Eprenetapopt (APR-246) and Azacitidine in TP53-Mutant Myelodysplastic Syndromes

David A Sallman, Amy E DeZern, Guillermo Garcia-Manero, David P Steensma, Gail J Roboz, Mikkael A Sekeres, Thomas Cluzeau, Kendra L Sweet, Amy McLemore, Kathy L McGraw, John Puskas, Ling Zhang, Jiqiang Yao, Qianxing Mo, Lisa Nardelli, Najla H Al Ali, Eric Padron, Greg Korbel, Eyal C Attar, Hagop M Kantarjian, Jeffrey E Lancet, Pierre Fenaux, Alan F List, Rami S Komrokji, David A Sallman, Amy E DeZern, Guillermo Garcia-Manero, David P Steensma, Gail J Roboz, Mikkael A Sekeres, Thomas Cluzeau, Kendra L Sweet, Amy McLemore, Kathy L McGraw, John Puskas, Ling Zhang, Jiqiang Yao, Qianxing Mo, Lisa Nardelli, Najla H Al Ali, Eric Padron, Greg Korbel, Eyal C Attar, Hagop M Kantarjian, Jeffrey E Lancet, Pierre Fenaux, Alan F List, Rami S Komrokji

Abstract

Purpose: Approximately 20% of patients with TP53-mutant myelodysplastic syndromes (MDS) achieve complete remission (CR) with hypomethylating agents. Eprenetapopt (APR-246) is a novel, first-in-class, small molecule that restores wild-type p53 functions in TP53-mutant cells.

Methods: This was a phase Ib/II study to determine the safety, recommended phase II dose, and efficacy of eprenetapopt administered in combination with azacitidine in patients with TP53-mutant MDS or acute myeloid leukemia (AML) with 20%-30% marrow blasts (ClinicalTrials.gov identifier: NCT03072043).

Results: Fifty-five patients (40 MDS, 11 AML, and four MDS/myeloproliferative neoplasms) with at least one TP53 mutation were treated. The overall response rate was 71% with 44% achieving CR. Of patients with MDS, 73% (n = 29) responded with 50% (n = 20) achieving CR and 58% (23/40) a cytogenetic response. The overall response rate and CR rate for patients with AML was 64% (n = 7) and 36% (n = 4), respectively. Patients with only TP53 mutations by next-generation sequencing had higher rates of CR (69% v 25%; P = .006). Responding patients had significant reductions in TP53 variant allele frequency and p53 expression by immunohistochemistry, with 21 (38%) achieving complete molecular remission (variant allele frequency < 5%). Median overall survival was 10.8 months with significant improvement in responding versus nonresponding patients by landmark analysis (14.6 v 7.5 months; P = .0005). Overall, 19/55 (35%) patients underwent allogeneic hematopoietic stem-cell transplant, with a median overall survival of 14.7 months. Adverse events were similar to those reported for azacitidine or eprenetapopt monotherapy, with the most common grade ≥ 3 adverse events being febrile neutropenia (33%), leukopenia (29%), and neutropenia (29%).

Conclusion: Combination treatment with eprenetapopt and azacitidine is well-tolerated yielding high rates of clinical response and molecular remissions in patients with TP53-mutant MDS and oligoblastic AML.

Conflict of interest statement

David A. SallmanConsulting or Advisory Role: Celyad, Agios, Abbvie, Aprea AB, Bristol-Myers Squibb, Gilead Sciences, Intellia Therapeutics, Kite Pharma, Magenta Therapeutics, Novartis, SyndaxSpeakers' Bureau: Agios, Incyte, Bristol-Myers SquibbResearch Funding: Celgene, Jazz PharmaceuticalsPatents, Royalties, Other Intellectual Property: Intellectual Property Patent for LB-100 in MDS Amy E. DeZernConsulting or Advisory Role: Celgene, Novartis, Gilead SciencesResearch Funding: Celgene, Astex PharmaceuticalsTravel, Accommodations, Expenses: Abbvie Guillermo Garcia-ManeroHonoraria: Celgene, Astex Pharmaceuticals, Acceleron Pharma, Helssin, AbbvieConsulting or Advisory Role: Celgene, Astex Pharmaceuticals, Acceleron Pharma, Jazz Pharmaceuticals, Bristol-Myers Squibb, Helsinn TherapeuticsResearch Funding: Celgene, Astex Pharmaceuticals, Amphivena, Helsinn Therapeutics, Novartis, Abbvie, Bristol-Myers Squibb, Onconova Therapeutics, H3 Biomedicine, Merck David P. SteensmaStock and Other Ownership Interests: Arrowhead Pharmaceuticals, Sage TherapeuticsHonoraria: Daiichi Sankyo, Summer Road, Stemline Therapeutics, Celgene, Astex PharmaceuticalsConsulting or Advisory Role: Pfizer, Janssen Oncology, Agios, Onconova Therapeutics, Geron, Astex PharmaceuticalsResearch Funding: Aprea AB, Celgene/BMS, H3 Biomedicine Gail J. RobozConsulting or Advisory Role: Amphivena, Janssen, Amgen, Astex Pharmaceuticals, Celgene, Genoptix, MedImmune, Novartis, Pfizer, Abbvie, Argenx, Array BioPharma, Bayer, Celltrion, Jazz Pharmaceuticals, Orsenix, Genentech/Roche, Sandoz, Actinium Pharmaceuticals, Astellas Pharma, Eisai, Bayer, Daiichi Sankyo, MEI Pharma, Otsuka, Takeda, Roche, Agios, Trovagene, GlaxoSmithKline, Bristol-Myers Squibb, Helsinn Therapeutics, MesoblastResearch Funding: Abbvie, Agios, Astex Pharmaceuticals, Celgene, CTI, Karyopharm Therapeutics, MedImmune, MEI Pharma, Moffitt, Novartis, Onconova Therapeutics, Pfizer, Sunesis Pharmaceuticals, Tensha Therapeutics, Cellectis, Janssen, AmphivenaTravel, Accommodations, Expenses: Amphivena, Astex Pharmaceuticals, Janssen, Pfizer, Array BioPharma, Novartis, Abbvie, Jazz Pharmaceuticals, Celgene, Celltrion, Roche/Genentech, Sandoz, Bayer, Clovis Oncology, Amgen, Sunesis Pharmaceuticals, Eisai, Agios Mikkael A. SekeresConsulting or Advisory Role: Celgene, Millennium, Syros PharmaceuticalsResearch Funding: Takeda, Pfizer, Bristol-Myers Squibb Thomas CluzeauConsulting or Advisory Role: Abbvie, Agios, Bristol-Myers Squibb, Jazz Pharmaceuticals, Novartis, Roche, Takeda, Syros PharmaceuticalsSpeakers' Bureau: Novartis, Amgen, Sanofi, Astellas PharmaTravel, Accommodations, Expenses: Bristol-Myers Squibb, Novartis, Pfizer, Sanofi Kendra L. SweetLeadership: ImmtechConsulting or Advisory Role: Astellas Pharma, Bristol-Myers Squibb, Novartis, Takeda, Stemline TherapeuticsResearch Funding: Incyte Kathy L. McGrawResearch Funding: Genentech, Celgene Eric PadronHonoraria: Stemline Therapeutics, Blueprint MedicinesSpeakers' Bureau: Novartis, Taiho PharmaceuticalResearch Funding: Incyte, Bristol-Myers Squibb, Kura Oncology Greg KorbelEmployment: Aprea Therapeutics, IncLeadership: Aprea Therapeutics, IncStock and Other Ownership Interests: Aprea Therapeutics, Inc Eyal C. AttarEmployment: Agios, Aprea ABLeadership: Aprea ABStock and Other Ownership Interests: Agios, Aprea ABConsulting or Advisory Role: TeladocTravel, Accommodations, Expenses: Aprea AB, Agios Hagop M. KantarjianHonoraria: Abbvie, Amgen, ARIAD, Bristol-Myers Squibb, Immunogen, Orsenix, Pfizer, Agios, Takeda, Actinium PharmaceuticalsResearch Funding: Pfizer, Amgen, Bristol-Myers Squibb, Novartis, ARIAD, Astex Pharmaceuticals, Abbvie, Agios, Cyclacel, Immunogen, Jazz Pharmaceuticals Jeffrey E. LancetStock and Other Ownership Interests: ArvinasConsulting or Advisory Role: Jazz Pharmaceuticals, Astellas Pharma, Abbvie, Agios, BerGenBio, Daiichi Sankyo, ElevateBioResearch Funding: Pfizer Pierre FenauxHonoraria: CelgeneResearch Funding: Celgene Alan F. ListHonoraria: Celgene, Aileron Therapeutics, Cellular Biomedicine GroupConsulting or Advisory Role: Celgene, Cellular Biomedicine Group, Aileron Therapeutics, Acceleron Pharma, International Personalized Cancer Center, Precision Biosciences, CTI BioPharma Corp, Prelude TherapeuticsResearch Funding: CelgeneTravel, Accommodations, Expenses: Celgene, Cellular Biomedicine GroupOther Relationship: Thousand Talents Award Rami S. KomrokjiStock and Other Ownership Interests: AbbvieConsulting or Advisory Role: Novartis, Incyte, Bristol-Myers Squibb, Jazz Pharmaceuticals, Abbvie, Geron, Acceleron PharmaSpeakers' Bureau: Jazz Pharmaceuticals, Bristol-Myers Squibb, AgiosTravel, Accommodations, Expenses: Incyte, Jazz Pharmaceuticals, Bristol-Myers Squibb, AgiosNo other potential conflicts of interest were reported.

Figures

FIG 1.
FIG 1.
(A) Heat map of individual patient baseline risk characteristics including molecular and genetic features and best response to treatment, and (B) matchstick plot of TP53 mutation types and locations by best response. AML, acute myeloid leukemia; CMML, chronic myelomonocytic leukemia; CR, complete remission; DBD, DNA-binding domain; HI, hematologic improvement; INT, intermediate; IPSS-R, revised International Prognostic Scoring System; mCR, marrow complete remission; MDS, myelodysplastic syndromes; MDS/MPN, myelodysplastic syndromes/myeloproliferative neoplasms; NE, not evaluable; NR, not reported; ORR, overall response rate; PD, progressive disease; PRD, proline-rich domain; REG, C-terminal regulatory domain; SD, stable disease; TAD, transactivation domain; TET, tetramerization domain.
FIG 2.
FIG 2.
(A) Treatment response and duration for all 45 response-evaluable patients, (B) TP53 variant allele frequency by best response, and (C) serial p53 IHC positivity by best response. AE, adverse event; AML, acute myeloid leukemia; CMML, chronic myelomonocytic leukemia; CR, complete remission; HI, hematologic improvement; IHC, immunohistochemistry; mCR, marrow complete remission; MDS, myelodysplastic syndromes; MDS/MPN, myelodysplastic syndromes/myeloproliferative neoplasms; NE, not evaluable; NR, not reported; SD, stable disease; VAF, variant allele frequency.
FIG 3.
FIG 3.
Kaplan-Meier curves for OS (A) in intention-to-treat population, (B) in patients with response versus no response by landmark analysis at 4 months, and (C) in patients with CR and non-CR response versus no response by landmark analysis at 4 months. CR, complete remission; OS, overall survival.

References

    1. Papaemmanuil E Gerstung M Malcovati L, et al. : Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 122:3616-3627, 2013
    1. Haferlach T Nagata Y Grossmann V, et al. : Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 28:241-247, 2014
    1. Walter MJ Shen D Shao J, et al. : Clonal diversity of recurrently mutated genes in myelodysplastic syndromes. Leukemia 27:1275-1282, 2013
    1. Bejar R Stevenson K Abdel-Wahab O, et al. : Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med 364:2496-2506, 2011
    1. Sasaki K Kanagal-Shamanna R Montalban-Bravo G, et al. : Impact of the variant allele frequency of ASXL1, DNMT3A, JAK2, TET2, TP53, and NPM1 on the outcomes of patients with newly diagnosed acute myeloid leukemia. Cancer 126:765-774, 2020
    1. Hunter AM, Sallman DA: Current status and new treatment approaches in TP53 mutated AML. Best Pract Res Clin Haematol 32:134-144, 2019
    1. Singhal D Wee LYA Kutyna MM, et al. : The mutational burden of therapy-related myeloid neoplasms is similar to primary myelodysplastic syndrome but has a distinctive distribution. Leukemia 33:2842-2853, 2019
    1. Sallman DA Komrokji R Vaupel C, et al. : Impact of TP53 mutation variant allele frequency on phenotype and outcomes in myelodysplastic syndromes. Leukemia 30:666-673, 2016
    1. Bernard E Nannya Y Hasserjian RP, et al. : Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes. Nat Med 26:1549-1556, 2020
    1. Haase D Stevenson KE Neuberg D, et al. : TP53 mutation status divides myelodysplastic syndromes with complex karyotypes into distinct prognostic subgroups. Leukemia 33:1747-1758, 2019
    1. Kulasekararaj AG Smith AE Mian SA, et al. : TP53 mutations in myelodysplastic syndrome are strongly correlated with aberrations of chromosome 5, and correlate with adverse prognosis. Br J Haematol 160:660-672, 2013
    1. Montalban-Bravo G Kanagal-Shamanna R Benton CB, et al. : Genomic context and TP53 allele frequency define clinical outcomes in TP53-mutated myelodysplastic syndromes. Blood Adv 4:482-495, 2020
    1. Sallman DA Al Ali N Yun S, et al. : Clonal suppression of TP53 mutant MDS and oligoblastic AML with hypomethylating agent therapy improves overall survival. Blood 132:1817, 2018
    1. Fenaux P Mufti GJ 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 10:223-232, 2009
    1. Bally C Adès L Renneville A, et al. : Prognostic value of TP53 gene mutations in myelodysplastic syndromes and acute myeloid leukemia treated with azacitidine. Leuk Res 38:751-755, 2014
    1. Falconi G Fabiani E Piciocchi A, et al. : Somatic mutations as markers of outcome after azacitidine and allogeneic stem cell transplantation in higher-risk myelodysplastic syndromes. Leukemia 33:785-790, 2019
    1. Craddock CF Houlton AE Quek LS, et al. : Outcome of azacitidine therapy in acute myeloid leukemia is not improved by concurrent vorinostat therapy but is predicted by a diagnostic molecular signature. Clin Cancer Res 23:6430, 2017
    1. Welch JS Petti AA Miller CA, et al. : TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N Engl J Med 375:2023-2036, 2016
    1. Short NJ Kantarjian HM Loghavi S, et al. : Treatment with a 5-day versus a 10-day schedule of decitabine in older patients with newly diagnosed acute myeloid leukaemia: A randomised phase 2 trial. Lancet Haematol 6:e29-e37, 2019
    1. Zeidan AM Wang R Wang X, et al. : Clinical outcomes of older patients with AML receiving hypomethylating agents: A large population-based study in the United States. Blood Adv 4:2192-2201, 2020
    1. Lambert JM Gorzov P Veprintsev DB, et al. : PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell 15:376-388, 2009
    1. Bykov VJ Zhang Q Zhang M, et al. : Targeting of mutant p53 and the cellular redox balance by APR-246 as a strategy for efficient cancer therapy. Front Oncol 6:21, 2016
    1. Peng X Zhang MQ Conserva F, et al. : APR-246/PRIMA-1MET inhibits thioredoxin reductase 1 and converts the enzyme to a dedicated NADPH oxidase. Cell Death Dis 4:e881, 2013
    1. Maslah N Salomao N Drevon L, et al. : Synergistic effects of PRIMA-1Met (APR-246) and 5-azacitidine in TP53-mutated myelodysplastic syndromes and acute myeloid leukemia. Haematologica 105:1539, 2020
    1. Lehmann S Bykov VJ Ali D, et al. : Targeting p53 in vivo: A first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J Clin Oncol 30:3633-3639, 2012
    1. Deneberg S Cherif H Lazarevic V, et al. : An open-label phase I dose-finding study of APR-246 in hematological malignancies. Blood Cancer J 6:e447, 2016
    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 108:419-425, 2006
    1. Ganster C Schabb R Shirneshan K, et al. : TP53 status and outcome in myelodysplastic syndromes with complex aberrant karyotypes. EHA Libr 294702:EP785, 2020
    1. Kantarjian H Issa J-PJ Rosenfeld CS, et al. : Decitabine improves patient outcomes in myelodysplastic syndromes. Cancer 106:1794-1803, 2006
    1. Bejar R Stevenson KE Caughey B, et al. : Somatic mutations predict poor outcome in patients with myelodysplastic syndrome after hematopoietic stem-cell transplantation. J Clin Oncol 32:2691-2698, 2014
    1. Yun S Geyer SM Komrokji RS, et al. : Prognostic significance of serial molecular annotation in myelodysplastic syndromes (MDS) and secondary acute myeloid leukemia (sAML). Leukemia 10.1038/s41375-020-0997-4[epub ahead of print on July 29, 2020]
    1. DiNardo CD Pratz K Pullarkat V, et al. : Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 133:7-17, 2019
    1. Wei AH Strickland SA Hou J-Z, et al. : Venetoclax combined with low-dose cytarabine for previously untreated patients with acute myeloid leukemia: Results from a phase Ib/II study. J Clin Oncol 37:1277-1284, 2019
    1. Swords RT Coutre S Maris MB, et al. : Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, combined with azacitidine in patients with AML. Blood 131:1415-1424, 2018
    1. Sallman DA Al Malki M Asch AS, et al. : Tolerability and efficacy of the first-in-class anti-CD47 antibody magrolimab combined with azacitidine in MDS and AML patients: Phase Ib results. J Clin Oncol 38:7507, 2020. (suppl 15)
    1. Sekeres MA Schoonen WM Kantarjian H, et al. : Characteristics of US patients with myelodysplastic syndromes: Results of six cross-sectional physician surveys. J Natl Cancer Inst 100:1542-1551, 2008
    1. Cluzeau T Sebert M Rahmé R, et al. : Eprenetapopt plus azacitidine in TP53-mutated myelodysplastic syndromes and acute myeloid leukemia: A phase II study by the Groupe Francophone des Myélodysplasies (GFM). J Clin Oncol 39:1575-1583, 2021

Source: PubMed

3
Sottoscrivi