DNMT3A mutation is associated with increased age and adverse outcome in adult T-cell acute lymphoblastic leukemia

Jonathan Bond, Aurore Touzart, Stéphane Leprêtre, Carlos Graux, Mario Bargetzi, Ludovic Lhermitte, Guillaume Hypolite, Thibaut Leguay, Yosr Hicheri, Gaëlle Guillerm, Karin Bilger, Véronique Lhéritier, Mathilde Hunault, Françoise Huguet, Yves Chalandon, Norbert Ifrah, Elizabeth Macintyre, Hervé Dombret, Vahid Asnafi, Nicolas Boissel, Jonathan Bond, Aurore Touzart, Stéphane Leprêtre, Carlos Graux, Mario Bargetzi, Ludovic Lhermitte, Guillaume Hypolite, Thibaut Leguay, Yosr Hicheri, Gaëlle Guillerm, Karin Bilger, Véronique Lhéritier, Mathilde Hunault, Françoise Huguet, Yves Chalandon, Norbert Ifrah, Elizabeth Macintyre, Hervé Dombret, Vahid Asnafi, Nicolas Boissel

Abstract

The prognostic implications of DNMT3A genotype in T-cell acute lymphoblastic leukemia are incompletely understood. We performed comprehensive genetic and clinico-biological analyses of T-cell acute lymphoblastic leukemia patients with DNMT3A mutations treated during the GRAALL-2003 and -2005 studies. Eighteen of 198 cases (9.1%) had DNMT3A alterations. Two patients also had DNMT3A mutations in non-leukemic cell DNA, providing the first potential evidence of age-related clonal hematopoiesis in T-cell acute lymphoblastic leukemia. DNMT3A mutation was associated with older age (median 43.9 years vs 29.4 years, P<0.001), immature T-cell receptor genotype (53.3% vs 24.4%, P=0.016) and lower remission rates (72.2% mutated vs 94.4% non-mutated, P=0.006). DNMT3A alterations were significantly associated with worse clinical outcome, with higher cumulative incidence of relapse (HR 2.33, 95% CI: 1.05-5.16, P=0.037) and markedly poorer event-free survival (HR 3.22, 95% CI: 1.81-5.72, P<0.001) and overall survival (HR 2.91, 95% CI: 1.56-5.43, P=0.001). Adjusting for age as a covariate, or restricting the analysis to patients over 40 years, who account for almost 90% of DNMT3A-mutated cases, did not modify these observations. In multivariate analysis using the risk factors that were used to stratify treatment during the GRAALL studies, DNMT3A mutation was significantly associated with shorter event-free survival (HR 2.33, 95% CI: 1.06 - 4.04, P=0.02). Altogether, these results identify DNMT3A genotype as a predictor of aggressive T-cell acute lymphoblastic leukemia biology. The GRAALL-2003 and -2005 studies were registered at http://www.ClinicalTrials.gov as #NCT00222027 and #NCT00327678, respectively.

Copyright© 2019 Ferrata Storti Foundation.

Figures

Figure 1.
Figure 1.
DNMT3A mutations in T-cell acute lymphoblastic leukemia. (A) Schematic representation of the 21 mutations detected in this study. Further patient-specific details are provided in Online Supplementary Table S3. (B) Comparison of the mutational genotypes of DNMT3A altered (n=18) and DNMT3A wild-type (n=180) T-cell acute lymphoblastic leukemia. Percentage frequencies in each group are depicted. Functional categories are listed in bold.
Figure 2.
Figure 2.
Evidence of DNMT3A mutations in non-leukemic DNA. (A) Direct sequencing of DNMT3A exons 14 and 15, NOTCH1 and NRAS in diagnostic (left panels) and remission (right panels) samples. (B) Mutational assessment of DNA extracted from leukemic and non-leukemic fractions of samples from patients with T-cell acute lymphoblastic leukemia. Sequencing results of DNMT3A and NOTCH1 in leukemic (left panels) and non-leukemic (right panels) DNA are shown. Cases are numbered according to the listing in Online Supplementary Table S3.
Figure 3.
Figure 3.
DNMT3A mutation correlates with poor outcome in T-cell acute lymphoblastic leukemia. Comparisons of outcomes for patients with (n=18) and without (n=180) DNMT3A mutations are shown for: (A) cumulative incidence of relapse; (B) event-free survival; and (C) overall survival. The 5-year results were as follows: cumulative incidence of relapse 53.9% mutated vs. 28.7% non-mutated; event-free survival 27.8% mutated vs. 61.0% non-mutated; overall survival 38.8% mutated vs. 68.7% non-mutated. P values are indicated.
Figure 4.
Figure 4.
DNMT3A genotype predicts outcome in the age group of patients at risk of mutation. Comparisons of outcomes for patients with (n=16) and without (n=34) mutations in patients >40 years are shown for: (A) cumulative incidence of relapse; (B) event-free survival; and (C) overall survival. The 5-year results were as follows: cumulative incidence of relapse 58.3% mutated vs. 21.7% non-mutated; event-free survival, 25.0% mutated vs. 56.7% non-mutated; overall survival 37.5% mutated vs. 62.1% non-mutated. P values are indicated.

References

    1. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424–2433.
    1. Renneville A, Boissel N, Nibourel O, et al. Prognostic significance of DNA methyltransferase 3A mutations in cytogenetically normal acute myeloid leukemia: a study by the Acute Leukemia French Association. Leukemia. 2012;26(6):1247–1254.
    1. Gale RE, Lamb K, Allen C, et al. Simpson’s paradox and the impact of different DNMT3A mutations on outcome in younger adults with acute myeloid leukemia. J Clin Oncol. 2015;33(18):2072–2083.
    1. Marcucci G, Metzeler KH, Schwind S, et al. Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia. J Clin Oncol. 2012;30(7): 742–750.
    1. Yan XJ, Xu J, Gu ZH, et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat Genet. 2013;43(4):309–315.
    1. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 2014;28(2):241–247.
    1. Stegelmann F, Bullinger L, Schlenk RF, et al. DNMT3A mutations in myeloproliferative neoplasms. Leukemia. 2011;25(7):1217–1219.
    1. Jankowska AM, Makishima H, Tiu RV, et al. Mutational spectrum analysis of chronic myelomonocytic leukemia includes genes associated with epigenetic regulation: UTX, EZH2, and DNMT3A. Blood. 2011;118(14): 3932–3941.
    1. Roller A, Grossmann V, Bacher U, et al. Landmark analysis of DNMT3A mutations in hematological malignancies. Leukemia. 2013;27(7):1573–1578.
    1. Van Vlierberghe P, Ambesi-Impiombato A, Perez-Garcia A, et al. ETV6 mutations in early immature human T cell leukemias. J Exp Med. 2011;208(13):2571–2579.
    1. Grossmann V, Haferlach C, Weissmann S, et al. The molecular profile of adult T-cell acute lymphoblastic leukemia: mutations in RUNX1 and DNMT3A are associated with poor prognosis in T-ALL. Genes Chromosomes Cancer. 2013;52(4):410–422.
    1. Neumann M, Heesch S, Schlee C, et al. Whole-exome sequencing in adult ETP-ALL reveals a high rate of DNMT3A mutations. Blood. 2013;121(23):4749–4752.
    1. Van Vlierberghe P, Ambesi-Impiombato A, De Keersmaecker K, et al. Prognostic relevance of integrated genetic profiling in adult T-cell acute lymphoblastic leukemia. Blood. 2013;122(1):74–82.
    1. Couronne L, Bastard C, Bernard OA. TET2 and DNMT3A mutations in human T-cell lymphoma. N Engl J Med. 2012;366(1):95–96.
    1. Sakata-Yanagimoto M, Enami T, Yoshida K, et al. Somatic RHOA mutation in angioimmunoblastic T cell lymphoma. Nat Genet. 2014;46(2):171–175.
    1. Choi J, Goh G, Walradt T, et al. Genomic landscape of cutaneous T cell lymphoma. Nat Genet. 2015;47(9):1011–1019.
    1. Ho PA, Kutny MA, Alonzo TA, et al. Leukemic mutations in the methylation-associated genes DNMT3A and IDH2 are rare events in pediatric AML: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2011;57(2):204–209.
    1. Huether R, Dong L, Chen X, et al. The landscape of somatic mutations in epigenetic regulators across 1,000 paediatric cancer genomes. Nat Commun. 2014;5:3630.
    1. Shiba N, Taki T, Park MJ, et al. DNMT3A mutations are rare in childhood acute myeloid leukaemia, myelodysplastic syndromes and juvenile myelomonocytic leukaemia. Br J Haematol. 2012;156(3):413–414.
    1. Shen Y, Zhu YM, Fan X, et al. Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood. 2011;118(20): 5593–5603.
    1. Ribeiro AF, Pratcorona M, Erpelinck-Verschueren C, et al. Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia. Blood. 2012;119(24):5824–5831.
    1. Tie R, Zhang T, Fu H, et al. Association between DNMT3A mutations and prognosis of adults with de novo acute myeloid leukemia: a systematic review and meta-analysis. PloS One. 2014;9(6):e93353.
    1. Zhang J, Ding L, Holmfeldt L, et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature. 2012;481(7380):157–163.
    1. Bond J, Graux C, Lhermitte L, et al. Early response-based therapy stratification improves survival in adult early thymic precursor acute lymphoblastic leukemia: a group for research on adult acute lymphoblastic leukemia study. J Clin Oncol. 2017;35(23):2683–2691.
    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. Lin ME, Hou HA, Tsai CH, et al. Dynamics of DNMT3A mutation and prognostic relevance in patients with primary myelodysplastic syndrome. Clin Epigenetics. 2018;10:42.
    1. Genovese G, Kahler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371(26):2477–2487.
    1. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488–2498.
    1. Xie M, Lu C, Wang J, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med. 2014;20(12):1472–1478.
    1. Shlush LI, Zandi S, Mitchell A, et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature. 2014;506(7488):328–333.
    1. Corces-Zimmerman MR, Hong WJ, Weissman IL, Medeiros BC, Majeti R. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Natl Acad Sci U S A. 2014;111(7):2548–2553.
    1. Asnafi V, Beldjord K, Boulanger E, et al. Analysis of TCR, pT alpha, and RAG-1 in T-acute lymphoblastic leukemias improves understanding of early human T-lymphoid lineage commitment. Blood. 2003;101(7): 2693–2703.
    1. Coustan-Smith E, Mullighan CG, Onciu M, et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol. 2009;10(2):147–156.
    1. Huguet F, Leguay T, Raffoux E, et al. Pediatric-inspired therapy in adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: the GRAALL-2003 study. J Clin Oncol. 2009;27(6):911–918.
    1. Bond J, Marchand T, Touzart A, et al. An early thymic precursor phenotype predicts outcome exclusively in HOXA-overexpressing adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia study. Haematologica. 2016;101(6):732–740.
    1. Eguchi-Ishimae M, Eguchi M, Kempski H, Greaves M. NOTCH1 mutation can be an early, prenatal genetic event in T-ALL. Blood. 2008;111(1):376–378.
    1. Trinquand A, Tanguy-Schmidt A, Ben Abdelali R, et al. Toward a NOTCH1/FBXW7/RAS/PTEN-based oncogenetic risk classification of adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia study. J Clin Oncol. 2013;31(34):4333–4342.
    1. Huguet Fo, Leguay T, Thomas X, et al. The upper age limit for a pediatric-inspired therapy in younger adults with Ph-negative acute lymphoblastic leukemia (ALL)? Analysis of the GRAALL-2005 study. 2016:762–762.
    1. Metzeler KH, Walker A, Geyer S, et al. DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia. Leukemia. 2012;26(5): 1106–1107.

Source: PubMed

3
구독하다