What are the new drugs for Myelodysplastic Syndromes?

17 March 2025

Overview of Myelodysplastic Syndromes 
Myelodysplastic syndromes (MDS) comprise a heterogeneous group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis, dysplasia in one or more blood cell lineages, and peripheral cytopenias that often lead to symptoms such as fatigue, infection risk, and bleeding complications. The diagnosis of MDS relies on an integration of bone marrow morphological assessment, cytogenetic evaluation, and increasingly, molecular genetic information. Traditionally, classification systems such as the World Health Organization (WHO) criteria and prognostic scoring systems like the International Prognostic Scoring System (IPSS) and its revised version (IPSS-R) have been used to stratify patients into risk categories that guide clinical management. These classifications consider variables such as blast percentage, number and severity of cytopenias, and cytogenetic abnormalities. The evolving molecular landscape—with recurrent somatic mutations in genes such as TET2, SF3B1, ASXL1, and TP53—has extended our understanding of disease biology and prompted proposals for integrating mutational data into risk models, as exemplified by the recently developed IPSS-M. Thus, MDS is identified not only by its clinical and hematologic features but also by an increasingly rich genetic signature that defines subtypes and guides treatment decisions.

Current Treatment Landscape 
Historically, treatment for MDS has centered on supportive care measures including red blood cell transfusions, treatment with erythropoiesis-stimulating agents (ESAs), iron chelation, and, in select patients, immunosuppressive therapy. For patients with lower-risk disease—especially those harboring the 5q deletion—lenalidomide has become an established treatment option. In higher-risk MDS, hypomethylating agents such as azacitidine and decitabine are used although outcomes remain modest for many patients. These conventional therapies largely aim to relieve cytopenias, reduce transfusion dependence, and delay the risk of transformation to acute myeloid leukemia (AML). Nevertheless, despite decades of research, the clinical outcomes in MDS have been limited by the underlying biological heterogeneity of the disease and the modest improvements in survival offered by these treatments, prompting a strong impetus to develop new, more effective drugs that target specific molecular pathways involved in MDS pathogenesis.

Recent Drug Developments for MDS

Newly Approved Drugs 
Recent years have witnessed significant advances in drug development for MDS, with several new agents emerging on the market, designed to address the specific unmet needs of the disease. Among these, luspatercept stands out as a recently approved drug specifically designed to target anemia in lower-risk MDS patients with ring sideroblasts who are refractory to ESAs. Luspatercept is a recombinant fusion protein that acts as a ligand trap for select members of the transforming growth factor-beta (TGF-β) superfamily. By sequestering ligands that normally inhibit late-stage erythroid maturation, luspatercept improves red blood cell production and has demonstrated robust clinical activity with a favorable safety profile in this subset of patients. 

Another important development is the approval of the oral fixed-dose combination of decitabine and cedazuridine (brand name INQOVI® in the United States, Canada, and Australia) for the treatment of MDS. This combination was designed to overcome the first-pass metabolism issues that limit the bioavailability of decitabine when given orally. The inclusion of cedazuridine, a cytidine deaminase inhibitor, allows oral decitabine to achieve pharmacokinetic exposures equivalent to intravenous decitabine, thereby offering a more convenient treatment option with similar efficacy and manageable adverse events. 

Lenalidomide, once a mainstay in MDS treatment for patients with del(5q), continues to be an essential component of the therapeutic landscape. Although it has been in use for some time, its role has been further refined by ongoing clinical trials and mechanistic insights that have rekindled interest in its optimal sequencing and combination with other agents. 

Drugs in Clinical Trials 
Beyond these newly approved drugs, a robust pipeline of investigational agents is currently undergoing clinical evaluation. Many of these drugs aim to address the limitations of conventional therapies and to provide a more targeted approach based on the molecular profile of the disease. Among the drugs in clinical trials are novel hypomethylating agents and second-generation formulations intended to enhance efficacy, specificity, and ease of administration. For example, several studies are investigating extended dosing schedules or combination regimens of hypomethylating agents with other targeted therapies to overcome resistance mechanisms. 

Imetelstat, a telomerase inhibitor, has garnered significant attention in clinical trials for lower-risk MDS. Telomerase reactivation is a common feature in MDS, and imetelstat functions by inhibiting telomerase activity, which in turn may reduce the proliferative capacity of malignant clones. Early-phase trials have demonstrated promising hematologic improvements and durable responses in certain patient subsets. 

Roxadustat, a hypoxia-inducible factor (HIF) prolyl hydroxylase inhibitor, is another agent under study in MDS. Although originally developed for the treatment of anemia in chronic kidney disease, roxadustat may have a role in MDS by stimulating erythropoiesis through modulation of the HIF pathway, thereby increasing endogenous erythropoietin production and improving iron utilization. 

In addition, immunotherapeutic approaches are being incorporated into clinical trial designs. Monoclonal antibodies and immune checkpoint inhibitors—targeting molecules such as CD47 (with agents like magrolimab) and TIM3 (with agents like sabatolimab)—are being evaluated in combination with standard hypomethylating agents to enhance anti-tumor immunity and improve survival outcomes in higher-risk MDS patients. Venetoclax, a BCL-2 inhibitor widely used in other hematologic malignancies, is also under investigation in combination with hypomethylating agents for MDS, especially to potentially overcome resistance and improve the depth of responses. 

These investigational therapies are being assessed not only as single agents but frequently in various combination regimens aimed at targeting multiple pathogenic pathways simultaneously to achieve synergistic effects, improve response duration, and ultimately enhance overall survival.

Mechanisms of Action

Biological Targets 
The new drugs for MDS target a wide array of biological pathways implicated in the pathogenesis of the disease. Luspatercept, for example, targets the TGF-β signaling pathway—a critical mechanism that inhibits late-stage erythroid maturation. By binding to ligands such as GDF11 and activin B, luspatercept releases the inhibitory brake on erythroid differentiation, thereby improving red blood cell production and alleviating anemia in appropriate MDS patients. 

The oral decitabine/cedazuridine combination works by targeting the epigenetic machinery. Decitabine, a nucleoside analog, gets incorporated into DNA and inhibits DNA methyltransferases, leading to hypomethylation and subsequent reactivation of silenced genes that control differentiation and apoptosis. Cedazuridine’s role in this combination is to inhibit cytidine deaminase in the gut and liver, thus preventing the rapid degradation of decitabine and enhancing its bioavailability. 

Imetelstat targets telomerase, an enzyme that maintains telomere length and is abnormally active in many MDS clones. By binding to the RNA template of telomerase, imetelstat causes telomere shortening over successive cell cycles, leading to apoptosis and decreased proliferative capacity of malignant cells. 

Other investigational agents such as roxadustat act on the HIF pathway. By inhibiting HIF prolyl hydroxylase enzymes, roxadustat stabilizes HIF-α subunits, leading to increased endogenous erythropoietin production and improved erythropoiesis. This mechanism may offer benefits in patients where traditional ESAs have failed. 

Immune-directed therapies, including magrolimab and sabatolimab, target key regulators of the immune response. Magrolimab targets CD47, a “don’t eat me” signal overexpressed on MDS cells; by blocking this signal, macrophages can effectively phagocytose and eliminate malignant cells. Sabatolimab, on the other hand, interferes with the TIM3–galectin-9 interaction, thereby modulating T-cell function and potentially enhancing anti-tumor immunity. Venetoclax, a BCL-2 inhibitor, works by restoring apoptotic processes in MDS cells that have overexpressed anti-apoptotic proteins, allowing for controlled cell death and potential reduction of the malignant clone. 

Pharmacodynamics and Pharmacokinetics 
Each of these drugs exhibits unique pharmacodynamic and pharmacokinetic properties that are tailored to their mechanisms of action. For instance, the pharmacokinetic profile of the oral decitabine/cedazuridine combination mirrors that of intravenous decitabine. Cedazuridine’s inhibition of cytidine deaminase ensures that decitabine reaches therapeutic concentrations in the bloodstream, providing predictable exposure and facilitating an oral formulation that is equally effective as its intravenous counterpart. 

In the case of luspatercept, its long half-life and the sustained modulation of TGF-β ligands contribute to its ability to produce durable hematologic responses in clinical scenarios. The pharmacodynamics of luspatercept involve a gradual improvement in erythroid output that correlates with its mechanism of releasing the blockade on terminal erythroid differentiation. 

Imetelstat’s pharmacodynamics rely on its capacity to inhibit telomerase gradually as malignant cells divide. Its dosing regimen is designed to achieve sustained telomerase inhibition, leading to progressive telomere shortening and eventual cell death, while its pharmacokinetic properties ensure that therapeutic levels are maintained over the treatment period. 

For immune therapies such as magrolimab, the pharmacodynamics involve rapid target engagement of CD47 on MDS cells, leading to immediate restoration of macrophage-mediated clearance. The pharmacokinetic parameters of these monoclonal antibodies are characterized by long elimination half-lives, which enable infrequent dosing while maintaining continuous target inhibition. 

Roxadustat’s pharmacokinetic profile is optimized for its oral administration, with a relatively rapid absorption phase and a half-life that supports once-daily dosing. Its pharmacodynamics, determined by the stabilization of HIF-α, lead to increased erythropoietin production and improved iron metabolism over a sustained period, contributing to its therapeutic potential in MDS-related anemia. 

Clinical Efficacy and Safety

Clinical Trial Outcomes 
The clinical outcomes reported in trials of these new drugs for MDS have been encouraging yet varied by mechanism and patient population. Luspatercept, for instance, has demonstrated robust efficacy, with clinical trials showing significant improvement in transfusion independence rates among lower-risk MDS patients with ring sideroblasts who had limited responses to ESAs. In phase II and III studies, patients treated with luspatercept experienced marked increases in hemoglobin levels and reductions in transfusion burden, with response rates that have translated into improved health-related quality of life. 

The oral decitabine/cedazuridine combination has been rigorously evaluated in multiple studies, with pharmacokinetic equivalence to intravenous decitabine demonstrated in phase III trials. Patients receiving this combination have shown comparable overall response rates, similar rates of complete response (CR), and durable responses that mirror those seen with the IV formulation. The convenience of oral dosing further contributes to patient adherence and quality of life. 

In early phase clinical trials, imetelstat has shown potential in reducing transfusion dependence and improving marrow function in select lower-risk MDS patients. Although response rates vary and longer-term data are needed, the evidence of durable responses in a subset of patients underlines the promise of telomerase inhibition as a therapeutic strategy. 

Investigational immunotherapies have also reported early positive signals. Trials combining magrolimab with hypomethylating agents have indicated enhanced anti-leukemic activity and favorable response kinetics in higher-risk MDS patients. Similarly, studies evaluating sabatolimab in combination regimens have reported preliminary efficacy data that support further investigation. 

Roxadustat’s efficacy in the MDS setting is still under exploration, but early signals suggest that its ability to stimulate endogenous erythropoietin production and improve iron utilization can lead to hematologic improvements in patients who are refractory to conventional ESAs. 

Side Effects and Risk Assessment 
Although the new drugs have shown promising clinical efficacy, each agent is associated with its own set of adverse effects and potential risks. Luspatercept is generally well tolerated, with the most common side effects being mild to moderate in severity and including fatigue, headache, and musculoskeletal pain. Importantly, the rate of significant myelosuppression appears lower than that observed with traditional hypomethylating agents, making it an attractive option for patients with less aggressive disease. 

The safety profile of the oral decitabine/cedazuridine combination closely resembles that of intravenous decitabine, with common adverse events including neutropenia, thrombocytopenia, and gastrointestinal disturbances. However, the oral formulation has not shown any unexpected toxicities, and the adverse event profile is manageable with dose modifications and supportive care measures. 

Imetelstat’s adverse effects include cytopenias and liver function abnormalities, which require careful monitoring during treatment. As with other agents that target proliferative pathways, there remains a potential risk for off-target effects that could exacerbate marrow failure or impact non-malignant tissues. 

For immunotherapies such as magrolimab and sabatolimab, infusion-related reactions, immune-mediated adverse events, and infections are potential concerns. These therapies, by modulating the immune system, might increase the risk of autoimmunity or lead to unexpected inflammatory responses. Rigorous monitoring protocols and prophylactic measures are therefore integral parts of clinical trial designs for these agents. 

Roxadustat has been associated with adverse events related to its mechanism of action, such as hypertension, thromboembolic events, and gastrointestinal symptoms, although the overall safety profile appears acceptable when used within its approved dosing parameters in chronic kidney disease populations. Ongoing studies in MDS will clarify its risk–benefit balance in this new setting. 

Future Directions in MDS Treatment

Emerging Therapies 
The future of drug development for MDS is centered on further refining targeted therapies and combining modalities to address the multifactorial nature of the disease. Emerging agents include a second generation of hypomethylating agents with improved specificity and alternative dosing regimens designed to overcome resistance and extend durability of responses. Novel targeted agents such as APR-246, which aims to reactivate mutant TP53, are being explored particularly in high-risk patients with poor prognoses, and early data suggest that combining these with standard agents may improve outcomes. 

Immunotherapeutic strategies continue to evolve, with emerging trials investigating multiagent regimens that combine immune checkpoint inhibitors, monoclonal antibodies, and other agents that modulate the tumor microenvironment. For example, continued development of CD47 inhibitors like magrolimab in combination with hypomethylating agents holds promise for disrupting the “don’t eat me” signal and enhancing immune clearance of malignant cells. Additionally, agents targeting splicing machinery mutations and other novel genetic aberrations are on the horizon, reflecting the ongoing integration of genomic data into therapeutic strategies. 

Gene therapy and RNA-based therapies also represent a frontier for MDS treatment. Advances in next-generation sequencing and single-cell analyses are opening the door to tailoring treatments not just based on risk stratification, but also on the individual mutational landscape. Such approaches could lead to the development of personalized vaccines or adoptive cellular therapies that directly target the malignant clone or restore normal hematopoiesis. 

Challenges and Research Opportunities 
Despite significant advances, several challenges persist in the development of new drugs for MDS. The heterogeneity of the disease remains a primary obstacle, as differences in genetic, epigenetic, and microenvironmental factors contribute to variable responses to therapy. This complexity necessitates robust biomarker-driven approaches to identify the most appropriate candidates for specific treatments and to monitor therapeutic responses over time. 

Another challenge is managing the toxicity profiles of these new agents while maximizing therapeutic benefits. Although many novel drugs have more targeted mechanisms of action than traditional chemotherapeutics, their potential to disrupt normal hematopoiesis or induce immune-mediated adverse events requires careful study. Future research must focus on developing predictive models for adverse event risk and incorporating these into clinical decision-making frameworks. 

The integration of multi-agent regimens also presents both opportunities and challenges. While the combination of agents with different mechanisms of action holds the potential for synergistic effects and improved response rates, the increased complexity of such regimens makes it essential to design trials that can parse out individual agent contributions and optimize dosing schedules. Adaptive trial designs and the use of real-world evidence may help to accelerate this process. 

Furthermore, emerging research endeavors are increasingly focusing on real-life patient populations, including older patients and those with comorbidities who are often underrepresented in clinical trials. This inclusive approach is critical for ensuring that new therapies translate into meaningful improvements in survival and quality of life across the entire spectrum of patients with MDS. 

Conclusion 
In summary, the landscape of new drug development for myelodysplastic syndromes is advancing rapidly, driven by an improved understanding of the disease’s genetic and epigenetic basis and a commitment to refining treatment approaches. Newly approved therapies such as luspatercept and the oral decitabine/cedazuridine combination have already begun to change the treatment paradigm by offering more specific mechanisms of action and greater convenience for patients. In parallel, a wide array of drugs in clinical trials—including imetelstat, roxadustat, and various immunotherapeutic agents—are being investigated with the goal of overcoming the limitations of conventional therapy and addressing the underlying pathogenic pathways of MDS.

Different classes of drugs target distinct biological mechanisms. Luspatercept improves erythroid maturation via TGF-β ligand trapping, while decitabine/cedazuridine targets aberrant DNA hypermethylation to restore normal gene expression. Other agents, such as imetelstat, inhibit telomerase, a key driver of malignant cell longevity, and emerging immunotherapies work by dismantling cancer’s immune evasion strategies. Detailed pharmacodynamic and pharmacokinetic research underscores how these drugs achieve desired therapeutic exposures while maintaining an acceptable safety profile.

Clinical trials have underscored the efficacy of these interventions, with notable improvements in response rates, transfusion independence, and overall quality of life. However, challenges remain due to the inherent heterogeneity of MDS, the need for predictive biomarkers to tailor treatment, and the balancing act between efficacy and toxicity. The future of MDS treatment lies in the development of personalized regimens that combine conventional therapies with novel targeted agents, adaptive trial designs, and real-world data integration to optimize outcomes for all patient subsets.

Ultimately, while significant progress has been made, ongoing research and collaborative efforts are crucial for fully harnessing the potential of these new drugs. The integration of molecular diagnostics with innovative treatment strategies promises an era of more effective, less toxic, and truly personalized therapies for patients suffering from this challenging group of diseases. The continuous expansion of our therapeutic arsenal and the refinement of treatment paradigms represent hopeful prospects for improving survival and quality of life in MDS patients, marking an exciting time in the field of hematologic oncology.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.