 58.3–80.1)
2y-OS 76% (95% CI:52–90)
NCT01541280Guillaume, et al., Transpl and Cel Ther, 2021 [93]
PanobinostatI/IIPhase III TerminatedAML, MDSPhase II results:
CIR 20% (95% 7–33)
2y-RFS 75% (63–90).
2yr-OS 81% (95% CI: 69–95)
NCT04326764 [94]Bug et al., Leukemia, 2017 [95]
AML: acute myeloid leukemia, CI: confidence interval, CIR: cumulative incidence rate of relapse, DLI: donor lymphocyte infusion, IDH: isocitrate dehydrogenase, IL: interleukin, MDS: myelodysplastic syndromes, MPN: myeloproliferative neoplasms, OS: overall survival, PFS: progression-free survival.
Table 4. Selected Clinical Trials of HR-MDS.
Table 4. Selected Clinical Trials of HR-MDS.
Agent/Drug CombinationTrial PHASETarget/MOAPopulationResultsStatusClinical Trial ID
Venetoclax + AZA [102]IIIBCL-2 inhibitor + DNMTiHR-MDSDid not meet primary endpoint of OS; HR 0.908, p = 0.3772Active, not recruitingNCT04401748
APR-246
(Eprenetapopt) + AZA [103]
IIITP53TP53-mutant HR-MDSAPR-246 + AZA arm: CR 34.6%
AZA arm: CR 22.4%
CompletedNCT03745716
Rigosertib vs. BSC [104]IIIMicrotubule-destabilizing agentHR-MDS after failure of HMAsRigosertib: OS 8.2 months
(95% CI: 6.1–10.1)
Best supportive care: OS 5.9 months (95% CI: 4.1–9.3)
(HR 0.87, 95% CI: 0.67–1.14; p = 0.33)
CompletedNCT02562443
AZA and
Cedazuridine
(ASTX030) [105]
Multi-phaseOral DNMTiMDS, CMML, AMLNAActive, recruitingNCT04256317
AZA + pevonedistat [106] IIINEDD8-activating enzymeHR-MDS, CMML or AML with 20–30% blastsPevonedistat + AZA arm: median EFS 17.7 months
AZA arm: median EFS 15.7 months (HR 0.968; 95% CI: 0.757–1.238; p = 0.557) in the HR-MDS cohort
CompletedNCT03268954
AZA + magrolimab [107]IIIAnti-CD47 monoclonal antibody + DNMTiHR-MDSMagrolimab + AZA arm: CR 21.3% AZA arm: CR 23.6%TerminatedNCT04313881
AZA +
sabatolimab [108]
IIITIM-3 inhibitor + DNMTiMDS, CMML-2Median PFS:
Sabatolimab + AZA arm:
11.1 months
AZA arm: 8.5 months (p = 0.102)
CR rate:
Sabatolimab + AZA arm: 21.5%
AZA arm: 17.7% (p = 0.769)
TerminatedNCT04266301
AZA +
tamibarotene [109]
III (RARα) agonist + DNMTiHR-MDSTamibarotene + AZA arm: CR 23.8% AZA arm: CR 18.8% (p = 0.2084)TerminatedNCT04797780
AZA +
Lisaftoclax [110]
IIIBCL-2 inhibitor + DNMTiHR-MDSNARecruitingNCT06641414
AZA + AK117 [111]I/IIAnti-CD47 monoclonal antibody + DNMTiHR-MDSNAActive, not recruitingNCT04900350
AZA +/−
BGB-11417 [112]
I/IIBCL-2 inhibitor + DNMTiMDS, AMLNARecruitingNCT04771130
Ivosidenib [113]IIDH1 inhibitor +/− DNMTiHematologic malignancies with IDH1 mutationsORR of 83.3% and CR rate of 38.9% in 18 patientsActive, recruitingNCT02074839
Enasidenib [114]IIIDH2 inhibitor +/− DNMTiR/R MDS, HR-MDS, LR-MDS resistant to ESAEnasidenib monotherapy (cohort A):best OR 42.9% after 3–6 cyclesActive, not recruitingNCT03744390
AZA: azacitidine, AML: acute myeloid leukemia, BSC: best supportive care, DNMTi: DNA methyltransferase inhibitor, CI: confidence interval, CMML: chronic myelomonocytic leukemia, CR: complete remission, EFS: event free survival, ESA: erythropoiesis-stimulating agents, HMA: hypomethylating agents, HR: hazard ratio, HR-MDS: higher risk myelodysplastic syndrome, LR-MDS: lower risk MDS, MOA: mechanism of action, NA: not available, OR: overall response, ORR: overall response rate, OS: overall survival, PFS: progression free survival, R/R: relapse refractory, TIM-3: T-cell immunoglobulin domain and mucin domain-3.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Stempel, J.M.; Kewan, T.; Zeidan, A.M. Advances and Challenges in the Management of Myelodysplastic Syndromes. Cancers 2025, 17, 2469. https://doi.org/10.3390/cancers17152469

AMA Style

Stempel JM, Kewan T, Zeidan AM. Advances and Challenges in the Management of Myelodysplastic Syndromes. Cancers. 2025; 17(15):2469. https://doi.org/10.3390/cancers17152469

Chicago/Turabian Style

Stempel, Jessica M., Tariq Kewan, and Amer M. Zeidan. 2025. "Advances and Challenges in the Management of Myelodysplastic Syndromes" Cancers 17, no. 15: 2469. https://doi.org/10.3390/cancers17152469

APA Style

Stempel, J. M., Kewan, T., & Zeidan, A. M. (2025). Advances and Challenges in the Management of Myelodysplastic Syndromes. Cancers, 17(15), 2469. https://doi.org/10.3390/cancers17152469

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop