Adding venetoclax to fludarabine/busulfan RIC transplant for high-risk MDS and AML is feasible, safe, and active

Jacqueline S Garcia, Haesook T Kim, H Moses Murdock, Corey S Cutler, Jennifer Brock, Mahasweta Gooptu, Vincent T Ho, John Koreth, Sarah Nikiforow, Rizwan Romee, Roman Shapiro, Fiona Loschi, Jeremy Ryan, Geoffrey Fell, Hannah Q Karp, Fabienne Lucas, Annette S Kim, Danielle Potter, Thelma Mashaka, Richard M Stone, Daniel J DeAngelo, Anthony Letai, R Coleman Lindsley, Robert J Soiffer, Joseph H Antin, Jacqueline S Garcia, Haesook T Kim, H Moses Murdock, Corey S Cutler, Jennifer Brock, Mahasweta Gooptu, Vincent T Ho, John Koreth, Sarah Nikiforow, Rizwan Romee, Roman Shapiro, Fiona Loschi, Jeremy Ryan, Geoffrey Fell, Hannah Q Karp, Fabienne Lucas, Annette S Kim, Danielle Potter, Thelma Mashaka, Richard M Stone, Daniel J DeAngelo, Anthony Letai, R Coleman Lindsley, Robert J Soiffer, Joseph H Antin

Abstract

Adding the selective BCL-2 inhibitor venetoclax to reduced-intensity conditioning chemotherapy (fludarabine and busulfan [FluBu2]) may enhance antileukemic cytotoxicity and thereby reduce the risk of posttransplant relapse. This phase 1 study investigated the recommended phase 2 dose (RP2D) of venetoclax, a BCL-2 selective inhibitor, when added to FluBu2 in adult patients with high-risk acute myeloid leukemia (AML), myelodysplastic syndromes (MDS), and MDS/myeloproliferative neoplasms (MPN) undergoing transplant. Patients received dose-escalated venetoclax (200-400 mg daily starting day -8 for 6-7 doses) in combination with fludarabine 30 mg/m2 per day for 4 doses and busulfan 0.8 mg/kg twice daily for 8 doses on day -5 to day -2 (FluBu2). Transplant related-toxicity was evaluated from the first venetoclax dose on day -8 to day 28. Twenty-two patients were treated. At study entry, 5 patients with MDS and MDS/MPN had 5% to 10% marrow blasts, and 18 (82%) of 22 had a persistent detectable mutation. Grade 3 adverse events included mucositis, diarrhea, and liver transaminitis (n = 3 each). Neutrophil/platelet recovery and acute/chronic graft-versus-host-disease rates were similar to those of standard FluBu2. No dose-limiting toxicities were observed. The RP2D of venetoclax was 400 mg daily for 7 doses. With a median follow-up of 14.7 months (range, 8.6-24.8 months), median overall survival was not reached, and progression-free survival was 12.2 months (95% confidence interval, 6.0-not estimable). In patients with high-risk AML, MDS, and MDS/MPN, adding venetoclax to FluBu2 was feasible and safe. To further address relapse risk, assessment of maintenance therapy after venetoclax plus FluBu2 transplant is ongoing. This study was registered at clinicaltrials.gov as #NCT03613532.

© 2021 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.
Baseline disease and genetic profiles of study patients. Comutation plot of diagnostic mutations amenable to MRD tracking. Columns represent individual patients, and rows represent clinical variables or the presence of mutation(s) identified at diagnosis or mutations at screening with VAF ≥2%. This VAF cutoff is suggestive of a diagnostic mutation that was not confirmed at diagnosis due to lack of diagnostic sample or technical assay differences. Asterisk represents a BCR-ABL–mutated case (detected by reverse transcriptase polymerase chain reaction).
Figure 2.
Figure 2.
Engraftment, donor chimerism, and GVHD rate after venetoclax plus FluBu2 allo-HCT. (A) No evidence for impairment in neutrophil (red) and platelet (blue) engraftment. (B) CD3 T-cell donor chimerism was evaluated at days 28 and 100. In the T-cell chimerism plot, individual patients’ chimerism are connected by lines at days 28 and 100. The black solid line connects 2 medians at days 28 and 100 (P = .0015). (C) Risk for acute (blue) and chronic (red) GVHD were plotted. No grade IV acute GVHD (aGVHD) events occurred. cGVHD, chronic GVHD; D, day; mod/sev, moderate/severe.
Figure 3.
Figure 3.
Outcomes after venetoclax plus FluBu2 allo-HCT. Kaplan-Meier estimate and log-rank test of probability of OS (blue) and PFS (red) (A) and probability of NRM (blue) and relapse (red) (B). 95% CIs are reported for each outcome. Probability of OS (C) and PFS (D) in relation to ECOG PS of 0 to 1 (blue) vs ECOG PS of 2 (red).
Figure 4.
Figure 4.
Serial genetic MRD monitoring before and after transplant. (A) NGS swimmer plot. Each bar represents a patient, with the length of the gray bars indicating follow-up time. Symbols indicate NGS status at serial time points, relapse, and death. Patients arranged according to NGS status at days 28/100, and then by follow-up time. Mutation VAF, relapse, and post-relapse course in patients 1 and 5 are shown. (B) Patient 1 had a baseline mutation in TP53, relapsed >6 months’ posttransplant, and acquired new mutations in NF1, CSF3R, and KRAS. (C) Patient 5 was another TP53-mutated patient who relapsed but later achieved durable CR2 after salvage therapy (decitabine and venetoclax) followed by donor lymphocyte infusion. TP53 VAF and read level support indicated at time points leading up to and including relapse. CR2 indicates timepoint of second complete remission achieved after two cycles of decitabine and venetoclax.

References

    1. Armand P, Kim HT, Zhang MJ, et al. . Classifying cytogenetics in patients with acute myelogenous leukemia in complete remission undergoing allogeneic transplantation: a Center for International Blood and Marrow Transplant Research study. Biol Blood Marrow Transplant. 2012;18(2):280-288.
    1. Lindsley RC, Saber W, Mar BG, et al. . Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation. N Engl J Med. 2017; 376(6):536-547.
    1. Duncavage EJ, Jacoby MA, Chang GS, et al. . Mutation clearance after transplantation for myelodysplastic syndrome. N Engl J Med. 2018;379(11):1028-1041.
    1. Festuccia M, Deeg HJ, Gooley TA, et al. . Minimal identifiable disease and the role of conditioning intensity in hematopoietic cell transplantation for myelodysplastic syndrome and acute myelogenous leukemia evolving from myelodysplastic syndrome. Biol Blood Marrow Transplant. 2016;22(7):1227-1233.
    1. Hourigan CS, Dillon LW, Gui G, et al. . Impact of conditioning intensity of allogeneic transplantation for acute myeloid leukemia with genomic evidence of residual disease. J Clin Oncol. 2020;38(12):1273-1283.
    1. Heuser M, Heida B, Büttner K, et al. . Posttransplantation MRD monitoring in patients with AML by next-generation sequencing using DTA and non-DTA mutations. Blood Adv. 2021;5(9):2294-2304.
    1. Devine SM, Owzar K, Blum W, et al. . Phase II study of allogeneic transplantation for older patients with acute myeloid leukemia in first complete remission using a reduced-intensity conditioning regimen: results from Cancer and Leukemia Group B 100103 (Alliance for Clinical Trials in Oncology)/Blood and Marrow Transplant Clinical Trial Network 0502. J Clin Oncol. 2015;33(35):4167-4175.
    1. Vo TT, Ryan J, Carrasco R, et al. . Relative mitochondrial priming of myeloblasts and normal HSCs determines chemotherapeutic success in AML. Cell. 2012;151(2):344-355.
    1. Bhola PD, Mar BG, Lindsley RC, et al. . Functionally identifiable apoptosis-insensitive subpopulations determine chemoresistance in acute myeloid leukemia. J Clin Invest. 2016;126(10):3827-3836.
    1. Wei AH, Montesinos P, Ivanov V, et al. . Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood. 2020;135(24):2137-2145.
    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. Döhner 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. Lindsley RC, Mar BG, Mazzola E, et al. . Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood. 2015;125(9):1367-1376.
    1. Schuurhuis GJ, Heuser M, Freeman S, et al. . Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD Working Party. Blood. 2018;131(12):1275-1291.
    1. Walter RB, Gooley TA, Wood BL, et al. . Impact of pretransplantation minimal residual disease, as detected by multiparametric flow cytometry, on outcome of myeloablative hematopoietic cell transplantation for acute myeloid leukemia. J Clin Oncol. 2011;29(9):1190-1197.
    1. Such E, Cervera J, Costa D, et al. . Cytogenetic risk stratification in chronic myelomonocytic leukemia. Haematologica. 2011;96(3):375-383.
    1. Itzykson R, Kosmider O, Renneville A, et al. . Prognostic score including gene mutations in chronic myelomonocytic leukemia. J Clin Oncol. 2013;31(19):2428-2436.
    1. Przepiorka D, Weisdorf D, Martin P, et al. . 1994 Consensus conference on acute GVHD grading. Bone Marrow Transplant. 1995;15(6):825-828.
    1. Jagasia MH, Greinix HT, Arora M, et al. . National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant. 2015;21(3):389-401.e1.
    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. Savona MR, Malcovati L, Komrokji R, et al. ; MDS/MPN International Working Group . An international consortium proposal of uniform response criteria for myelodysplastic/myeloproliferative neoplasms (MDS/MPN) in adults. Blood. 2015;125(12):1857-1865.
    1. Kluk MJ, Lindsley RC, Aster JC, et al. . Validation and implementation of a custom next-generation sequencing clinical assay for hematologic malignancies. J Mol Diagn. 2016;18(4):507-515.
    1. Gray RJ. A class of K-sample tests for comparing the cumulative incidence of a competing risk. Ann Stat. 1988;16(3):1141-1154.
    1. Scott BL, Pasquini MC, Fei M, et al. . Myeloablative versus reduced-intensity conditioning for hematopoietic cell transplantation in acute myelogenous leukemia and myelodysplastic syndromes—long-term follow-up of the BMT CTN 0901 Clinical Trial. Transplant Cell Ther. 2021;27(6):483.e1-483.e6. DOI: .
    1. Craddock C, Jackson A, Loke J, et al. . Augmented reduced-intensity regimen does not improve postallogeneic transplant outcomes in acute myeloid leukemia. Clin Oncol. 2021;39(7):768-778.
    1. Konopleva M, Pollyea DA, Potluri J, et al. . Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 2016;6(10):1106-1117.
    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. 2019;133(1):7-17.
    1. Kekre N, Kim HT, Thanarajasingam G, et al. . Efficacy of immune suppression tapering in treating relapse after reduced intensity allogeneic stem cell transplantation. Haematologica. 2015;100(9):1222-1227.
    1. Chen YB, Li S, Lane AA, et al. . Phase I trial of maintenance sorafenib after allogeneic hematopoietic stem cell transplantation for fms-like tyrosine kinase 3 internal tandem duplication acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(12):2042-2048.
    1. Goodyear OC, Dennis M, Jilani NY, et al. . Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). Blood. 2012;119(14):3361-3369.
    1. Sánchez-Abarca LI, Gutierrez-Cosio S, Santamaría C, et al. . Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting. Blood. 2010;115(1):107-121.
    1. Oran B, de Lima M, Garcia-Manero G, et al. . A phase 3 randomized study of 5-azacitidine maintenance vs observation after transplant in high-risk AML and MDS patients [published correction appears in Blood Adv. 2021;5(6):1755-1756]. Blood Adv. 2020;4(21):5580-5588.
    1. Bhatt S, Pioso MS, Olesinski EA, et al. . Reduced mitochondrial apoptotic priming drives resistance to BH3 mimetics in acute myeloid leukemia. Cancer Cell. 2020;38(6):872-890.e6.
    1. Chua CC, Roberts AW, Reynolds J, et al. . Chemotherapy and Venetoclax in Elderly Acute Myeloid Leukemia Trial (CAVEAT): a phase Ib dose-escalation study of venetoclax combined with modified intensive chemotherapy. J Clin Oncol. 2020;38(30):3506-3517.
    1. Luskin MR, DeAngelo DJ. Mini-hyper-CVD combinations for older adults: results of recent trials and a glimpse into the future. Clin Lymphoma Myeloma Leuk. 2020;20(suppl 1):S44-S47.
    1. Pollyea DA, Amaya M, Strati P, Konopleva MY. Venetoclax for AML: changing the treatment paradigm [published correction appears in Blood Adv. 2020;4(6):1020]. Blood Adv. 2019;3(24):4326-4335.
    1. DiNardo CD, Lachowiez CA, Takahashi K, et al. . Venetoclax combined with FLAG-IDA induction and consolidation in newly diagnosed and relapsed or refractory acute myeloid leukemia. J Clin Oncol. 2021;39(25):2768-2778.
    1. Ciurea SO, Kongtim P, Varma A, et al. . Is there an optimal conditioning for older patients with AML receiving allogeneic hematopoietic cell transplantation? Blood. 2020;135(6):449-452.
    1. Beelen DW, Trenschel R, Stelljes M, et al. . Treosulfan or busulfan plus fludarabine as conditioning treatment before allogeneic haemopoietic stem cell transplantation for older patients with acute myeloid leukaemia or myelodysplastic syndrome (MC-FludT.14/L): a randomised, non-inferiority, phase 3 trial. Lancet Haematol. 2020;7(1):e28-e39.
    1. Bolaños-Meade J, Reshef R, Fraser R, et al. . Three prophylaxis regimens (tacrolimus, mycophenolate mofetil, and cyclophosphamide; tacrolimus, methotrexate, and bortezomib; or tacrolimus, methotrexate, and maraviroc) versus tacrolimus and methotrexate for prevention of graft-versus-host disease with haematopoietic cell transplantation with reduced-intensity conditioning: a randomised phase 2 trial with a non-randomised contemporaneous control group (BMT CTN 1203). Lancet Haematol. 2019;6(3):e132-e143.
    1. De Jong CN, Meijer E, Bakunina K, et al. . Post-transplantation cyclophosphamide after allogeneic hematopoietic stem cell transplantation: results of the prospective randomized HOVON-96 trial in recipients of matched related and unrelated donors. Blood. 2019;134(suppl 1):1.

Source: PubMed

3
Se inscrever