Myelodysplastic syndromes (MDS) have historically been challenging diseases for drug development due to their biology, preclinical modeling, and the affected patient population. In April 2022, the FDA convened a panel of regulators and academic experts in MDS to discuss approaches to improve MDS drug development. The panel reviewed challenges in MDS clinical trial design and endpoints and outlined considerations for future trial design in MDS to facilitate drug development to meaningfully meet patient needs. Challenges for defining clinical benefit in patients with MDS include cumbersome response criteria, standardized transfusion thresholds, and application and validation of patient reported outcome instruments. Clinical trials should reflect the biology of disease evolution, the advanced age of patients with MDS, and how patients are treated in real-world settings to maximize the likelihood of identifying active drugs. In patients with lower-risk disease, response criteria for anemic patients should be based on baseline transfusion dependency, improvement in symptoms, and quality of life. For higher-risk patients with MDS, trials should include guidance to prevent dose reductions or delays that could limit efficacy, specify minimal durations of treatment (in the absence of toxicity or progression), and have endpoints focused on overall survival and durable responses. MDS trials should be designed from the outset to allow the practicable application of new therapies in this high-needs population, with drugs that can be administered and tolerated in community settings, and with endpoints that meaningfully improve patients’ lives over existing therapies.

Despite increased understanding of the pathobiology and molecular underpinnings of myelodysplastic syndromes (MDS), therapeutic options remain limited. To date, only five drugs have been approved by the FDA for patients with MDS or subsets of MDS: azacitidine, lenalidomide, decitabine, luspatercept, and decitabine-cedazuridine tablets (1–5). Three of these are hypomethylating agents (HMA), which remain the standard of care for higher-risk patients with MDS (HR-MDS) and lower-risk patients with multiple cytopenias requiring therapy. Unfortunately, less than half of patients respond to HMAs, and even the best responses are transient and noncurative without hematopoietic cell transplantation (HCT; ref. 6). This standard of care has not changed for nearly two decades (Fig. 1), underscoring a need for new therapies.

Figure 1.

FDA approvals for MDS over time.

Figure 1.

FDA approvals for MDS over time.

Close modal

Recognizing this need, in April 2022, the FDA brought together a panel of regulators and academic experts in MDS to discuss approaches to improve MDS drug development. At this meeting, the panel reviewed challenges in MDS clinical trial design and endpoints and outlined considerations for future trial design in MDS to facilitate drug development to meaningfully meet patient needs.

MDS has historically been a challenging disease for drug development, for a variety of reasons. MDS is characterized by significant heterogeneity in pathobiology due to its genetic landscape (with a spectrum of over 50 myeloid mutations identified in patients with MDS; ref. 7). Consequently, outcomes of affected patients, who tend to be older, have comorbidities, and face logistical challenges in trial participation, are also heterogeneous (8, 9).

Unlike many other cancers, presentations can vary from incidentally found low, but relatively stable, blood counts, to a sudden diagnosis requiring urgent intervention, similar to acute myeloid leukemia (AML). Reflecting this, survival ranges from a few months to a decade or more (10). In addition, preclinical testing of drugs and new agents in MDS has been challenging due to the historic lack of animal models that resemble human MDS, or that can faithfully recapitulate the same complex biology to a degree high enough to reliably predict success or failure of an agent in humans (11).

Even when translated to the clinical setting, trial designs may not be amenable to the patients needing the drugs. For example, the standard management of HR-MDS includes repeated monthly cycles of 5 to 7 consecutive days of injectable HMAs, which have been shown prospectively to improve outcomes and are recommended to be continued ad infinitum, as long as the patient is responding and not experiencing prohibitive toxicities. Therefore, most clinical trials of new drugs for patients with HR-MDS typically combine another agent to an HMA backbone. However, as the median age at diagnosis of MDS is typically the eighth decade of life, and patients often have poor functional status, multiple comorbidities, and/or limited social support, trial participation at tertiary centers requiring multiple visits monthly becomes prohibitive (12). From a practical standpoint, nonspecialty settings may also be challenged to manage the adverse events and supportive care needs of vulnerable patients treated aggressively.

Most clinical trials for MDS are conducted in large centers, while the majority of patients with MDS in the United States are treated in community settings. As MDS is rare (with an estimated yearly incidence rate of 4/100,000 people), clinical trials often underaccrue and take a long time to complete, discouraging industry sponsors from conducting trials in patients with MDS until agents have been explored in other cancers traditionally managed at larger centers, such as AML. Add to this complexity the post-HMA failure setting, where patients are typically too sick either to enroll in clinical trials or remain on treatment long enough to realize a potential benefit. Drug development in this setting is further challenged by a lack of understanding of resistance mechanisms that lead to HMA failure, precluding rational drug design targeting specific mechanisms.

The landscape of therapeutics in MDS remains unchanged, as underscored by the failure of multiple large, randomized trials conducted in HR-MDS because the FDA approval of azacitidine due to an inability to demonstrate an improvement in outcomes over azacitidine, or after HMA failure (13–16). Compare this therapeutic lag with other blood cancers: in AML, for example, nine drugs have been approved since 2017. Clearly, we need to rethink clinical trial design specifically for MDS.

Challenges and considerations for future trial design in MDS

Clinical trials for patients with MDS should be designed to reflect the biology of disease evolution, including molecular mutations and overlap with other disorders; the advanced age of patients with MDS and how patients are treated in real-world settings; and to maximize the likelihood of identifying truly active drugs (Table 1).

Table 1.

Key considerations for MDS trial design.

Phase I/II 
  • Emphasize dose optimization to prevent excess toxicity in later-phase trials

 
Phase II/III 
  • Randomize whenever possible, to adjust for placebo or transfusion effect (double-blinded design preferable)

  • For combination trials with HMA, consider allowing local administration of HMA for days when patients are receiving HMA alone without the investigational drug

  • Consider early engagement of community oncology practices in clinical trials to aid with patient recruitment and future roll-out postapproval

 
All phases 
  • Select endpoints that are clinically meaningful

  • For lower-risk MDS trials, consider endpoints that capture trilineage count recovery and durable transfusion benefit, with supportive improvement in patient-reported outcomes and stratification by baseline cytopenias and transfusion status (as applicable)

  • For higher-risk MDS trials, consider endpoints that reflect modification of the natural history of the disease, such as overall survival or durable CR. Alternative endpoints should be prospectively validated.

  • Consider MRD assessment and bridge to transplant as exploratory endpoints.

  • Define relapsed/refractory MDS distinctly from treatment-naïve MDS

  • Consider inclusion of CCUS in lower-risk MDS trials if appropriate and with adequate stratification

  • Consider inclusion of patients with lower-risk MDS on higher-risk MDS trials, if otherwise eligible for HMA therapy

  • Consider inclusion of oligoblastic AML in higher-risk MDS trials if appropriate and with adequate stratification

  • Eliminate “transplant ineligible” language from eligibility criteria

  • Eliminate use of IPSS/IPSS-R score for inclusion to trials following HMA failure

  • Include strict protocol language to limit dose adjustments or delays of the HMA backbone for patients on HMA-based therapy for a minimum of six treatment cycles or until treatment response

  • Incorporate NGS to help identify genetic or biologic MDS subsets that may benefit from an intervention

 
Phase I/II 
  • Emphasize dose optimization to prevent excess toxicity in later-phase trials

 
Phase II/III 
  • Randomize whenever possible, to adjust for placebo or transfusion effect (double-blinded design preferable)

  • For combination trials with HMA, consider allowing local administration of HMA for days when patients are receiving HMA alone without the investigational drug

  • Consider early engagement of community oncology practices in clinical trials to aid with patient recruitment and future roll-out postapproval

 
All phases 
  • Select endpoints that are clinically meaningful

  • For lower-risk MDS trials, consider endpoints that capture trilineage count recovery and durable transfusion benefit, with supportive improvement in patient-reported outcomes and stratification by baseline cytopenias and transfusion status (as applicable)

  • For higher-risk MDS trials, consider endpoints that reflect modification of the natural history of the disease, such as overall survival or durable CR. Alternative endpoints should be prospectively validated.

  • Consider MRD assessment and bridge to transplant as exploratory endpoints.

  • Define relapsed/refractory MDS distinctly from treatment-naïve MDS

  • Consider inclusion of CCUS in lower-risk MDS trials if appropriate and with adequate stratification

  • Consider inclusion of patients with lower-risk MDS on higher-risk MDS trials, if otherwise eligible for HMA therapy

  • Consider inclusion of oligoblastic AML in higher-risk MDS trials if appropriate and with adequate stratification

  • Eliminate “transplant ineligible” language from eligibility criteria

  • Eliminate use of IPSS/IPSS-R score for inclusion to trials following HMA failure

  • Include strict protocol language to limit dose adjustments or delays of the HMA backbone for patients on HMA-based therapy for a minimum of six treatment cycles or until treatment response

  • Incorporate NGS to help identify genetic or biologic MDS subsets that may benefit from an intervention

 

Increasingly, molecular characterization of MDS and incorporation of molecular mutations into clinicopathologic scoring systems has become standard, with increased accuracy of molecular systems in predicting leukemia transformation and overall survival (OS) and almost 50% of patients being restratified into risk categories that differ from those suggested by the International Prognostic Scoring System (IPSS) or Revised IPSS (IPSS-R) (17, 18). Thus, future MDS clinical trials ideally would include next-generation sequencing (NGS)results, both to better risk stratify patients and to help identify genetic or biologic subsets who could benefit from a therapeutic intervention—or subsets who would not benefit. It is also reasonable for trials to be designed independent of MDS risk stratification (with inclusion of patients with secondary MDS), provided disease severity can be assessed in the context of treatment efficacy and toxicity.

In designing trials for treatment-naïve MDS, one real-world consideration is that patients with MDS initially may be evaluated and started on treatment at local clinics, and then referred to specialty centers for a trial. For such trials, patients could still be considered to have “treatment-naïve” MDS if they have received, for example, a single cycle of HMA or erythropoiesis stimulating agent therapy previously. Once a patient is enrolled in a clinical trial, administration of standard-of-care therapies (including blood product transfusions or drugs such as HMAs considered standard) could be allowed at local treatment locations, as frequent study visits to a specialized center may be a barrier to trial participation. Further engagement of community sites should also be pursued to decentralize clinical trials and develop more practicable therapies for a broad population of patients with MDS across the country.

In addition, clinical trials for MDS should be designed to maximize the chance of identifying truly active drugs. The diagnosis of MDS can be challenging, as is reflected in large databases or in the rate of discrepancy between an MDS diagnosis and alternative diagnoses [such as AML, MDS/myeloproliferative neoplasms (MPN), or nonmalignant conditions] >25% of the time (19, 20). Thus, central review of the MDS diagnosis is needed to reduce these discrepancies on trials and should include risk stratification.

It has long been recognized that older adults who develop MDS may have comorbidities associated with anemia, which can complicate efficacy assessments of MDS drugs intended to correct anemia (21). Consequently, randomization in advanced phase trials is needed to adjust for this effect.

Multiple trials in higher-risk patients (including histone deacetylase inhibitors or immunomodulatory drugs combined with HMAs) have demonstrated that dose reductions, delays or premature cessation of HMA-based therapy may have compromised efficacy (13, 14, 22, 23). On the other hand, in the recent randomized trial of the NEDD8 activating enzyme pevonedistat combined with azacitidine versus azacitidine monotherapy, strict protocol language limiting dose adjustments led to a 32% complete response (CR) rate for the control arm (azacitidine monotherapy; ref. 13). Thus, protocols should include instructions to maintain dose, avoid cycle delays, and continue HMA-based therapy for a minimum of six treatment cycles (in the absence of clear progression or unacceptable toxicity) prior to determining failure of response.

For most patients with HR-MDS, the standard backbone of therapy includes an HMA, which as monotherapy results in a typical median response rate (complete + partial) of approximately 20%, and response duration and survival of approximately 18 months (24, 25). The paradigm of recent clinical therapeutic trial interventions has been to introduce combination therapies upfront, one being an HMA, to improve on these mediocre outcomes (i.e., add-on trials). However, a head-to-head comparison versus HMA alone could be considered for agents with compelling monotherapy data in the relapsed or refractory setting. As patients with HR-MDS have a median survival often measured in months, trials should ideally be powered to show differences in treatment arms based on an appropriate response rate, duration, quality of life, and survival in the comparator arm based upon data from control arms of recent clinical trials (12, 26).

Considerations for eligibility criteria in MDS trials

MDS is typically divided into lower- and higher-risk (LR- and HR-MDS) categories commonly defined by the IPSS-R, which serves as a default staging system, but only for newly diagnosed patients (10). Consequently, trials are often constructed for patients with either lower- or higher-risk disease, with eligibility for a given trial being mutually exclusive for one or the other.

However, disease severity can evolve as patients acquire additional molecular mutations or progress through multiple lines of therapy (7, 27)—aspects of advanced MDS that may not be captured through existing prognostic scoring systems, and that make HCT or intensive therapies viable options (28, 29). Thus, clinical trials should avoid eligibility language referencing a patient's potential for HCT, as this is variably interpreted.

“Relapsed or refractory” MDS should be defined distinctly from treatment-naïve MDS, and consistently across trials (e.g., for patients with lower-risk disease, previous exposure for a defined time period to erythropoiesis stimulating agents, and/or to prior immunomodulatory or HMAs; for higher-risk disease, previous exposure to hypomethylating agents for a defined time period and/or prior HCT).

The very definition of MDS is evolving, as data suggest that patients with clonal cytopenias of undetermined significance (CCUS) with discrete molecular abnormalities have similar outcomes to patients with LR-MDS, and that patients with HR-MDS with excess blasts have similar outcomes to patients with oligoblastic AML (30, 31). This is now reflected in recent revisions in classification of myeloid neoplasms from the World Health Organization and the International Consensus Classification, which further blur the line between MDS and AML, and identify discrete categories of MDS based on molecular profile (31, 32). In contradistinction, patients with certain MDS/MPN, such as chronic myelomonocytic leukemia, may not have similar outcomes or drug responsiveness as pure MDS (13, 14). Thus, trials for patients with LR-MDS may consider allowing enrollment of patients with CCUS, and trials for patients with HR-MDS may consider allowing enrollment of patients with oligoblastic leukemia (particularly if targeting a common mutation; ref. 31), provided patients in either scenario are otherwise eligible, can be stratified, and treatment and endpoints can be harmonized.

In addition, patients in HR-MDS trials should reflect the real-world MDS population—the majority of whom are over age 60 years and with managed medical comorbidities. Patients should not be excluded from trial eligibility based on prior therapy for lower-risk disease that is distinct from the therapy being evaluated for HR-MDS. Patients potentially eligible for HCT should also be included as noted above.

Recommendations for clinically meaningful endpoints for LR-MDS

Patients with LR-MDS remain a heterogenous group with a variable degree of presenting cytopenias and clinical outcomes. Morbidity and mortality in this group are mainly related to cytopenia complications or interplay with other comorbidities. In addition, approximately one-third of lower-risk patients progress to have HR-MDS or AML (33, 34). Thus, clinically meaningful endpoints would capture important treatment effects such as alleviating cytopenias associated with symptoms and morbidity, preventing disease progression, or improving OS. Moreover, such endpoints should be practical, with applicability both to pivotal clinical trials and to clinical practice to assess patient benefit objectively. As such, we propose that the response criteria for patients with LR-MDS should be based on the predominant cytopenia or clinical feature—similar to a “target lesion” in solid tumor trials.

In LR-MDS, the clinical manifestations of anemia dominate the initial presentation in 85% of patients and drive the need for therapeutic intervention (35). Regular red blood cell (RBC) transfusion requirements are associated with a worse prognosis, and hemoglobin (Hgb) levels below 9 g/dL in men and 8 g/dL in women are associated with reduced OS and a higher risk of nonleukemic and cardiac death (36). However, existing criteria defining achievement of transfusion independence or lack of transfusion dependence are problematic.

For example, despite the arbitrary threshold for transfusion of RBCs for a Hgb ≤ 9 g/dL specified in International Working Group (IWG) 2006, most clinicians do not give a transfusion until patients are symptomatic or for a Hgb < 7–8 g/dL, likely adopting thresholds from trials in patients with septic shock, surgical considerations, or inpatient therapy, and from concerns for iron overload, transfusion burden, and product availability (37–40). Fairness and use of limited resources are issues that have been highlighted during the COVID-19 pandemic where global blood shortages affected when transfusions could occur.

A proposal enumerating stricter criteria for defining hematologic improvement (HI) has provided initial guidance for improving definitions, and for making them more clinically meaningful (41). The IWG 2006 criteria require receipt of at least 4 units of packed red blood cells for a Hgb <9 g/dL within an 8-week period to define transfusion dependence. The 2018 modified criteria called for this period to be extended to 16 weeks and to stratify patients as not transfusion dependent, low transfusion dependent and high transfusion dependent—to better reflect the heterogeneity of anemia burden in patients with MDS, and to set the stage for defining achievement of transfusion independence that must last 16 weeks or longer.

For patients who are not transfusion dependent (NTD; defined as no RBC transfusion in the 16 weeks prior to starting treatment), initiation of therapy can be considered if patients are symptomatic and have a Hgb ≤ 9 g/dL. Following the IWG 2018 response criteria (41), an objective Hgb increase of at least 1.5 g/dL for 16 weeks or more may be considered clinically meaningful only if associated with objective improvement in quality of life using patient reported outcomes (PRO) tools such as the MDS-QUALMS (42) or significant prolongation of time-to-transfusion dependency/transfusion-free survival.

For patients with low transfusion burden (LTB; defined as 3–7 RBCs transfusions in 16 weeks with at least two separate transfusion episodes and a maximum of three in 8 weeks), a clinically meaningful response is defined as complete elimination of RBC transfusion dependence for 16 weeks or more. Assessment of PRO benefit may be more challenging in this group as RBC transfusions in a comparison group not receiving active therapy may alleviate many anemia-related symptoms, thus potentially leveling PRO assessments between study arms.

For patients with high transfusion burden (defined as ≥8 RBCs in 16 weeks, ≥4 in 8 weeks), complete elimination of RBC transfusions for 16 weeks or more is a meaningful endpoint. A ≥50% reduction in transfusion burden sustained for 16 weeks or more may be clinically meaningful if objectively associated with improvement in quality of life as measured by validated PRO tools.

In patients with isolated or predominating thrombocytopenia, platelet improvement should be associated with no bleeding events and/or platelet transfusions. The definition of a complete platelet response includes a platelet count of at least 100 × 10⁹/L without bleeding events.

For patients with isolated neutropenia, treatment should not be initiated merely based on neutrophil count. Clinically meaningful response could include a reduction in recurrent infections, an endpoint that would need to be validated in a randomized trial.

In LR-MDS trials, PROs also have the potential to identify therapies with biological activity that results in clinical benefit. How a patient feels (symptom relief) and functions (increased daily activity) are critical indicators of clinical benefit that are not required by established response criteria alongside achievement of HI. Yet, capturing symptom improvement from a patient's perspective was a key secondary endpoint that contributed to the approval of ruxolitinib in the COMFORT I–II trials in myelofibrosis, and this endpoint was also included in the prescribing information (43, 44). Employing a tool that accurately captures symptom improvement tracking with disease improvement, and that can distinguish improvement between treatment and control trial arms, would be useful.

Future clinical trials should take advantage of advances in technology for assessing PRO improvement using tools such as wearables and make efforts to assess objective data on the impact of disease through health care resource use endpoints. Furthermore, clinical trials using targeted therapies need to assess cytogenetic and molecular responses by measuring minimal residual disease (MRD) to develop this endpoint as a potential surrogate marker of clinical benefit.

Of note, HI and PRO endpoints have not been used to support efficacy for new MDS therapies to date. Thus, the proposed endpoints would need to be validated in the setting of prospective, randomized controlled trials along with appropriate stratification by baseline cytopenias and transfusion status, as applicable. Clinical trial designs would need to carefully define the intended patient population as well as stipulate an analysis plan that rigorously tests the selected efficacy endpoints. For HI endpoints targeting a single cell line, care would need to be taken to “do no harm,” ensuring that other cell lines are not negatively impacted by the therapy, particularly for patients who are NTD or LTB at baseline where the margin of benefit is smaller.

In addition, among patients risk stratified as LR-MDS, the CR definition applied in HR-MDS can be used similarly as a reasonable marker of benefit. This use of CR with full count recovery may be used as an efficacy endpoint to capture responses while preserving trilineage hematopoiesis.

The ultimate clinically meaningful endpoint—improvement in OS, is more challenging to achieve in LR-MDS, where no approved therapies have prospectively achieved this benefit. As progression to AML is less frequent and survival is longer compared with those with HR-MDS, OS may be a more relevant efficacy endpoint for those whose “low-risk” disease features an adverse risk mutation (biologically contradicting the low-risk designation), such as those in EZH2, RUNX1, TP53, and ASXL1 (45). Furthermore, OS may serve as a critical safety endpoint for randomized LR-MDS studies to ensure there is no increased mortality due to toxicities from the experimental agent.

Recommendations for clinically meaningful endpoints for HR-MDS

A close look at the initial 2000 and subsequent 2006 IWG-modified response criteria for MDS highlights some challenges in drug development (46, 47). One overarching problem related to these response criteria lies in how cumbersome they are to apply or interpret compared with those for AML (48–50).

Meaningful clinical endpoints specific to HR-MDS should be identified starting with earlier-phase studies (such as randomized phase II trials). Primary endpoints should reflect true modification of the natural history of the disease, such as OS. One challenge with event-free survival (EFS), defined by the IWG 2006 as “failure or death from any cause,” is that it is seldom used due to its ambiguity. An “event” in HR-MDS could be defined to signify a demonstrable change in disease biology that is associated with worse outcomes. Future revisions to the IWG response criteria ideally would reflect meaningful clinical benefit to a patient with enhanced practicability, reduced ambiguity, and enriched interobserver consistency. While there is experience (13) using a more specific and alternative EFS definition of “death or progression to AML with >50% increase in blasts to >20%,” which conveys a metric of the lack or loss of benefit from an investigational intervention, use of EFS by any definition requires further evaluation and validation in prospective randomized, controlled trials.

Earlier clinical endpoints could also be considered. CR, as currently defined, lacks distinctness and has not regularly translated to an OS benefit from the phase II to the phase III setting (51). Study drugs added to an azacitidine backbone may be myelosuppressive, making therapy-related cytopenias in the setting of disease resolution difficult to discern from lack of drug benefit. In addition, certain requirements of an MDS CR are stricter than those for an AML CR: a Hgb ≥11 g/dL is required for an MDS CR, whereas no Hgb requirement is specified for AML CR; requiring CR confirmation is also unique to HR-MDS. Finally, the designation of a marrow CR may lead to misleading clinical conclusions. Without concomitant HI, achievement of marrow CR has similar clinical benefit to that of stable disease (52).

We suggest that studies prospectively collect data to evaluate alternative CR definitions using: (i) a decrease in the mandatory Hgb threshold for CR determination to 10 g/dL or consideration of RBC transfusion independence; (ii) CR lasting a clinically meaningful duration of time. For overall response rate (ORR) reporting, this would be limited to true CR patients, along with partial response (PR) and potentially HI (given its inconsistent translation to a meaningful benefit). Marrow CR particularly without HI can be avoided, as the reduction of marrow blasts to less than 5% has no true clinical correlation.

PROs to capture quality of life or symptom improvement are similarly critical for defining clinical benefit in HR-MDS. Real-life data consistently show how clinical outcomes fall short of the those achieved within the context of a clinical trial (25, 53). Patient-specific factors such as frailty and comorbidities have yet to be consistently included in response assessments or adequately captured on clinical trials. Investigational therapies that do not improve disease-related symptom burden may speak to both the drug's ability to truly modify the disease, and to the feasibility of using it in the real world (54).

The MDS field is sensitive to the necessity of interim markers of benefit and secondary endpoints to better reflect the spectrum of improvement. Incorporation of PROs specific to MDS (55) is key. The durability of time without transfusion and improved PRO are also secondary endpoints with clinical relevance, particularly in combination with CR and even PR to demonstrate altered natural history. An endpoint including patients who undergo HCT should also be evaluated. This would be annotated as alive and with disease control that allows the HCT procedure to occur. This endpoint has been evaluated in the AML setting (56) and is increasingly viewed as a measure of modified goals of care, as HCT potentially extends OS. Furthermore, outcomes in HR-MDS trials need not be censored at the time of HCT, as both the bridge to HCT as well as ability to survive past HCT are meaningful.

An area of burgeoning clinical and translational research in HR MDS includes robust specimen banking, requirement to document full molecular panels (NGS), and measurable residual disease assessments. These should be standard in trials to correlate with EFS, OS, and ORR, and to illustrate role the role of certain phenotypes and genotypes more likely to respond to a given treatment.

MDS is a complicated collection of heterogeneous disorders with highly variable presentations and outcomes, and that predominantly affects a vulnerable population of older adults. Trials should be designed from the outset to allow the practicable application of new therapies in this high-needs population, with drugs that can be administered and tolerated in community settings, and with endpoints that meaningfully improve patients’ lives over existing therapies, rather than simply providing supportive care. Trials should also capitalize on our expanding understanding of the biology of these disorders to effect substantive change in patient outcomes.

M.A. Sekeres reports personal fees from BMS, Novartis, and Kurome outside the submitted work. A.E. DeZern reports personal fees from Geron, Novartis, Sobi, and BMS outside the submitted work. A.M. Zeidan reports personal fees from Novartis, AbbVie, Genentech, BMS, Geron, Agios, Syros, Aprea, ASTEX, Kura, Taiho, Chiesi, Takeda, Regeneron, Gilead, Orum, Schrodinger, Syndax, and Ionis outside the submitted work. R.S. Komrokji reports grants and personal fees from BMS and personal fees from Novartis, JAZZ, AbbVie, CTI Biopharma, Geron, PharmaEssentia, Servier, Taiho, Takeda, Gilead, and Rigel outside the submitted work. No disclosures were reported by the other authors.

This article reflects the views of the authors and should not be construed to represent FDA's views or policies.

1.
Celgene
;
U.S. Food and Drug Administration
.
VIDAZA (azacitidine) [drug label]
. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021790s025lbl.pdf.
2.
Celgene
;
U.S. Food and Drug Administration
.
REVLIMID (lenalidomide) [drug label]
. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/021880s065lbl.pdf.
3.
Otsuka
;
U.S. Food and Drug Administration
.
DACOGEN (decitabine) [drug label]
. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021790s025lbl.pdf.
4.
Celgene
;
U.S. Food and Drug Administration
.
REBLOYZYL (luspatercept-aamt) [drug label]
. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/761136s006lbl.pdf.
5.
Otsuka; U.S. Food and Drug Administration
.
INQOVI (decitabine and cedazuridine tablets) [drug label]
. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/212576s000lbl.pdf.
6.
Craddock
C
,
Quek
L
,
Goardon
N
,
Freeman
S
,
Siddique
S
,
Raghavan
M
, et al
.
Azacitidine fails to eradicate leukemic stem/progenitor cell populations in patients with acute myeloid leukemia and myelodysplasia
.
Leukemia
2013
;
27
:
1028
36
.
7.
Makishima
H
,
Yoshizato
T
,
Yoshida
K
,
Sekeres
MA
,
Radivoyevitch
T
,
Suzuki
H
, et al
.
Dynamics of clonal evolution in myelodysplastic syndromes
.
Nat Genet
2017
;
49
:
204
12
,.
8.
Brierley
CK
,
Zabor
EC
,
Komrokji
RS
,
Dezern
AE
,
Roboz
GJ
,
Brunner
AM
, et al
.
Low participation rates and disparities in participation in interventional clinical trials for myelodysplastic syndromes
.
Cancer
2020
;
126
:
4735
43
.
9.
Zeidan
AM
,
Stahl
M
,
Sekeres
MA
,
Steensma
DP
,
Komrokji
RS
,
Gore
SD
.
A call for action: increasing enrollment of untreated patients with higher-risk myelodysplastic syndromes in first-line clinical trials
.
Cancer
2017
;
123
:
3662
72
.
10.
Greenberg
PL
,
Tuechler
H
,
Schanz
J
,
Sanz
G
,
Garcia-Manero
G
,
Sole
F
, et al
.
Revised international prognostic scoring system for myelodysplastic syndromes
.
Blood
2012
;
120
:
2454
65
.
11.
Liu
W
,
Teodorescu
P
,
Halene
S
,
Ghiaur
G
.
The coming of age of preclinical models of MDS
.
Front Oncol
2022
;
12
:
815037
.
12.
Zeidan
AM
,
Salimi
T
,
Epstein
RS
.
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
75
.
13.
Adès
L
,
Girshova
L
,
Doronin
VA
,
Díez-Campelo
M
,
Valcárcel
D
,
Kambhampati
S
, 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
45
.
14.
Sekeres
MA
,
Othus
M
,
List
AF
,
Odenike
O
,
Stone
RM
,
Gore
SD
, 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
53
,
15.
Zeidan
AM
,
Boss
I
,
Beach
CL
,
Copeland
WB
,
Thompson
E
,
Fox
BA
, et al
.
A randomized phase 2 trial of azacitidine with or without durvalumab as first-line therapy for higher-risk myelodysplastic syndromes
.
Blood Adv
2022
;
6
:
2207
18
.
16.
Garcia-Manero
G
,
Fenaux
P
,
Al-Kali
A
,
Baer
MR
,
Sekeres
MA
,
Roboz
GJ
, 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
,.
17.
Bernard
E
,
Tuechler
H
,
Greenberg
PL
,
Hasserjian
RP
,
Ossa
JEA
,
Nannya
Y
, et al
.
Molecular international prognostic scoring system for myelodysplastic syndromes
.
NEJM Evidence
2022
;
1
:
EVIDoa2200008
.
18.
Nazha
A
,
Komrokji
R
,
Meggendorfer
M
,
Jia
X
,
Radakovich
N
,
Shreve
J
, et al
.
Personalized prediction model to risk stratify patients with myelodysplastic syndromes
.
J Clin Oncol
2021
;
39
:
3737
46
.
19.
Zhang
L
,
Stablein
DM
,
Epling-Burnette
P
,
Harrington
AM
,
Moscinski
LC
,
Kroft
S
, et al
.
Diagnosis of myelodysplastic syndromes and related conditions: rates of discordance between local and central review in the NHLBI MDS natural history study
.
Blood
2018
;
132
:
4370
.
20.
Rollison
DE
,
Howlader
N
,
Smith
MT
,
Strom
SS
,
Merritt
WD
,
Ries
LA
, et al
.
Epidemiology of myelodysplastic syndromes and chronic myeloproliferative disorders in the United States, 2001–2004, using data from the NAACCR and SEER programs
.
Blood
2008
;
112
:
45
52
.
21.
Fenaux
P
,
Platzbecker
U
,
Mufti
GJ
,
Garcia-Manero
G
,
Buckstein
R
,
Santini
V
, et al
.
Luspatercept in patients with lower-risk myelodysplastic syndromes
.
N Engl J Med
2020
;
382
:
140
51
.
22.
Garcia-Manero
G
,
Montalban-Bravo
G
,
Berdeja
JG
,
Abaza
Y
,
Jabbour
E
,
Essell
J
, et al
.
Phase 2, randomized, double-blind study of pracinostat in combination with azacitidine in patients with untreated, higher-risk myelodysplastic syndromes
.
Cancer
2017
;
123
:
994
1002
.
23.
Sekeres
MA
,
Watts
J
,
Radinoff
A
,
Sangerman
MA
,
Cerrano
M
,
Lopez
PF
, 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
24
.
24.
Corman
S
,
Joshi
N
,
Wert
T
,
Kale
H
,
Hill
K
,
Zeidan
AM
.
Under-use of hypomethylating agents in patients with higher-risk myelodysplastic syndrome in the United States: a large population-based analysis
.
Clin Lymphoma Myeloma Leuk
2021
;
21
:
e206
11
.
25.
Bernal
T
,
Martinez-Camblor
P
,
Sanchez-Garcia
J
,
de Paz
R
,
Luno
E
,
Nomdedeu
B
, et al
.
Effectiveness of azacitidine in unselected high-risk myelodysplastic syndromes: results from the Spanish registry
.
Leukemia
2015
;
29
:
1875
81
.
26.
Zeidan
AM
,
Davidoff
AJ
,
Long
JB
,
Hu
X
,
Wang
R
,
Ma
X
, et al
.
Comparative clinical effectiveness of azacitidine versus decitabine in older patients with myelodysplastic syndromes
.
Br J Haematol
2016
;
175
:
829
40
.
27.
Jabbour
EJ
,
Garcia-Manero
G
,
Strati
P
,
Mishra
A
,
Al Ali
NH
,
Padron
E
, et al
.
Outcome of patients with low-risk and intermediate-1-risk myelodysplastic syndrome after hypomethylating agent failure: a report on behalf of the MDS clinical research consortium
.
Cancer
2015
;
121
:
876
82
.
28.
Lindsley
RC
,
Saber
W
,
Mar
BG
,
Redd
R
,
Wang
T
,
Haagenson
MD
, et al
.
Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation
.
N Engl J Med
2017
;
376
:
536
47
.
29.
Nakamura
R
,
Saber
W
,
Martens
MJ
,
Ramirez
A
,
Scott
B
,
Oran
B
, 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
39
.
30.
Malcovati
L
,
Gallì
A
,
Travaglino
E
,
Ambaglio
I
,
Rizzo
E
,
Molteni
E
, et al
.
Clinical significance of somatic mutation in unexplained blood cytopenia
.
Blood
2017
;
129
:
3371
8
.
31.
Arber
DA
,
Orazi
A
,
Hasserjian
RP
,
Borowitz
MJ
,
Calvo
KR
,
Kvasnicka
H-M
, et al
.
The international consensus classification of myeloid neoplasms and acute leukemias: integrating morphological, clinical, and genomic data
.
Blood
2022
;
140
:
1200
28
.
32.
Khoury
JD
,
Solary
E
,
Abla
O
,
Akkari
Y
,
Alaggio
R
,
Apperley
JF
, et al
.
The 5th edition of the world health organization classification of haematolymphoid tumours: myeloid and histiocytic/dendritic neoplasms
.
Leukemia
2022
;
36
:
1703
19
.
33.
Jain
AG
,
Ball
S
,
Aguirre
LE
,
Al Ali
N
,
Kaldas
D
,
Tinsley-Vance
S
, et al
.
The natural history of lower risk MDS: factors predicting progression to high-risk myelodysplastic syndrome and acute myeloid leukemia in patients with very low and low risk MDS according to the R-IPSS criteria
.
Blood
2021
;
138
:
2600
.
34.
Komrokji
RS
,
George
TI
,
Erba
HP
,
Scott
BL
,
Grinblatt
DL
,
Maciejewski
JP
, et al
.
Treatment patterns and outcomes of patients with lower-risk myelodysplastic syndromes in the connect ® myeloid disease registry
.
Blood
2021
;
138
:
3686
.
35.
Platzbecker
U
,
Hofbauer
LC
,
Ehninger
G
,
Holig
K
.
The clinical, quality of life, and economic consequences of chronic anemia and transfusion support in patients with myelodysplastic syndromes
.
Leuk Res
2012
;
36
:
525
36
.
36.
Malcovati
L
,
Della Porta
MG
,
Strupp
C
,
Ambaglio
I
,
Kuendgen
A
,
Nachtkamp
K
, et al
.
Impact of the degree of anemia on the outcome of patients with myelodysplastic syndrome and its integration into the WHO classification-based prognostic scoring system (WPSS)
.
Haematologica
2011
;
96
:
1433
40
.
37.
Holst
LB
,
Haase
N
,
Wetterslev
J
,
Wernerman
J
,
Guttormsen
AB
,
Karlsson
S
, et al
.
Lower versus higher hemoglobin threshold for transfusion in septic shock
.
N Engl J Med
2014
;
371
:
1381
91
.
38.
Hebert
PC
,
Carson
JL
.
Transfusion threshold of 7 g per deciliter–the new normal
.
N Engl J Med
2014
;
371
:
1459
61
.
39.
Dezern
AE
,
Williams
K
,
Zahurak
M
,
Hand
W
,
Stephens
RS
,
King
KE
, et al
.
Red blood cell transfusion triggers in acute leukemia: a randomized pilot study
.
Transfusion
2016
;
56
:
1750
7
.
40.
Carson
JL
,
Terrin
ML
,
Noveck
H
,
Sanders
DW
,
Chaitman
BR
,
Rhoads
GG
, et al
.
Liberal or restrictive transfusion in high-risk patients after hip surgery
.
N Engl J Med
2011
;
365
:
2453
62
.
41.
Platzbecker
U
,
Fenaux
P
,
Ades
L
,
Giagounidis
A
,
Santini
V
,
van de Loosdrecht
AA
, et al
.
Proposals for revised IWG 2018 hematological response criteria in patients with MDS included in clinical trials
.
Blood
2019
;
133
:
1020
30
.
42.
Abel
GA
,
Hebert
D
,
Lee
C
,
Foran
JM
,
Gore
SD
,
Saber
W
, et al
.
Patient-reported outcomes and frailty among participants in the NHLBI MDS natural history study
.
Blood
2020
;
136
:
15
16
.
43.
Harrison
C
,
Kiladjian
JJ
,
A-A
HK
,
Gisslinger
H
,
Waltzman
R
,
Stalbovskaya
V
, et al
.
JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis
.
N Engl J Med
2012
;
366
:
787
98
.
44.
Verstovsek
S
,
Mesa
RA
,
Gotlib
J
,
Levy
RS
,
Gupta
V
,
DiPersio
JF
, et al
.
A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis
.
N Engl J Med
2012
;
366
:
799
807
.
45.
Bejar
R
,
Stevenson
KE
,
Caughey
BA
,
Abdel-Wahab
O
,
Steensma
DP
,
Galili
N
, et al
.
Validation of a prognostic model and the impact of mutations in patients with lower-risk myelodysplastic syndromes
.
J Clin Oncol
2012
;
30
:
3376
82
,
46.
Cheson
BD
,
Bennett
JM
,
Kantarjian
H
,
Pinto
A
,
Schiffer
CA
,
Nimer
SD
, et al
.
Report of an international working group to standardize response criteria for myelodysplastic syndromes
.
Blood
2000
;
96
:
3671
4
.
47.
Cheson
BD
,
Greenberg
PL
,
Bennett
JM
,
Lowenberg
B
,
Wijermans
PW
,
Nimer
SD
, et al
.
Clinical application and proposal for modification of the international working group (IWG) response criteria in myelodysplasia
.
Blood
2006
;
108
:
419
25
.
48.
Dohner
H
,
Estey
E
,
Grimwade
D
,
Amadori
S
,
Appelbaum
FR
,
Buchner
T
, et al
.
Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel
.
Blood
2017
;
129
:
424
47
.
49.
Cheson
BD
,
Bennett
JM
,
Kopecky
KJ
,
Buchner
T
,
Willman
CL
,
Estey
EH
, et al
.
Revised recommendations of the international working group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia
.
J Clin Oncol
2003
;
21
:
4642
9
.
50.
Döhner
H
,
Wei
AH
,
Appelbaum
FR
,
Craddock
C
,
DiNardo
CD
,
Dombret
H
, et al
.
Diagnosis and management of acute myeloid leukemia in adults: 2022 recommendations from an international expert panel, on behalf of the European LeukemiaNet
.
Blood
2022
;
140
:
1345
77
.
51.
Sallman
DA
,
DeZern
AE
,
Garcia-Manero
G
,
Steensma
DP
,
Roboz
GJ
,
Sekeres
MA
, et al
.
Eprenetapopt (APR-246) and azacitidine in TP53-mutant myelodysplastic syndromes
.
J Clin Oncol
2021
;
39
:
1584
94
.
52.
Komrokji
RS
,
Al Ali
NH
,
Sallman
D
,
Padron
E
,
DeZern
AE
,
Barnard
J
, et al
.
Validation of international working group response criteria in higher-risk myelodysplastic syndromes: a report on behalf of the MDS clinical research consortium
.
Cancer Med
2021
;
10
:
447
53
.
53.
Fenaux
P
,
Mufti
GJ
,
Hellstrom-Lindberg
E
,
Santini
V
,
Finelli
C
,
Giagounidis
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
32
.
54.
Abel
GA
,
Klepin
HD
.
Frailty and the management of hematologic malignancies
.
Blood
2018
;
131
:
515
24
.
55.
Abel
GA
,
Efficace
F
,
Buckstein
RJ
,
Tinsley
S
,
Jurcic
JG
,
Martins
Y
, et al
.
Prospective international validation of the quality of life in myelodysplasia scale (QUALMS)
.
Haematologica
2016
;
101
:
781
8
.
56.
Lancet
JE
,
Uy
GL
,
Cortes
JE
,
Newell
LF
,
Lin
TL
,
Ritchie
EK
, et al
.
CPX-351 (cytarabine and daunorubicin) liposome for injection versus conventional cytarabine plus daunorubicin in older patients with newly diagnosed secondary acute myeloid leukemia
.
J Clin Oncol
2018
;
36
:
2684
92
.