Pevonedistat plus azacitidine vs azacitidine alone in higher-risk MDS/chronic myelomonocytic leukemia or low-blast-percentage AML

Lionel Adès, Larisa Girshova, Vadim A Doronin, María Díez-Campelo, David Valcárcel, Suman Kambhampati, Nora-Athina Viniou, Dariusz Woszczyk, Raquel De Paz Arias, Argiris Symeonidis, Achilles Anagnostopoulos, Eduardo Ciliao Munhoz, Uwe Platzbecker, Valeria Santini, Robert J Fram, Ying Yuan, Sharon Friedlander, Douglas V Faller, Mikkael A Sekeres, Lionel Adès, Larisa Girshova, Vadim A Doronin, María Díez-Campelo, David Valcárcel, Suman Kambhampati, Nora-Athina Viniou, Dariusz Woszczyk, Raquel De Paz Arias, Argiris Symeonidis, Achilles Anagnostopoulos, Eduardo Ciliao Munhoz, Uwe Platzbecker, Valeria Santini, Robert J Fram, Ying Yuan, Sharon Friedlander, Douglas V Faller, Mikkael A Sekeres

Abstract

PANTHER is a global, randomized phase 3 trial of pevonedistat+azacitidine (n = 227) vs azacitidine monotherapy (n = 227) in patients with newly diagnosed higher-risk myelodysplastic syndromes (MDS; n = 324), higher-risk chronic myelomonocytic leukemia (n = 27), or acute myeloid leukemia (AML) with 20% to 30% blasts (n = 103). The primary end point was event-free survival (EFS). In the intent-to-treat population, the median EFS was 17.7 months with pevonedistat+azacitidine vs 15.7 months with azacitidine (hazard ratio [HR], 0.968; 95% confidence interval [CI], 0.757-1.238; P = .557) and in the higher-risk MDS cohort, median EFS was 19.2 vs 15.6 months (HR, 0.887; 95% CI, 0.659-1.193; P = .431). Median overall survival (OS) in the higher-risk MDS cohort was 21.6 vs 17.5 months (HR, 0.785; P = .092), and in patients with AML with 20% to 30% blasts was 14.5 vs 14.7 months (HR, 1.107; P = .664). In a post hoc analysis, median OS in the higher-risk MDS cohort for patients receiving >3 cycles was 23.8 vs 20.6 months (P = .021) and for >6 cycles was 27.1 vs 22.5 months (P = .008). No new safety signals were identified, and the azacitidine dose intensity was maintained. Common hematologic grade ≥3 treatment emergent adverse events were anemia (33% vs 34%), neutropenia (31% vs 33%), and thrombocytopenia (30% vs 30%). These results underscore the importance of large, randomized controlled trials in these heterogeneous myeloid diseases and the value of continuing therapy for >3 cycles. The trial was registered on clinicaltrials.gov as #NCT03268954.

© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Distribution of poor prognostic and frequently mutated genes in patients with higher-risk MDS by treatment arm. Mutational analysis was conducted on 270 bone marrow aspirate samples collected at screening (n = 135 samples from each treatment arm).
Figure 2.
Figure 2.
EFS and OS in the ITT population. Kaplan-Meier curve of EFS (A) and Kaplan-Meier curve of OS (B). ITT, intent-to-treat.
Figure 3.
Figure 3.
EFS in the higher-risk MDS cohort. Kaplan-Meier curve of EFS (A) and forest plot of subgroup analysis of EFS (B). *Lower confidence interval is truncated at 0.125. NE, not estimable.
Figure 4.
Figure 4.
OS in the higher-risk MDS cohort.
Figure 5.
Figure 5.
OS in patients with AML with 20% to 30% blasts. Kaplan-Meier curve of OS (A) and forest plot of subgroup analysis of OS (B). *Upper confidence interval is truncated at 8 and lower confidence interval is truncated at 0.125. ELN, European LeukemiaNet; NE, not estimable; WBC, white blood cell.
Figure 6.
Figure 6.
EFS and OS in patients with higher-risk CMML. Kaplan-Meier curve of EFS (A) and Kaplan-Meier curve of OS (B). NE, not estimable.
Figure 7.
Figure 7.
OS in patients with higher-risk MDS by number of cycles received. Patients who received greater than 3 cycles (A) and patients who received greater than 6 cycles (B).

References

    1. Granfeldt Østgård LS, Medeiros BC. Epidemiology and impact of preceding or underlying disease in secondary acute myeloid leukemia. HemaSphere. 2018;2(S2):153-155.
    1. Ogawa S. Genetics of MDS. Blood. 2019;133(10):1049-1059.
    1. Fisher DAC, Fowles JS, Zhou A, Oh ST. Inflammatory pathophysiology as a contributor to myeloproliferative neoplasms. Front Immunol. 2021;12:683401.
    1. Garcia-Manero G, Chien KS, Montalban-Bravo G. Myelodysplastic syndromes: 2021 update on diagnosis, risk stratification and management. Am J Hematol. 2020;95(11):1399-1420.
    1. Kwon J. Diagnosis and treatment of chronic myelomonocytic leukemia. Blood Res. 2021;56(S1):S5-S16.
    1. Greenberg PL, Stone RM, Al-Kali A, et al. . Myelodysplastic syndromes, version 2.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2017;15(1):60-87.
    1. Hasserjian RP, Buckstein R, Patnaik MM. Navigating myelodysplastic and myelodysplastic/myeloproliferative overlap syndromes. Am Soc Clin Oncol Educ Book. 2021;41(41):328-350.
    1. Roman E, Smith A, Appleton S, et al. . Myeloid malignancies in the real-world: occurrence, progression and survival in the UK’s population-based Haematological Malignancy Research Network 2004-15. Cancer Epidemiol. 2016;42:186-198.
    1. Howlader N, Noone AM, Krapcho M, et al, eds. SEER Cancer Statistics Review, 1975-2017. Based on November 2019 SEER data submission, posted to the SEER website. Bethesda, MD: National Cancer Institute; April 2020.
    1. Catenacci DV, Schiller GJ. Myelodysplasic syndromes: a comprehensive review. Blood Rev. 2005;19(6):301-319.
    1. Griffiths EA, Carraway HE, Chandhok NS, Prebet T. Advances in non-intensive chemotherapy treatment options for adults diagnosed with acute myeloid leukemia. Leuk Res. 2020;91:106339.
    1. Bewersdorf JP, Zeidan AM. Risk-adapted, individualized treatment strategies of myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML). Cancers (Basel). 2021;13(7):1610.
    1. Kayser S, Levis MJ. Updates on targeted therapies for acute myeloid leukaemia. Br J Haematol. 2022;196(2):316-328.
    1. Garcia JS, Swords RT, Roboz GJ, et al. . A systematic review of higher-risk myelodysplastic syndromes clinical trials to determine the benchmark of azacitidine and explore alternative endpoints for overall survival [published correction appears in Leuk Res. 2021;105;10655]. Leuk Res. 2021;104:106555.
    1. Bell JA, Galaznik A, Blazer M, et al. . Transfusion-free interval is associated with improved survival in patients with higher-risk myelodysplastic syndromes engaged in routine care. Leuk Lymphoma. 2019;60(1):49-59.
    1. Zeidan AM, Davidoff AJ, Long JB, et al. . Comparative clinical effectiveness of azacitidine versus decitabine in older patients with myelodysplastic syndromes. Br J Haematol. 2016;175(5):829-840.
    1. Bernal T, Martínez-Camblor P, Sánchez-García J, et al. ; Spanish Group on Myelodysplastic Syndromes; PETHEMA Foundation; Spanish Society of Hematology . Effectiveness of azacitidine in unselected high-risk myelodysplastic syndromes: results from the Spanish registry. Leukemia. 2015; 29(9):1875-1881.
    1. Soucy TA, Smith PG, Milhollen MA, et al. . An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. Nature. 2009;458(7239):732-736.
    1. Brownell JE, Sintchak MD, Gavin JM, et al. . Substrate-assisted inhibition of ubiquitin-like protein-activating enzymes: the NEDD8 E1 inhibitor MLN4924 forms a NEDD8-AMP mimetic in situ. Mol Cell. 2010;37(1):102-111.
    1. Soucy TA, Dick LR, Smith PG, Milhollen MA, Brownell JE. The NEDD8 conjugation pathway and its relevance in cancer biology and therapy. Genes Cancer. 2010;1(7):708-716.
    1. Swords RT, Kelly KR, Smith PG, et al. . Inhibition of NEDD8-activating enzyme: a novel approach for the treatment of acute myeloid leukemia. Blood. 2010;115(18):3796-3800.
    1. Zhou L, Zhang W, Sun Y, Jia L. Protein neddylation and its alterations in human cancers for targeted therapy. Cell Signal. 2018;44:92-102.
    1. Sekeres MA, Watts J, Radinoff A, et al. . Randomized phase 2 trial of pevonedistat plus azacitidine versus azacitidine for higher-risk MDS/CMML or low-blast AML [published correction appears in Leukemia. 2021;35(12):3637]. Leukemia. 2021;35(7):2119-2124.
    1. Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood. 2002;100(7):2292-2302.
    1. Greenberg PL, Tuechler H, Schanz J, et al. . Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012;120(12):2454-2465.
    1. Cheson BD, Greenberg PL, Bennett JM, et al. . Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood. 2006;108(2):419-425.
    1. Cheson BD, Bennett JM, Kopecky KJ, et al. ; International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia . 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(24):4642-4649.
    1. Sekeres MA, Othus M, List AF, 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.
    1. Garcia-Manero G, Sekeres MA, Egyed M, et al. . A phase 1b/2b multicenter study of oral panobinostat plus azacitidine in adults with MDS, CMML or AML with ⩽30% blasts. Leukemia. 2017;31(12):2799-2806.
    1. Celgene Corporation. Highlights of prescribing information: VIDAZA (azacitidine for injection), for subcutaneous or intravenous use. 2004. Available at: . Accessed 15 December 2021.
    1. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. ; International Vidaza High-Risk MDS Survival Study Group . 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.
    1. Garcia-Manero G, Montalban-Bravo G, Berdeja JG, 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(6):994-1002.
    1. Sallman D, Al Malki M, Asch A, et al. The first-in-class anti-CD47 antibody magrolimab combined with azacitidine is well-tolerated and effective in MDS patients: phase 1b results [oral presentation]. European Hematology Association (EHA) 2020. Presentation S187.
    1. Wei A, Esteve J, Porkka K, et al. Sabatolimab plus hypomethylating agents (HMAs) in patients (pts) with high-/very high-risk myelodysplastic syndrome (HR/VHR-MDS) and acute myeloid leukemia (AML): subgroup analysis of a phase 1 study [oral presentation]. European Hematology Association (EHA) 2021: Presentation S168.
    1. Garcia JS, Wei AH, Borate U, et al. . Safety, efficacy, and patient-reported outcomes of venetoclax in combination with azacitidine for the treatment of patients with higher-risk myelodysplastic syndrome: a phase 1b study. [abstract] Blood. 2020;136(suppl 1):55-57. Abstract 656.
    1. Sallman DA, DeZern AE, Garcia-Manero G, et al. . Eprenetapopt (APR-246) and azacitidine in TP53-mutant myelodysplastic syndromes. J Clin Oncol. 2021;39(14):1584-1594.
    1. Aprea Therapeutics. Aprea Therapeutics announces results of primary endpoint from phase 3 trial of eprenetapopt in TP53 mutant myelodysplastic syndromes (MDS). 2020. Available at: . Accessed 15 December 2021.

Source: PubMed

3
Suscribir