Skip to main content
eClinicalMedicine logoLink to eClinicalMedicine
. 2025 Oct 10;89:103546. doi: 10.1016/j.eclinm.2025.103546

Omacetaxine and azacitidine for untreated patients with myelodysplastic syndromes and excess blasts: a phase I/II clinical trial

Daniel A Pollyea a,, Brett M Stevens a, Diana Abbott b, Maria Amaya a, Jonathan A Gutman a, Andrew Kent a, Christine McMahon a, Marc Schwartz a, Clayton Smith a, Jessie Dell-Martin a, Connor Sohalski a, Alexandra Ellis a, Mary Haag c, Jeffrey Schowinsky c, Zenggang Pan c, Sweta B Patel a, Monica Ransom a, Austin Gillen a, Craig T Jordan a, Eric M Pietras a
PMCID: PMC12547833  PMID: 41140451

Summary

Background

Patients with myelodysplastic syndromes (MDS) with excess blasts (MDS-EB) have poor long-term outcomes. Our preclinical studies showed MDS-EB stem cells are dependent upon protein synthesis. We designed a phase 1/2 clinical trial to examine the safety/efficacy of the protein synthesis inhibitor omacetaxine mepesuccinate (oma) with the hypomethylating agent (HMA) azacitidine (aza) for patients with untreated MDS-EB.

Methods

Enrollment occurred from September 2018 to March 2024 and the study was registered at clinicaltrials.gov (NCT03564873). The phase 1 primary endpoint was to determine the maximum tolerated dose (MTD) and the phase 2 primary endpoint was to determine the overall response rate. Aza 75 mg/m2 was administered daily and oma twice daily days 1–7. Oma was escalated in three cohorts: 0.75 mg/m2, 1.0 mg/m2 and 1.25 mg/m2, with a de-escalation cohort (0.5 mg/m2), to find the maximum tolerated dose (MTD). Responders who tolerated therapy could continue sequential cycles. Those who did not respond, progressed, had significant toxicity or proceeded to allogeneic stem cell transplantation (ASCT) discontinued.

Findings

The MTD of oma was 0.5 mg/m2; dose limiting toxicities included hypoxia, respiratory failure, gastrointestinal bleed and gout. Common adverse events included thrombocytopenia, anemia, neutropenia and febrile neutropenia. Overall response rate was 13/24 (54%) with four complete remissions (CR). Ten patients were bridged to ASCT. With median follow-up time of 3.5 years, median response duration and progression-free survival were 719 and 92 days, respectively. Median overall survival was 1.5 years.

Interpretation

The MTD of oma in MDS-EB has been established. Responses, including CRs, occurred rapidly. This therapeutic combination, conceived based on data that it targets the malignant stem cell population, could be further studied for patients with MDS-EB but high toxicity needs to be taken into account.

Funding

HYPERLINCI, Edward P. Evans Foundation, Leukemia and Lymphoma Society Career Development Program, VA Merit, V-Foundation.

Keywords: Myelodysplastic syndromes, Clinical trial, Phase 1/2, Omacetaxine, Azacitidine


Research in context.

Evidence before this study

This clinical trial is the first to combine the protein synthesis inhibitor omacetaxine mepesuccinate with azacitidine for previously untreated patients with myelodysplastic syndromes (MDS) with excess blasts (EB). We searched PubMed and ClinicalTrials.gov from July 1, 2014 to December 1, 2024 for prior evidence of such a combination in this population and did not find any that met these search criteria. The clinical trial was developed based on pre-clinical data suggesting this combination could be effective in this population. This paper, published September 12, 2018, showed that protein synthesis inhibition could target MDS stem cells and inspired this clinical strategy.

Added value of this study

With this study we follow up on a promising hypothesis related to a new therapeutic direction, the targeting of an MDS stem cell population through protein synthesis inhibition, with a clinical trial in patients with this disease. We have detailed the toxicity profile of a novel drug combination, established the recommended dose for future studies, and report preliminary efficacy of this combination.

Implications of all the available evidence

It is possible to use omacetaxine mepesuccinate with azacitidine to target a malignant stem cell population in MDS with excess blasts. Further studies with this regimen or other similar regimens can now be planned or designed to potentially understand this mechanism, and how it can positively impact patients more optimally while avoiding adverse events.

Introduction

Patients with myelodysplastic syndromes (MDS) with excess blasts (MDS-EB)1 have poor long term outcomes.2 Currently the only approved therapeutic option for these patients is a hypomethylating agent (HMA). The HMA azacitidine (aza) was superior to best supportive care for high-risk, newly diagnosed MDS-EB patients, but efficacy with this agent is limited: the overall response rate (ORR) was 29% with a complete remission (CR) rate of 17%.3 In addition, a median time of 64 days to achieve a response was required.4 More potent, effective and rationally designed therapeutic strategies for this disease are needed. Recently, we determined that the BCL-2 inhibitor venetoclax targets the leukemia stem cell (LSC) compartment of a related myeloid malignancy, acute myeloid leukemia (AML).5 Widespread use of this agent has resulted in frequent, deep and durable remissions for patients with this disease.6 Given the biological similarities between AML and MDS-EB,7 we investigated whether a stem cell population exists in MDS-EB; we were indeed able to identify these cells, and found they showed distinct activation of oxidative phosphorylation and protein synthesis machinery. Furthermore, targeting protein synthesis showed eradication of the MDS stem cell population in primary patient specimens.8 This observation led us to design this phase 1/2 study of the protein synthesis inhibitor omacetaxine mepesuccinate (oma) with aza for untreated patients with MDS-EB. Here we report the clinical outcomes of this novel therapeutic combination.

Methods

Patients were eligible if they had MDS-EB, no prior therapy with a hypomethylating agent, an Eastern Cooperative Oncology Group performance status9 of ≤2, adequate organ function, and no uncontrolled diabetes or systemic infection. All patients received antimicrobial prophylaxis.

Aza 75 mg/m2 intravenously was administered daily on days 1–7. Oma was administered subcutaneously (SC), twice daily, on days 1–7. A cycle was defined as a 28-day period. There were three planned dose escalation cohorts of oma; 0.75 mg/m2, 1.0 mg/m2 and 1.25 mg/m2, with a dose de-escalation cohort (0.5 mg/m2). Dose escalation to find the maximum tolerated dose (MTD) was performed in a 3 + 3 fashion in which the first three patients were assigned to cohort 1; in the absence of dose limiting toxicity (DLT), escalation to the next cohort was permitted. If one of the first three patients in a cohort had DLT, a total of six patients were enrolled in that cohort; the cohort in which >1 patient experienced DLT was the maximally administered dose. The MTD was the cohort in which ≤1/6 patients had a DLT event. DLT was defined as the occurrence, in the first cycle of treatment, of: grade ≥3 non-hematologic toxicity, excluding grade ≥3 infection, fever or electrolyte abnormalities that resolved to < grade 2 in <72 h; absolute neutrophil count <500/μL or platelet count <10 × 109/L for >42 days in cycle one for patients who achieved a CR or marrow CR; any treatment related death.

Adverse events (AEs) were assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0. Serious AEs (SAEs) were defined as AEs that were life threatening, or resulted in death, hospitalization or prolongation of hospitalization. Bone marrow biopsies occurred at baseline, cycle 1 day 8, cycle 1 day 28 and again at the conclusion of cycle 4, 6 and every 6 cycles thereafter. Response assessments, hematologic improvement and transfusion independence were protocol defined per the 2006 International Working Group (IWG)10; for this report we have also updated these definitions using the 2023 version.11 Patients who responded and tolerated therapy could continue to receive sequential cycles of treatment indefinitely. Those who did not respond, progressed, had significant treatment related toxicity or proceeded to allogeneic stem cell transplantation (ASCT) came off the study.

Statistics

Kaplan–Meier survival analysis was used to calculate median survival times and accompanying 95% confidence intervals for overall survival (OS), progression-free survival (PFS), and duration of response among responders. OS was defined as time between the baseline assessment date and death date, or censor date for patients still alive. PFS was defined as time between the baseline assessment and date of relapse, date of death, date of first assessment among non-responders, or date of censoring among responders. Duration of response for responders was defined as time between the date of the first response and relapse or date of death. Reverse Kaplan–Meier analysis was used to calculate median follow-up time. For the phase II study, sample size was determined based on an alternative hypothesis of an ORR of 40% compared with a null hypothesis of a 20% ORR,12 using a Simon two-stage minimax design for phase II studies,13 with a significance level of 0.05 and a power of 80%. The primary objective of Phase 1 was to determine the MTD of oma in combination with aza; the primary objective of phase 2 was to determine the ORR of the combination.

Ethics

This single-institution investigator-initiated protocol was approved by the Colorado Multiple Institutional Review Board (#17-2215) and is included as a Supplemental file; patients were treated in accordance with the Declaration of Helsinki and the study was registered at clinicaltrials.gov (NCT03564873). Informed consent was obtained from all study patients.

Role of funding sources

Teva provided oma and financial support for the clinical trial and had input into the study design. Teva and other funding sources had no additional roles in the study, including data collection, analysis, interpretation or writing of the manuscript.

Results

Between September 2018 and March 2024, 24 patients were enrolled (Fig. 1). Nine patients were required to complete the phase 1 study. Baseline factors are listed in Table 1. Ten (42%) were male. The median age was 70 (range 53–80) and the median baseline blast percentage was 10% (range 6%–17%). The median molecular international prognostic scoring system (IPSS-M)14 score was 1.51 (very high risk) and 4 (17%) had TP53 mutations. Per the revised IPSS,15 cytogenetics were classified as good (N = 5), intermediate (N = 7), poor (N = 2) and very poor (N = 10). Four had MDS with increased blasts (IB)1 and 15 had MDS-IB2.16

Fig. 1.

Fig. 1

CONSORT flow chart.

Table 1.

Baseline characteristics of all patients and their outcomes.

Patient Age Sex Cohort Baseline Blasts (%) WHO Classification Mutations (VAF%) Cyto Category per IPSS-R IPSS-M Best Response Per 2006 IWG Best Response Per 2023 IWG Bridged to Transplant with Study Therapy Relapse
001 74 M 1 12 MDS-IB2 STAG2
56%
U2AF1
44%
Intermediate 1.79 (Very High) Marrow CR No Response Y N
002 76 M 1 7 MDS-IB1 RUNX1
9%
U2AF110%
BCOR
23%
STAG2
22%
Good 1.66 (Very High) Marrow CR with HI CRL (CRuni) Y N
003 61 F 1 7 MDS-IB1 None Very Poor 1.22 (High) Marrow CR with HI CRL (CRuni) N Y
004 70 F 0 9.5 MDS-IB2 ASXL1
34%
SRSF2
42%
STAG2
30%
Intermediate 1.18 (High) Marrow CR with HI CRL (CRbi) Y N
005 57 M 0 13 MDS-IB2 DNMT3A
45%
SETBP1
48%
SRSF2
47%
TET2
45%
SCF3R
60%
Poor 2.52 (Very High) Marrow CR CRL (CRuni) Y N
006 70 F 0 10 MDS-IB2 RUNX1
8%
PHF6
7%
Very Poor 0.65 (High) No response No Response N N/A
007 73 M 0 13.5 MDS-IB2 SF3B1
20%
Intermediate 0.45 (Moderate Low) CR CR Y N
008 74 F 0 11 MDS-IB2 TP53
44%
Very Poor 1.04 (High) Marrow CR CRL (CRuni) N Y
009 73 M 0 8 MDS-IB1 DDX41
17%
DDX41
22%
DDX41
51% JAK2
15%
CBL
5%
Good 0.72 (High) Marrow CR CRL (CRuni) Y N
010 71 F Expansion 17 MDS-IB2 ASXL1
9%
CBL
12%
CBL
11%
SF3B1
35%
Poor 3.15 (Very High) No Response HI-N N N
011 76 M Expansion 15 MDS-IB2 TET2
35%
NF1
25%
NF1
19%
SRSF2
32%
ASXL1
28%
TET2
33%
Good 0.48 (Moderate High) Marrow CR CRL (CRuni) N Y
012 70 F Expansion 13 MDS-IB2 NRAS
10%
NRAS
10%
TP53
63%
Very Poor 3.76 (Very High) No Response No Response N N/A
013 75 F Expansion 7.5 MDS-IB1 TP53
55%
DNMT3A
31%
Very Poor 1.11 (High) No Response No Response N N/A
014 70 M Expansion 12 MDS-IB2 ASXL1
43%
RUNX1
48%
CSF3R
48%
Very Poor 1.34 (High) No Response No Response N N/A
015 59 M Expansion 10 MDS-IB2 U2AF1
26%
BCOR
19%
ETV6
11%
Intermediate 1.30 (High) Marrow CR No Response Y N
016 69 F Expansion 6 MDS-IB1 DNMT3A
42%
TP53
73%
U2AF1
43%
Very Poor 3.64 (Very High) No Response No Response N N/A
017 53 F Expansion 10 MDS-IB2 No Mutations Intermediate 1.06 (High) No Response No Response N N/A
018 80 M Expansion 15 MDS-IB2 NRAS
6%
DNMT3A
20%
U2AF1
30%
BCORL1
33%
RUNX1
19%
BCOR
31%
Good 3.30 (Very High) CR CR Y N
019 63 M Expansion 12 MDS-IB2 KIT
17%
NRAS
19%
Intermediate 1.51 (Very High) Marrow CR with HI CRL (CRuni) N Y
020 70 F Expansion 10 MDS-IB2 TP53
46%
Very Poor 1.71 (Very High) CR CR Y Y
021 70 F Expansion 10 MDS-IB2 TET2
47%
TET2
13%
RUNX1
20%
DNMT3A
44%
CEBPA
35%
BCOR
43%
CALR
50%
Good 2.02 (Very High) No Response No Response N n/a
022 63 M Expansion 9 MDS-IB1 SF3B1
40%
TP53
78%
Very Poor 2.66 (Very High) Marrow CR No Response Y Y
023 62 F Expansion 10 MDS-IB2 TP53
35%
TP53
36%
Very Poor 3.26 (Very High) No Response No Response N n/a
024 76 M Expansion 15 MDS-IB2 No Mutations Intermediate 1.51 (Very High) CR CR N N

EB = excess blasts; IPSS-R = revised international prognostic scoring system; CR = complete remission; HI = hematologic improvement; CRL = CR with limited count recovery; CRuni = unilineage; CRbi = bilineage; N/A = not applicable due to not having had a response; WHO = World Health Organization; MDS-IB1 = MDS with increased blasts 1; MDS-IB2 = MDS with increased blasts 2; VAF% = variant allele frequency %.

Two patients experienced DLT events (grade 3 hypoxia and grade 4 respiratory failure) within the first three enrolled in cohort 1, requiring de-escalation to cohort 0. In cohort 0, 1/6 patients had a DLT event (grade 4 gastrointestinal bleed and grade 3 gout flare, occurring in the same patient). Therefore, the MTD was established as 0.5 mg/m2 SC omacetaxine BID on days 1–7 along with azacitidine 75 mg/m2 IV daily on days 1–7. Fifteen additional patients were enrolled at the MTD; all 24 are presented in the safety and efficacy analysis and detailed in Table 1. Five-hundred-and-six AEs were reported, including 36 SAEs. The most common grade >2 AEs regardless of attribution were thrombocytopenia (N = 17), anemia (N = 11), neutropenia (N = 8) and febrile neutropenia (N = 6); the most common SAEs were febrile neutropenia (N = 6), infection/pneumonia (N = 5), fever (N = 3) and sepsis (N = 3). There were two grade 5 events, both due to sepsis and deemed unrelated; one patient died within 30 days of treatment of sepsis and one patient died within 60 days of treatment from disease progression. Grade >2 AEs are listed in Table 2.

Table 2.

Grade > 2 adverse events and serious adverse events, regardless of attribution, and the number of patients who experienced each event.

Event Grade 3 Grade 4 Grade 5
Hematologic adverse events
 Thrombocytopenia 10 7 0
 Febrile neutropenia 5 1 0
 Anemia 10 1 0
 Neutropenia 3 5 0
Non-hematologic adverse events
 Systemic inflammatory response syndrome/Sepsis 1 4 2
 Pneumonia 2 1 0
 COVID-19 infection 1 0 0
 Sinusitis 1 0 0
 Skin infection 2 0 0
 Urinary tract infection 1 0 0
 Stridor 1 0 0
 Hypoxia/respiratory failure/dyspnea 5 1 0
 Acute kidney injury 2 0 0
 Dysphagia 2 0 0
 Hypotension 4 0 0
 Hypertension 1 0 0
 Hypophosphatemia 3 0 0
 Hypocalcemia 1 0 0
 Hypokalemia 1 0 0
 Hyponatremia 2 0 0
 Hyperglycemia 1 0 0
 Arthralgia 1 0 0
 Cardiac arrythmia 2 0 0
 Edema 2 0 0
 Gout 1 0 0
 Encephalopathy 2 0 0
 Fatigue 2 0 0
 Chest pain 0 1 0
 Fever 2 0 0
 Intracranial hemorrhage/stroke 2 0 0
 Gastrointestinal bleed 1 1 0
 Muscle weakness 0 1 0
 Pleural effusion 0 1 0

Highest grade of each event per patient is recorded once.

The median follow up time for all patients was 3.5 years (95% confidence internal [CI] 1.8, 4.9). Per 2023 IWG criteria,11 the ORR was 13/24 (54%) with N = 4 CR, N = 8 CR with limited count recovery (CRL) and N = 1 Hematologic Improvement. Using 2006 IWG10 criteria, the ORR was 15/24 (63%) (Table 3). The median total number of cycles received was 1 (1-3). Responses occurred quickly, after a median of 1 cycle (1-3). Ten patients were bridged to ASCT due to responses that occurred as a result of oma/aza; of these, 2 have relapsed. A total of 6/15 (40%) responders experienced disease progression. Of the 9 non-responders per 2006 IWG criteria, 8 received second-line salvage therapy. Seven received venetoclax with azacitidine; one received intensive chemotherapy with fludarabine, cytarabine, growth factor and idarubicin (FLAG-Ida). Six proceeded to ASCT. Median response duration was 2 years (95% CI 73 days, Not Reached). Using 2023 IWG response criteria, median progression free survival was 92 days (95% CI: 41, 611) (Fig. 2, left). Fifteen patients have died, from disease progression (N = 9), transplant related mortality (N = 3), sepsis (N = 2) and pulmonary hypertension (N = 1). Median OS was 1.5 years (Fig. 2, right); 3 years for those who had an ASCT and 7.3 months for those who did not. Individual patient outcomes are summarized in Fig. 3.

Table 3.

Responses.

2006 IWG Criteria 2023 IWG Criteria
Complete remission 4 (17%) 4 (17%)
Complete remission with limited count recovery (CRL) N/A 8 (33%)
 CR Bilineage (CRbi) N/A 1 (4%)
 CRL Unilineage (CRuni) N/A 7 (29%)
Hematologic improvement-neutrophil response N/A 1 (4%)
Marrow CR 11 (46%) N/A
 Erythroid Response 3 N/A
 Platelet Response 2 N/A
 Neutrophil Response 0 N/A
Overall response 15 (63%) 13 (54%)
No response 9 (38%) 11 (46%)

Fig. 2.

Fig. 2

Outcomes of phase 1/2 clinical trial of omacetaxine and azacitidine for previously untreated myelodysplastic syndrome with excess blasts patients. A) Progression-free survival; B) overall survival.

Fig. 3.

Fig. 3

Swim lane plot showing individual patient response and outcomes. CR = complete remission; CRL = CR with limited count recovery; CRuni = unilineage; CRbi = bilineage; HI-N = hematologic improvement, neutrophils.

Discussion

Effective treatment of MDS-EB remains a major challenge.2,17, 18, 19, 20, 21 Standard-of-care regimens employing HMAs exhibit suboptimal response rates and require a median of 4–6 cycles to achieve,3,22,23 indicating that HMAs alone are unlikely to effectively target the malignant stem cells that serve as a reservoir for disease. However, recent efforts to add therapies to an HMA backbone in newly-diagnosed higher-risk MDS have not succeeded. A study that randomized patients to receive aza, aza with lenalidomide or aza with vorinostat did not result in improved response rates.24 The addition of the NEDD-8 activating enzyme inhibitor pevonedistat with aza did improve response rates but not event-free survival.25 The TP53 targeting therapy eprenetapopt,26 T-cell immunoglobulin domain and mucin domain −2 (TIM-3) inhibitor sabatolimab27 and CD47-targeting monoclonal antibody magrolimab28 also either failed definitive studies or are no longer being developed. Currently, a definitive trial of the BCL-2 inhibitor venetoclax with aza awaits publication (NCT04401748); the phase 1b results were recently published and were promising29 but a press release on June 16, 2025 stated the study did not meet its primary endpoint of OS. Notably, venetoclax has stem-cell targeting properties in AML,5,30,31 and therefore, we focused on the augmentation of aza backbone therapies with novel strategies to target MDS stem cells.

Therefore, to rationally investigate other potential aza partners we exhaustively characterized the biological properties of primary human MDS stem cell populations.8 We established that MDS stem cells exhibited high rates of protein translation and, using the translation inhibitor omacetaxine mepesuccinate in preclinical in vivo models of high-risk MDS, showed specific targeting of MDS stem cells that significantly augmented the activity of aza,8 hence providing preclinical rationale for our trial. In this pilot study, we were able to establish the MTD of this combination. Response rates of roughly 50% in this small series would favorably compare to historical studies establishing aza as the standard of care3,4 and more recent control arms that have employed aza alone.25,27 A relatively high rate of patients proceeded to ASCT but despite this, there are few long-term survivors. This may be a reflection of the small sample size and relatively high-risk population. However, it is notable that three patients died of transplant-related mortality. While no obvious associations with this outcome and the study exist, there may be as-yet unknown associations.

Those who proceeded to ASCT had a longer OS; these patients had a higher response rate (70%) than those who did not proceed to ASCT (43%). The median number of cycles administered is lower than other studies; patients moved quickly to ASCT in some cases, and in others, once they achieved remission, were maintained on aza alone.

This was a small, single-center study, and these results would need to be investigated in a larger population for more enthusiasm to be reasonable. In addition, the toxicity impact of adding a second agent to HMAs in this population can be considerable, and was a limitation in this trial. However, employing this type of stem-cell directed strategy to a group of MDS-EB patients who are being steered toward ASCT, allowing for ideally deeper pre-ASCT responses that can translate into better post-ASCT options, could be viable.

Contributors

DAP designed the clinical trial, recruited and treated patients, verified the underlying data and contributed to data analysis. DA led data analysis and verified the underlying data. MA, JAG, AK, CM, MS, CS, JD-M, CS and recruited and treated patients and contributed to data analysis. JD-M and CS verified the underlying data. MH, JS and ZP contributed data. BMS, SBP, MR, AG, CTJ and EMP performed correlative work and related data analysis. DAP, EMP, BMS and CTJ wrote and edited the manuscript. All authors read and approved the final version of the manuscript.

Data sharing statement

Data collected for this study, including de-identified individual patient data and a data dictionary defining each field in the set, can be made available on reasonable request and after signing appropriate data sharing agreements. Please send data access requests to the corresponding author. Such requests must be approved by the respective ethics boards and appropriate data custodians.

Declaration of interests

Teva supplied omacetaxine and financial support for the clinical trial in the form of payments made to the University of Colorado. CS receives salary and equity for OncoVerity and consulting fees from RefinedScience. CM served on an advisory board for Syndax and Kura and serves on a safety monitoring committee for Kura. DA has served on a data safety monitoring board for Merck. DAP received consulting fees from Abbvie, Gilead and Syros.

Acknowledgements

B.M.S. was supported by R01 CA286717 and an Edward P. Evans Foundation Young Investigator Award. C.T.J. was supported by the Nancy Carroll Allen Chair in Hematology Research and R01 CA200707. E.M.P. was supported by the Cleo Meador and George Ryland Scott Chair in Hematology Research, a Leukemia and Lymphoma Society Scholar Award, R01 CA286717, and an Edward P. Evans Foundation Discovery Research Grant. DAP was supported by the Robert H. Allen MD Chair in Hematology Research, the Leukemia and Lymphoma Society Scholar in Clinical Research Achievement Award, the V-Foundation and the University of Colorado Department of Medicine Outstanding Early Career Scholar Program. This work was also supported by the University of Colorado Cancer Center P30.

Footnotes

Appendix A

Supplementary data related to this article can be found at https://doi.org/10.1016/j.eclinm.2025.103546.

Appendix A. Supplementary data

2024.08.29_Oma_Aza_Protocol
mmc1.pdf (680.5KB, pdf)

References

  • 1.Arber D.A., Orazi A., Hasserjian R., et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–2405. doi: 10.1182/blood-2016-03-643544. [DOI] [PubMed] [Google Scholar]
  • 2.Sekeres M.A., Taylor J. Diagnosis and treatment of myelodysplastic syndromes: a review. JAMA. 2022;328(9):872–880. doi: 10.1001/jama.2022.14578. [DOI] [PubMed] [Google Scholar]
  • 3.Fenaux P., Mufti G.J., Hellstrom-Lindberg E., 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(3):223–232. doi: 10.1016/S1470-2045(09)70003-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Silverman L.R., Demakos E.P., Peterson B.L., et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol. 2002;20(10):2429–2440. doi: 10.1200/JCO.2002.04.117. [DOI] [PubMed] [Google Scholar]
  • 5.Pollyea D.A., Stevens B.M., Jones C.L., et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat Med. 2018;24(12):1859–1866. doi: 10.1038/s41591-018-0233-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.DiNardo C.D., Jonas B.A., Pullarkat V., et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383(7):617–629. doi: 10.1056/NEJMoa2012971. [DOI] [PubMed] [Google Scholar]
  • 7.Arber D.A., Orazi A., Hasserjian R.P., et al. International consensus classification of myeloid neoplasms and acute leukemias: integrating morphologic, clinical, and genomic data. Blood. 2022;140(11):1200–1228. doi: 10.1182/blood.2022015850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Stevens B.M., Khan N., D'Alessandro A., et al. Characterization and targeting of malignant stem cells in patients with advanced myelodysplastic syndromes. Nat Commun. 2018;9(1):3694. doi: 10.1038/s41467-018-05984-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Oken M.M., Creech R.H., Tormey D.C., et al. Toxicity and response criteria of the eastern cooperative oncology Group. Am J Clin Oncol. 1982;5(6):649–655. [PubMed] [Google Scholar]
  • 10.Cheson B.D., Greenberg P.L., Bennett J.M., et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood. 2006;108(2):419–425. doi: 10.1182/blood-2005-10-4149. [DOI] [PubMed] [Google Scholar]
  • 11.Zeidan A.M., Platzbecker U., Bewersdorf J.P., et al. Consensus proposal for revised International Working Group 2023 response criteria for higher-risk myelodysplastic syndromes. Blood. 2023;141(17):2047–2061. doi: 10.1182/blood.2022018604. [DOI] [PubMed] [Google Scholar]
  • 12.Zeidan A.M., Sekeres M.A., Garcia-Manero G., et al. Comparison of risk stratification tools in predicting outcomes of patients with higher-risk myelodysplastic syndromes treated with azanucleosides. Leukemia. 2016;30(3):649–657. doi: 10.1038/leu.2015.283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Simon R. Optimal two-stage designs for phase II clinical trials. Control Clin Trials. 1989;10(1):1–10. doi: 10.1016/0197-2456(89)90015-9. [DOI] [PubMed] [Google Scholar]
  • 14.Bernard E., Tuechler H., Greenberg P.L., et al. Molecular international prognostic scoring system for myelodysplastic syndromes. NEJM Evid. 2022;1(7) doi: 10.1056/EVIDoa2200008. [DOI] [PubMed] [Google Scholar]
  • 15.Greenberg P.L., Tuechler H., Schanz J., et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012;120(12):2454–2465. doi: 10.1182/blood-2012-03-420489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Khoury J.D., Solary E., Abla O., et al. The 5th edition of the world Health Organization classification of haematolymphoid tumours: myeloid and Histiocytic/Dendritic neoplasms. Leukemia. 2022;36(7):1703–1719. doi: 10.1038/s41375-022-01613-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Platzbecker U., Kubasch A.S., Homer-Bouthiette C., Prebet T. Current challenges and unmet medical needs in myelodysplastic syndromes. Leukemia. 2021;35(8):2182–2198. doi: 10.1038/s41375-021-01265-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zhan D., Park C.Y. Stem cells in the myelodysplastic syndromes. Front Aging. 2021;2 doi: 10.3389/fragi.2021.719010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Woll P.S., Yoshizato T., Hellstrom-Lindberg E., Fioretos T., Ebert B.L., Jacobsen S.E.W. Targeting stem cells in myelodysplastic syndromes and acute myeloid leukemia. J Intern Med. 2022;292(2):262–277. doi: 10.1111/joim.13535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Rodriguez-Sevilla J.J., Colla S. A road map of relapse in MDS after allo-HSCT. Blood. 2024;143(11):941–943. doi: 10.1182/blood.2023023533. [DOI] [PubMed] [Google Scholar]
  • 21.Bewersdorf J.P., Xie Z., Bejar R., et al. Current landscape of translational and clinical research in myelodysplastic syndromes/neoplasms (MDS): proceedings from the 1(st) International Workshop on MDS (iwMDS) of the International Consortium for MDS (icMDS) Blood Rev. 2023;60 doi: 10.1016/j.blre.2023.101072. [DOI] [PubMed] [Google Scholar]
  • 22.Rajakumaraswamy N., Gandhi M., Wei A.H., et al. Real-world effectiveness of azacitidine in treatment-naive patients with higher-risk myelodysplastic syndromes. Clin Lymphoma Myeloma Leuk. 2024;24(4):260–268.e2. doi: 10.1016/j.clml.2023.12.008. [DOI] [PubMed] [Google Scholar]
  • 23.Khan C., Pathe N., Fazal S., Lister J., Rossetti J.M. Azacitidine in the management of patients with myelodysplastic syndromes. Ther Adv Hematol. 2012;3(6):355–373. doi: 10.1177/2040620712464882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sekeres M.A., Othus M., List A.F., 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(24):2745–2753. doi: 10.1200/JCO.2015.66.2510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ades L., Girshova L., Doronin V.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(17):5132–5145. doi: 10.1182/bloodadvances.2022007334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sallman D.A., DeZern A.E., Garcia-Manero G., et al. Eprenetapopt (APR-246) and azacitidine in TP53-Mutant myelodysplastic syndromes. J Clin Oncol. 2021;39(14):1584–1594. doi: 10.1200/JCO.20.02341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zeidan A.M., Ando K., Rauzy O., 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(1):e38–e50. doi: 10.1016/S2352-3026(23)00333-2. [DOI] [PubMed] [Google Scholar]
  • 28.Sallman D.A., Al Malki M.M., Asch A.S., 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(15):2815–2826. doi: 10.1200/JCO.22.01794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Garcia J.S., Platzbecker U., Odenike O., et al. Efficacy and safety of venetoclax plus azacitidine for patients with treatment-naive high-risk myelodysplastic syndromes. Blood. 2025;145(11):1126–1135. doi: 10.1182/blood.2024025464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Jones C.L., Stevens B.M., D'Alessandro A., et al. Cysteine depletion targets leukemia stem cells through inhibition of electron transport complex II. Blood. 2019;134(4):389–394. doi: 10.1182/blood.2019898114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Jones C.L., Stevens B.M., D'Alessandro A., et al. Inhibition of amino acid metabolism selectively targets human leukemia stem cells. Cancer Cell. 2018;34(5):724–740.e4. doi: 10.1016/j.ccell.2018.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

2024.08.29_Oma_Aza_Protocol
mmc1.pdf (680.5KB, pdf)

Articles from eClinicalMedicine are provided here courtesy of Elsevier

RESOURCES