6-month follow-up of VIALE-C demonstrates improved and durable efficacy in patients with untreated AML ineligible for intensive chemotherapy (141/150)

Andrew H Wei, Panayiotis Panayiotidis, Pau Montesinos, Kamel Laribi, Vladimir Ivanov, Inho Kim, Jan Novak, Don A Stevens, Walter Fiedler, Maria Pagoni, Julie Bergeron, Stephen B Ting, Jing-Zhou Hou, Achilles Anagnostopoulos, Andrew McDonald, Vidhya Murthy, Takahiro Yamauchi, Jianxiang Wang, Brenda Chyla, Yan Sun, Qi Jiang, Wellington Mendes, John Hayslip, Courtney D DiNardo, Andrew H Wei, Panayiotis Panayiotidis, Pau Montesinos, Kamel Laribi, Vladimir Ivanov, Inho Kim, Jan Novak, Don A Stevens, Walter Fiedler, Maria Pagoni, Julie Bergeron, Stephen B Ting, Jing-Zhou Hou, Achilles Anagnostopoulos, Andrew McDonald, Vidhya Murthy, Takahiro Yamauchi, Jianxiang Wang, Brenda Chyla, Yan Sun, Qi Jiang, Wellington Mendes, John Hayslip, Courtney D DiNardo

Abstract

VIALE-C compared the safety and efficacy of venetoclax or placebo plus low-dose cytarabine (+LDAC) in patients with untreated AML ineligible for intensive chemotherapy. Overall, 211 patients were enrolled (n = 143, venetoclax; n = 68, placebo). At the primary analysis, the study did not meet its primary endpoint of a statistically significant improvement in overall survival (OS), however, ~60% of patients had been on study for ≤6-months. Here, we present an additional 6-months of follow-up of VIALE-C (median follow-up 17.5 months; range 0.1-23.5). Median OS was (venetoclax +LDAC vs. placebo +LDAC) 8.4 vs. 4.1 months (HR = 0.70, 95% CI 0.50,0.99; P = 0.040); a 30% reduction in the risk of death with venetoclax. Complete response (CR)/CR with incomplete hematologic recovery (CRi) rates were 48.3% vs. 13.2%. Transfusion independence rates (RBC) were 43% vs.19% and median event-free survival was 4.9 vs. 2.1 months (HR = 0.61; 95% CI 0.44,0.84; P = 0.002). These results represent improved efficacy over the primary analysis. Incidence of grade ≥3 adverse events were similar between study arms and overall safety profiles were comparable to the primary analysis. These data support venetoclax +LDAC as a frontline treatment option for patients with AML ineligible for intensive chemotherapy.This trial was registered at www.clinicaltrials.gov as #NCT03069352.

Conflict of interest statement

AbbVie-sponsored the study (NCT03069352), contributed to its design, collection, analysis, and interpretation of the data, and participated in the writing, review, and approval of the manuscript. All authors had access to relevant data. No honoraria or payments were made for authorship. Venetoclax (ABT-199/GDC-0199) is being developed in collaboration between AbbVie and Genentech. A.H.W.: Consulting for AbbVie, Amgen, Astellas Pharma, Celgene, Janssen, MacroGenics, Novartis, Roche, and Servier; research funding from AbbVie, Celgene, Novartis, and Servier; former employee of Walter and Eliza Hall Institute of Medical Research, which receives royalties related to venetoclax, and Dr. Wei is entitled to a fraction of these payments. P.P.: Grant/research support from AbbVie, Genesis, Novartis, and Roche; honoraria from AbbVie, Genesis, Gilead, Janssen, Novartis, and Roche. P.M.: Grant/research support from Astellas Pharma, Celgene, Daiichi Sankyo, Janssen, Karyopharm Therapeutics, Novartis, Pfizer, and Teva; speaker/advisory role for AbbVie, Celgene, Daiichi Sankyo, Incyte, Janssen, Karyopharm Therapeutics, Novartis, Pfizer, Teva, and Tolero; consulting for Agios, Astellas Pharma, Celgene, Daiichi Sankyo, Oryzon, and Tolero. K.L.: Grant/research support from AbbVie, Novartis, Roche, Sandoz, and Takeda; personal fees from AbbVie, Astellas Pharma, BeiGene, Celgene, iQone Healthcare Switzerland, Janssen, Novartis, and Sandoz. V.I.: Investigator in AbbVie-sponsored clinical trials. I.K.: Investigator in AbbVie-sponsored clinical trials. J.N.: Consulting/advisory role for Amgen, Novartis, Pfizer, Roche, and Takeda; travel expenses from Amgen and Janssen. D.A.S.: Investigator in AbbVie-sponsored clinical trials. W.F.: Membership on an entity’s board of directors or advisory committee for AbbVie, Amgen, ARIAD/Incyte, Celgene, Jazz Pharmaceuticals, MorphoSys AG, Novartis, and Pfizer; patents and royalties from Amgen; support for meeting attendance Amgen, Daiichi Sankyo, Gilead, Jazz Pharmaceuticals, and Servier; research funding from Amgen and Pfizer. M.P.: Speaker/advisory role for AbbVie, Amgen, Astellas Pharma, Genesis, Janssen, Novartis, and Pfizer. J.B.: Consulting for AbbVie, Amgen, Astellas Pharma, BMS, Jazz Pharmaceuticals, Novartis, and Pfizer; travel support from Amgen and Novartis. S.B.T.: Consulting for AbbVie; investigator in AbbVie-sponsored clinical trials. J.Z.H., A.A., and A.M.: Investigator in AbbVie-sponsored clinical trials. V.M.: Conference attendance support from AbbVie, Celgene, Janssen, Novartis, and Takeda; consulting for Celgene, Novartis, and Janssen. T.Y.: Research support/honoraria from, and advisory role for AbbVie, Astellas Pharma, Gilead, Janssen, Nippon Shinyaku, Otsuka, Pfizer, Solasia, SymBio, and Takeda. J.W.: Advisory role for AbbVie; research support from Celgene. B.C., Y.S., Q.J., W.M., and J.H.: Employees of AbbVie and may hold stock or stock options. C.D.D.: Research support from AbbVie/Genentech, Agios, BMS/Celgene, Calithera, Cleave, Daiichi Sankyo, Immune-Onc, and Loxo; consulting/advisory board member for AbbVie, Agios, Aprea, BMS/Celgene, Immune-Onc, Kura, Novartis, Takeda, and Notable Labs.

© 2021. The Author(s).

Figures

Fig. 1. Overall Survival.
Fig. 1. Overall Survival.
A OS at 6-month follow-up. Kaplan–Meier plot showing the OS rate of all patients over time, separated by treatment arm; the number of patients at risk for each time point is shown below the graph. Tick marks indicate censored data. Republished with permission of Elsevier Science & Technology Journals, from Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial, Wei et al., volume 135, issue 24, copyright 2021; permission conveyed through Copyright Clearance Center, Inc. B Subgroup analysis of investigator-assessed OS. HR is from the unstratified Cox proportional-hazards model. Data included are subjected to a cut-off date of August 15, 2019. Median (95% CI) and HR (95% CI) are calculated only for subgroups with available data. AML acute myeloid leukemia, CI confidence interval, HMA hypomethylating agent, HR hazard ratio, LDAC low-dose cytarabine, NA not assessed, OS overall survival.
Fig. 2. OS in patients treated with…
Fig. 2. OS in patients treated with venetoclax +LDAC achieving CR/CRi by best post-baseline MRD value (−3 vs. ≥10−3).
Kaplan–Meier plot showing the OS rate in patients treated with venetoclax +LDAC who achieved a CR/CRi response, stratified by best post-baseline MRD value (−3 vs. ≥10−3). Unable to graph MRD data for placebo + LDAC arm due to small sample size (data summarized in Table 5). Tick marks indicate censored data. CI confidence interval, CR complete response, CRi CR with incomplete hematologic recovery, LDAC low-dose cytarabine, NR not reached, OS overall survival, VEN, venetoclax, MRD minimal residual disease.

References

    1. Surveillance, Epidemiology, and End Results. Cancer Statistics. . Accessed June 2020.
    1. Khwaja A, Bjorkholm M, Gale RE, Levine RL, Jordan CT, Ehninger G, et al. Acute myeloid leukaemia. Nat Rev Dis Prim. 2016;2:16010. doi: 10.1038/nrdp.2016.10.
    1. Appelbaum FR, Gundacker H, Head DR, Slovak ML, Willman CL, Godwin JE, et al. Age and acute myeloid leukemia. Blood. 2006;107:3481–5. doi: 10.1182/blood-2005-09-3724.
    1. Medeiros BC, Satram-Hoang S, Hurst D, Hoang KQ, Momin F, Reyes C. Big data analysis of treatment patterns and outcomes among elderly acute myeloid leukemia patients in the United States. Ann Hematol. 2015;94:1127–38. doi: 10.1007/s00277-015-2351-x.
    1. Burnett AK, Milligan D, Prentice AG, Goldstone AH, McMullin MF, Hills RK, et al. A comparison of low-dose cytarabine and hydroxyurea with or without all-trans retinoic acid for acute myeloid leukemia and high-risk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer. 2007;109:1114–24. doi: 10.1002/cncr.22496.
    1. Kantarjian HM, Thomas XG, Dmoszynska A, Wierzbowska A, Mazur G, Mayer J, et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol. 2012;30:2670–7. doi: 10.1200/JCO.2011.38.9429.
    1. Dennis M, Hills RK, Russell NH, Copland M, Thomas I, McMullin MFF, et al. An evaluation of 17 years of low dose cytarabine as therapy for AML patients not fit for intensive treatment, including patients with adverse cytogenetics, shows improving survival, potential underutilisation and highlights the need for new therapy. Blood. 2017;130:3874.
    1. Cortes JE, Heidel FH, Fiedler W, Smith BD, Robak T, Montesinos P, et al. Survival outcomes and clinical benefit in patients with acute myeloid leukemia treated with glasdegib and low-dose cytarabine according to response to therapy. J Hematol Oncol. 2020;13:92. doi: 10.1186/s13045-020-00929-8.
    1. Souers AJ, Leverson JD, Boghaert ER, Ackler SL, Catron ND, Chen J, et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med. 2013;19:202–8. doi: 10.1038/nm.3048.
    1. Pan R, Ruvolo VR, Wei J, Konopleva M, Reed JC, Pellecchia M, et al. Inhibition of Mcl-1 with the pan–Bcl-2 family inhibitor (–)BI97D6 overcomes ABT-737 resistance in acute myeloid leukemia. Blood. 2015;126:363–72. doi: 10.1182/blood-2014-10-604975.
    1. Wei AH, Strickland SA, Jr, Hou JZ, Fiedler W, Lin TL, Walter RB, 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. 2019;37:1277–84. doi: 10.1200/JCO.18.01600.
    1. Wei AH, Montesinos P, Ivanov V, DiNardo CD, Novak J, Laribi K, et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood. 2020;135:2137–45. doi: 10.1182/blood.2020004856.
    1. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127:2391–405. doi: 10.1182/blood-2016-03-643544.
    1. Howard SC, Jones DP, Pui CH. The tumor lysis syndrome. N Engl J Med. 2011;364:1844–54. doi: 10.1056/NEJMra0904569.
    1. Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129:424–47. doi: 10.1182/blood-2016-08-733196.
    1. Cheson BD, Bennett JM, Kopecky KJ, Büchner 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. doi: 10.1200/JCO.2003.04.036.
    1. Schuurhuis GJ, Heuser M, Freeman S, Béné MC, Buccisano F, Cloos J, et al. Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD working party. Blood. 2018;131:1275–91. doi: 10.1182/blood-2017-09-801498.
    1. Shallis RM, Boddu PC, Bewersdorf JP, Zeidan AM. The golden age for patients in their golden years: the progressive upheaval of age and the treatment of newly-diagnosed acute myeloid leukemia. Blood Rev. 2020;40:100639. doi: 10.1016/j.blre.2019.100639.
    1. DiNardo CD, Jonas BA, Pullarkat V, Thirman MJ, Garcia JS, Wei AH, et al. Azacitidine and venetoclax in previously untreated acute myeloid. Leuk N Engl J Med. 2020;383:617–29. doi: 10.1056/NEJMoa2012971.

Source: PubMed

3
Sottoscrivi