A study to assess the efficacy of enasidenib and risk-adapted addition of azacitidine in newly diagnosed IDH2-mutant AML

Sheng F Cai, Ying Huang, Jennie R Lance, Hsiaoyin Charlene Mao, Andrew J Dunbar, Samantha N McNulty, Todd Druley, Yan Li, Maria R Baer, Wendy Stock, Tibor Kovacsovics, William G Blum, Gary J Schiller, Rebecca L Olin, James M Foran, Mark Litzow, Tara Lin, Prapti Patel, Matthew C Foster, Michael Boyiadzis, Robert H Collins, Jordan Chervin, Abigail Shoben, Jo-Anne Vergilio, Nyla A Heerema, Leonard Rosenberg, Timothy L Chen, Ashley O Yocum, Franchesca Druggan, Sonja Marcus, Mona Stefanos, Brian J Druker, Alice S Mims, Uma Borate, Amy Burd, John C Byrd, Ross L Levine, Eytan M Stein, Sheng F Cai, Ying Huang, Jennie R Lance, Hsiaoyin Charlene Mao, Andrew J Dunbar, Samantha N McNulty, Todd Druley, Yan Li, Maria R Baer, Wendy Stock, Tibor Kovacsovics, William G Blum, Gary J Schiller, Rebecca L Olin, James M Foran, Mark Litzow, Tara Lin, Prapti Patel, Matthew C Foster, Michael Boyiadzis, Robert H Collins, Jordan Chervin, Abigail Shoben, Jo-Anne Vergilio, Nyla A Heerema, Leonard Rosenberg, Timothy L Chen, Ashley O Yocum, Franchesca Druggan, Sonja Marcus, Mona Stefanos, Brian J Druker, Alice S Mims, Uma Borate, Amy Burd, John C Byrd, Ross L Levine, Eytan M Stein

Abstract

Enasidenib (ENA) is an inhibitor of isocitrate dehydrogenase 2 (IDH2) approved for the treatment of patients with IDH2-mutant relapsed/refractory acute myeloid leukemia (AML). In this phase 2/1b Beat AML substudy, we applied a risk-adapted approach to assess the efficacy of ENA monotherapy for patients aged ≥60 years with newly diagnosed IDH2-mutant AML in whom genomic profiling demonstrated that mutant IDH2 was in the dominant leukemic clone. Patients for whom ENA monotherapy did not induce a complete remission (CR) or CR with incomplete blood count recovery (CRi) enrolled in a phase 1b cohort with the addition of azacitidine. The phase 2 portion assessing the overall response to ENA alone demonstrated efficacy, with a composite complete response (cCR) rate (CR/CRi) of 46% in 60 evaluable patients. Seventeen patients subsequently transitioned to phase 1b combination therapy, with a cCR rate of 41% and 1 dose-limiting toxicity. Correlative studies highlight mechanisms of clonal elimination with differentiation therapy as well as therapeutic resistance. This study demonstrates both efficacy of ENA monotherapy in the upfront setting and feasibility and applicability of a risk-adapted approach to the upfront treatment of IDH2-mutant AML. This trial is registered at www.clinicaltrials.gov as #NCT03013998.

Conflict of interest statement

Conflict-of-interest disclosure: S.F.C. is a consultant for and holds equity interest in Imago BioSciences. G.J.S. has commercial interests in Bristol Myers Squibb (BMS), Amgen, and Johnson & Johnson (J&J); has received fees from AbbVie, Agios, Amgen, Astellas, BMS, Incyte, Janssen, Jazz, Karyopharm, Kite, Pharmacyclics, Sanofi/Genzyme, and Stemline; and has received research funding from AbbVie, Actinium, Actuate, Arog, Astellas, AltruBio, AVM Bio, BMS/Celgene, Celator, Constellation, Daiichi-Sankyo, Deciphera, Delta-Fly, Forma, FujiFilm, Gamida, Genentech-Roche, GlycoMimetics, Geron, Incyte, Karyopharm, Kiadis, Kite/Gilead, Kura, Marker, Mateon, Onconova, Pfizer, PrECOG, Regimmune, Samus, Sangamo, Sellas, Stemline, Syros, Takeda, Tolero, Trovagene, Agios, Amgen, Jazz, Orca, Ono-UK, and Novartis. J.M.F. has received research funding from Takeda/Millennium. P.P. has served on the advisory board of Agios Pharmaceuticals and is currently an employee of Servier. J.-A.V. is an employee of Foundation Medicine Inc with equity in Roche. B.J.D. reports serving on the scientific advisory board of Adela Bio, Aileron Therapeutics, and Therapy Architects/ALLCRON (inactive) as well as Cepheid, Celgene, DNA SEQ, Nemucore Medical Innovations, Novartis, RUNX1 Research Program, and Vivid Biosciences (inactive); reports serving on scientific advisory board and owning stocks in Aptose Biosciences, Blueprint Medicines, Enliven Therapeutics, Iterion Therapeutics, Grail, and Recludix Pharma; serving on the board of directors and owning stocks in Amgen and Vincerx Pharma; serving on the board of directors in Burroughs Wellcome Fund and CureOne; being a member on the joint steering committee of Beat AML LLS; being a member on the advisory committee of Multicancer Early Detection Consortium; is the founder of VB Therapeutics; reports sponsored research agreements with Enliven Therapeutics and Recludix Pharma; clinical trial funding from Novartis and AstraZeneca; royalties from patent 6958335 (Novartis exclusive license) and Oregon Health and Science University (OHSU) and Dana-Farber Cancer Institute (1 Merck exclusive license, 1 CytoImage, Inc exclusive license, and 1 Sun Pharma Advanced Research Company nonexclusive license); and holds US patents 4326534, 6958335, 7416873, 7592142, 10473667, 10664967, and 11049247. A.S.M. has served on the advisory boards of Jazz Pharmaceuticals, AbbVie/Genentech, Astellas Pharma, PTC Therapeutics, Novartis, Agios Pharmaceuticals and Syndax Pharmaceuticals. U.B. has been a consultant for Genentech, Daiichi Sankyo, Takeda, Pfizer, AbbVie/Genentech, and Novartis. J.C.B. is a current equity holder in Vincerx Pharma Inc (a publicly traded company); holds membership on the board of directors or advisory committees of Vincerx, Newave, and Orange Grove Bio; and reports being a consultant or receiving honoraria from Novartis, Trillium, Astellas, AstraZeneca, Pharmacyclics, and Syndax. R.L.L. is on the supervisory board of Qiagen; is a scientific adviser to Imago, Mission Bio, Zentalis, Ajax, Auron, Prelude, C4 Therapeutics, and Isoplexis; receives research support from and has consulted for Celgene and Roche; has consulted for Incyte, Janssen, Astellas, MorphoSys, and Novartis; and has received honoraria from AstraZeneca, Roche, Lilly, and Amgen for invited lectures and from Gilead for grant reviews. E.M.S. has served on the advisory boards of Astellas Pharma, AbbVie, Genentech, Daiichi Sankyo, Novartis, Amgen, Seattle Genetics, Syros Pharmaceuticals, Syndax Pharmaceuticals, Agios Pharmaceuticals, and Celgene, and is an equity holder in Auron Therapeutics. The remaining authors declare no competing financial interests.

© 2024 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.
Patients with suspected AML harboring IDH2 R140/R172 mutations were screened under the Beat AML Master Trial and consented for this study (n = 64). Eligible patients started on ENA monotherapy with up to 5 cycles to achieve a CR or CRi (n = 60). Patients who achieved a CR/CRi by 5 cycles continued on ENA monotherapy until relapse (n = 29). Patients who did not achieve a CR/CRi started ENA plus AZA combination therapy (n = 17). Patients who experienced treatment failure, death, withdrew, or an adverse event warranting discontinuation during the first 5 cycles of ENA monotherapy went off study (n = 14). Patients were accrued in 2 stages, n = 24 and n = 36, respectively.
Figure 2.
Figure 2.
Durability of responses for patients who achieved a remission. Duration of response estimated by the Kaplan-Meier method for phase 1b (A) and phase 2 and Exp (B).
Figure 3.
Figure 3.
Survival assessments for patients enrolled on study. Overall survival estimated by the Kaplan-Meier method for phase 1b (A) and phase 2 and Exp (B).
Figure 4.
Figure 4.
Correlates of treatment response and resistance. (A) Serial bone marrow aspirate samples analyzed using multiparameter flow cytometry visualized using t-distributed Stochastic Neighbor Embedding (t-SNE) plots generated by Cytobank. (B) Correlating 2-HG data from serial plasma samples. (C) Serial bone marrow aspirate samples taken at baseline, CR and relapse were analyzed using the ArcherDX’s FusionPlex Myeloid and VariantPlex Myeloid panels.
Figure 4.
Figure 4.
Correlates of treatment response and resistance. (A) Serial bone marrow aspirate samples analyzed using multiparameter flow cytometry visualized using t-distributed Stochastic Neighbor Embedding (t-SNE) plots generated by Cytobank. (B) Correlating 2-HG data from serial plasma samples. (C) Serial bone marrow aspirate samples taken at baseline, CR and relapse were analyzed using the ArcherDX’s FusionPlex Myeloid and VariantPlex Myeloid panels.
Figure 5.
Figure 5.
Molecular correlates of response. Baseline mutations identified using the FoundationOne Heme panel and grouped by responses during phase 2 (A) or phase 1b (B). CRMRD, CR without minimal residual disease; MLFS, morphologic leukemia-free state; NE, not evaluated; PD, progressive disease; PR, partial remission; SD, stable disease; TF, treatment failure.

References

    1. Dohner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373(12):1136–1152.
    1. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209–2221.
    1. Dohner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–447.
    1. Stone RM, Larson RA, Dohner H. Midostaurin in FLT3-mutated acute myeloid leukemia. N Engl J Med. 2017;377(19):1903.
    1. Erba HP, Montesinos P, Vrhovac R, et al. EHA2022 Hybrid Congress. Vol. 6. HemaSphere; 2022. Quizartinib prolonged survival vs placebo plus intensive induction and consolidation therapy followed by single-agent continuation in patients aged 18-75 years with newly diagnosed FLT3-ITD+ AML; pp. 1–2.
    1. Burd A, Levine RL, Ruppert AS, et al. Precision medicine treatment in acute myeloid leukemia using prospective genomic profiling: feasibility and preliminary efficacy of the Beat AML Master Trial. Nat Med. 2020;26(12):1852–1858.
    1. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383(7):617–629.
    1. Montesinos P, Recher C, Vives S, et al. Ivosidenib and azacitidine in IDH1-mutated acute myeloid leukemia. N Engl J Med. 2022;386(16):1519–1531.
    1. Mims AS, Kohlschmidt J, Borate U, et al. A precision medicine classification for treatment of acute myeloid leukemia in older patients. J Hematol Oncol. 2021;14(1):96.
    1. Medeiros BC, Fathi AT, DiNardo CD, Pollyea DA, Chan SM, Swords R. Isocitrate dehydrogenase mutations in myeloid malignancies. Leukemia. 2017;31(2):272–281.
    1. Clark O, Yen K, Mellinghoff IK. Molecular pathways: isocitrate dehydrogenase mutations in cancer. Clin Cancer Res. 2016;22(8):1837–1842.
    1. Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553–567.
    1. Kats LM, Reschke M, Taulli R, et al. Proto-oncogenic role of mutant IDH2 in leukemia initiation and maintenance. Cell Stem Cell. 2014;14(3):329–341.
    1. Yen K, Travins J, Wang F, et al. AG-221, a first-in-class therapy targeting acute myeloid leukemia harboring oncogenic IDH2 mutations. Cancer Discov. 2017;7(5):478–493.
    1. DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med. 2018;378(25):2386–2398.
    1. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood. 2017;130(6):722–731.
    1. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood. 2015;126(3):291–299.
    1. DiNardo CD, Schuh AC, Stein EM, et al. Enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia (AG221-AML-005): a single-arm, phase 1b and randomised, phase 2 trial. Lancet Oncol. 2021;22(11):1597–1608.
    1. Venugopal S, Takahashi K, Daver N, et al. Efficacy and safety of enasidenib and azacitidine combination in patients with IDH2 mutated acute myeloid leukemia and not eligible for intensive chemotherapy. Blood Cancer J. 2022;12(1):10.
    1. Pollyea DA, DiNardo CD, Arellano ML, et al. Impact of venetoclax and azacitidine in treatment-naive patients with acute myeloid leukemia and IDH1/2 mutations. Clin Cancer Res. 2022;28(13):2753–2761.
    1. Kotecha N, Krutzik PO, Irish JM. Web-based analysis and publication of flow cytometry experiments. Curr Protoc Cytom. 2010;Chapter 10 Unit10.17.
    1. Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–404.
    1. Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):pl1.
    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. 2009;10(3):223–232.
    1. Rucker FG, Schlenk RF, Bullinger L, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood. 2012;119(9):2114–2121.
    1. Wang F, Morita K, DiNardo CD, et al. Leukemia stemness and co-occurring mutations drive resistance to IDH inhibitors in acute myeloid leukemia. Nat Commun. 2021;12(1):2607.
    1. Pollyea DA, Tallman MS, de Botton S, et al. Enasidenib, an inhibitor of mutant IDH2 proteins, induces durable remissions in older patients with newly diagnosed acute myeloid leukemia. Leukemia. 2019;33(11):2575–2584.

Source: PubMed

3
Suscribir