Outcomes of TP53-mutant acute myeloid leukemia with decitabine and venetoclax

Kunhwa Kim, Abhishek Maiti, Sanam Loghavi, Rasoul Pourebrahim, Tapan M Kadia, Caitlin R Rausch, Ken Furudate, Naval G Daver, Yesid Alvarado, Maro Ohanian, Koji Sasaki, Nicholas J Short, Koichi Takahashi, Musa Yilmaz, Guilin Tang, Farhad Ravandi, Hagop M Kantarjian, Courtney D DiNardo, Marina Y Konopleva, Kunhwa Kim, Abhishek Maiti, Sanam Loghavi, Rasoul Pourebrahim, Tapan M Kadia, Caitlin R Rausch, Ken Furudate, Naval G Daver, Yesid Alvarado, Maro Ohanian, Koji Sasaki, Nicholas J Short, Koichi Takahashi, Musa Yilmaz, Guilin Tang, Farhad Ravandi, Hagop M Kantarjian, Courtney D DiNardo, Marina Y Konopleva

Abstract

Background: TP53 mutation (TP53mut ) confers an adverse prognosis in acute myeloid leukemia (AML). Venetoclax with hypomethylating agents is a current standard for older patients; however, recent reports suggest that TP53mut confers resistance to venetoclax. The authors investigated the outcomes of patients with TP53mut AML who were treated with a 10-day decitabine and venetoclax (DEC10-VEN) (ClinicalTrials.gov identifier NCT03404193).

Methods: Patients with newly diagnosed AML received decitabine 20 mg/m2 for 10 days every 4 to 6 weeks for induction, followed by decitabine for 5 days after response. The venetoclax dose was 400 mg daily. TP53mut was identified in bone marrow samples using next-generation sequencing, with sensitivity of 5%. Outcomes were analyzed according to European LeukemiaNet 2017 guidelines.

Results: Among 118 patients (median age, 72 years; age range, 49-89 years), 63 (53%) had secondary AML, 39 (33%) had AML with complex karyotype, and 35 (30%) had TP53mut AML. The median TP53 variant allele frequency was 32% (interquartile range, 16%-65%), 8 patients (23%) had only a single TP53 mutation, 15 (43%) had multiple mutations, and 12 (34%) had mutation and deletion. Outcomes were significantly worse in patients who had TP53mut AML compared with those who had wild-type TP53 AML, with an overall response rate of 66% vs 89% (P = .002), a complete response/complete response with incomplete hematologic recovery rate of 57% vs 77% (P = .029), and a 60-day mortality of 26% vs 4% (P < .001), respectively. Patients with TP53mut versus wild-type TP53 had shorter overall survival at 5.2 versus 19.4 months, respectively (hazard ratio, 4.67; 95% CI, 2.44-8.93; P < .0001), and shorter relapse-free survival at 3.4 versus 18.9 months (hazard ratio, 4.80; 95% CI, 1.97-11.69; P < .0001), respectively. Outcomes with DEC10-VEN in patients with TP53mut AML were comparable to historical results with 10-day decitabine alone.

Conclusions: Patients with TP53mut AML have lower response rates and shorter survival with DEC10-VEN.

Keywords: TP53; acute myeloid leukemia (AML); decitabine; outcome; venetoclax.

© 2021 American Cancer Society.

References

    1. Olivier M, Hollstein M, Hainaut P. TP53 mutations in human cancers: origins, consequences, and clinical use. Cold Spring Harb Perspect Biol. 2010;2:a001008.
    1. Bowen D, Groves MJ, Burnett AK, et al. TP53 gene mutation is frequent in patients with acute myeloid leukemia and complex karyotype, and is associated with very poor prognosis. Leukemia. 2009;23:203-206.
    1. Ok CY, Patel KP, Garcia-Manero G, et al. Mutational profiling of therapy-related myelodysplastic syndromes and acute myeloid leukemia by next generation sequencing, a comparison with de novo diseases. Leuk Res. 2015;39:348-354.
    1. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374:2209-2221.
    1. Kadia TM, Jain P, Ravandi F, et al. TP53 mutations in newly diagnosed acute myeloid leukemia: clinicomolecular characteristics, response to therapy, and outcomes. Cancer. 2016;122:3484-3491.
    1. Hunter AM, Sallman DA. Current status and new treatment approaches in TP53 mutated AML. Best Pract Res Clin Haematol. 2019;32:134-144.
    1. Montalban-Bravo G, Takahashi K, Garcia-Manero G. Decitabine in TP53-mutated AML. N Engl J Med. 2017;376:796-797.
    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. 2019;6:e29-e37.
    1. Blum W, Garzon R, Klisovic RB, et al. Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine. PNAS. 2010;107:7473-7478.
    1. Bhatnagar B, Duong VH, Gourdin TS, et al. Ten-day decitabine as initial therapy for newly diagnosed patients with acute myeloid leukemia unfit for intensive chemotherapy. Leuk Lymphoma. 2014;55:1533-1537.
    1. Welch JS, Petti AA, Miller CA, et al. TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N Engl J Med. 2016;375:2023-2036.
    1. DiNardo CD, Pratz KW, Letai A, et al. Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study. Lancet Oncol. 2018;19:216-228.
    1. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383:617-629.
    1. DiNardo CD, Maiti A, Rausch CR, et al. 10-day decitabine with venetoclax for newly diagnosed intensive chemotherapy ineligible, and relapsed or refractory acute myeloid leukaemia: a single-centre, phase 2 trial. Lancet Haematol. 2020;7:e724-e736.
    1. Nechiporuk T, Kurtz SE, Nikolova O, et al. The TP53 apoptotic network is a primary mediator of resistance to BCL2 inhibition in AML cells. Cancer Discov. 2019;9:910-924.
    1. DiNardo CD, Tiong IS, Quaglieri A, et al. Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood. 2020;135:791-803.
    1. Aldoss I, Zhang J, Pillai R, et al. Venetoclax and hypomethylating agents in TP53-mutated acute myeloid leukaemia. Br J Haematol. 2019;187:e45-e48.
    1. Wang YW, Tsai CH, Lin CC, et al. Cytogenetics and mutations could predict outcome in relapsed and refractory acute myeloid leukemia patients receiving BCL-2 inhibitor venetoclax. Ann Hematol. 2020;99:501-511.
    1. Quesada AE, Routbort MJ, DiNardo CD, et al. DDX41 mutations in myeloid neoplasms are associated with male gender, TP53 mutations and high-risk disease. Am J Hematol. 2019;94:757-766.
    1. Xu J, Jorgensen JL, Wang SA. How do we use multicolor flow cytometry to detect minimal residual disease in acute myeloid leukemia? Clin Lab Med. 2017;37:787-802.
    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;129:424-447.
    1. Maiti A, DiNardo CD, Kadia TM, et al. Ten-day decitabine with venetoclax versus intensive chemotherapy in relapsed or refractory acute myeloid leukemia: a propensity score matched analysis. Blood. 2020;136(suppl 1):30-33.
    1. Boddu P, Kantarjian H, Ravandi F, et al. Outcomes with lower intensity therapy in TP53-mutated acute myeloid leukemia. Leuk Lymphoma. 2018;59:2238-2241.
    1. Bewersdorf JP, Shallis RM, Gowda L, et al. Clinical outcomes of patients (pts) with TP53-mutated acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS): a single center experience. Blood. 2019;134(suppl 1):5173.
    1. Short NJ, Montalban-Bravo G, Hwang H, et al. Prognostic and therapeutic impacts of mutant TP53 variant allelic frequency in newly diagnosed acute myeloid leukemia. Blood Adv. 2020;4:5681-5689.
    1. Pan R, Ruvolo V, Mu H, et al. Synthetic lethality of combined Bcl-2 inhibition and p53 activation in AML: mechanisms and superior antileukemic efficacy. Cancer Cell. 2017;32:748-760.e6.
    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. 2020;26:1549-1556.

Source: PubMed

3
Subscribe