DNMT3A mutations in acute myeloid leukemia

Olga Blau, Aline Kuehnel,, Franziska Behrenbeck, Nikola Suckert,, Igor Wolfgang Blau

Abstract


Abstract

Acute myeloid leukemia (AML) is a heterogeneous disease affecting pluripotent stem cells and characterized by ineffective hematopoiesis. In the last years, progresses in molecular technologies lead to identification of numerous gene mutations, which providing more complete insight in oncogenic events and allow to optimize risk stratification of AML patients. Moreover, definition of particular mutations allows personalized approaches to treatments that are based on an individual mutation profiles. More recently, a new class of mutations affecting genes for DNA methylation and post-translational histone modification was identified in AML.

These mutations frequently occur in the DNA nucleotide methyltransferase 3A gene (DNMT3A). DNMT3A belongs to the mammalian methyltransferase gene family that is responsible for tissue-specific gene expression. DNMT3A plays an important role in epigenetically regulated gene expression and repression. DNMT3A mutations represent a potential target in the pathogenesis of AML. The likely mechanism by which DNMT3A loss contributes to leukemogenesis is altered DNA methylation and the attendant gene expression changes. There are evidence that DNMT3A mutation are present in the early pre-leukemic cells, and seems to be a “founder” mutation, which can be implicated as functional components of AML evolution. 

Different studies have shown a negative impact of DNMT3A mutations on outcomes in patients with AML. Prognostic effect is known to depend on certain biological factors as well as a combination of cytogenetics and other mutations. 

In this review we address the biological features of DNMT3A gene, clinical and prognostic impact of DNMT3A, and therapeutic options. 



Keywords


AML, DNMT3A, Methylation

Full Text:

 Subscribers Only

References


References:

Renneville A, Roumier C,, Biggio V, Nibourel J, Boissel N, Fenaux P, et al., Cooperating gene mutations in acute myeloid leukemia: a review of the literature. Leukemia, 2008. 22(5): p. 915-931.

Knudson, A.G., Mutation and Cancer: Statistical Study of Retinoblastoma. Proceedings of the National Academy of Sciences, 1971. 68(4): p. 820-823.

Tucker, T. and J.M. Friedman, Pathogenesis of hereditary tumors: beyond the “two-hit” hypothesis. Clinical Genetics, 2002. 62(5): p. 345-357.

Park, S, Koh Y, Yoon S. Effects of Somatic Mutations Are Associated with SNP in the Progression of Individual Acute Myeloid Leukemia Patient: The Two-Hit Theory Explains Inherited Predisposition to Pathogenesis. Genomics Inform, 2013. 11(1): p. 34-37.

Reya, T., et al., Stem cells, cancer, and cancer stem cells. Nature, 2001. 414(6859): p. 105-111.

Grimwade D. The changing paradigm of prognostic factors in acute myeloid leukaemia. Best Pract Res Clin Haematol, 2012. 25(4): p. 419-25.

Patel JP, Gönen M, Figueroa ME, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366(12):1079–1089.

Döhner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453-474.

Marcucci, G., T. Haferlach, and H. Dohner, Molecular Genetics of Adult Acute Myeloid Leukemia: Prognostic and Therapeutic Implications. Journal of Clinical Oncology, 2011. 29(5): p. 475-486.

Blau, O., Berenstein R, Sindram A, et al., Molecular analysis of different FLT3-ITD mutations in acute myeloid leukemia. Leukemia & Lymphoma, 2013. 54(1): p. 145-152.

Pabst T, Eyholzer M, Fos J, Mueller BU. Heterogeneity within AML with CEBPA mutations; only CEBPA double mutations, but not single CEBPA mutations are associated with favourable prognosis. Br J Cancer. 2009;100(8):1343-1346.

Thakral G, Vierkoetter, Namiki S, Lawicki S, Fernandez X, Ige H, Kawahara W, Lum C . AML multi-gene panel testing: A review and comparison of two gene panels. Pathology – Research and Practice. 2016:212:372–380.

Taskesen E, Bullinger L, Corbacioglu A, et al. Prognostic impact,concurrent genetic mutations, and gene expression features of AML with CEBPA mutations in a cohort of 1182 cytogenetically normal AML patients: further evidence for CEBPA double mutant AML as a distinctive disease entity. Blood. 2011;117(8):2469-2475.

Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–2074.

Döhner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid Leukemia.N Engl J Med.2015;373(12):1136-1152.

Falini B, N.I., Bolli N, Martelli MP, Liso A, Gorello P et al. , Translocations and mutations involving the nucleophosmin (NPM1) gene in lymphomas and leukemias. . Haematologica 2007. 92: 519-532.

O'Donnell MR, Abboud CN, Altman J, et al. Acute myeloid leukemia. J Natl Compr Canc Netw. 2012;10(8):984-1021.

Ley TJ, Ding L, Walter MJ, et al, DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424-2433.

Thol F, Damm F, Lüdeking A, et al. Incidence and prognostic influence of DNMT3a mutations in acute myeloid leukemia. J Clin Oncol. 2011;29(21):2889-2896.

Gaidzik, VI, Schlenk RF, et al. Clinical impact of DNMT3A mutations in younger adult patients with acute myeloid leukemia: results of the AML Study Group (AMLSG). Vol. 121. 2013. 4769-4777.

Ryotokuji T, Yamaguchi H, Ueki T, Usuki K, Kurosawa S et al. Clinical characteristics and prognosis of acute myeloid leukemia associated with DNA-methylation regulatory gene mutations. Haematologica 2016 [Epub ahead of print]

Chou WC, Chou SC, Liu CY, et al. TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics. Blood. 2011;118(14):3803-3810.

Figueroa ME1, 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.

Boissel N, Nibourel O, Renneville A, et al. Prognostic impact of isocitrate dehydrogenase enzyme isoforms 1 and 2 mutations in acute myeloid leukemia: a study by the Acute Leukemia French Association group. J Clin Oncol. 2010;28(23):3717-3723.

Genovese, G., et al., Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence. New England Journal of Medicine, 2014. 371(26): p. 2477-2487.

Jaiswal, S., et al., Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med, 2014. 371(26): p. 2488-98.

Shlush, L.I., et al., Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature, 2014. 506(7488): p. 328-33.

Xie, M. et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nature Med. 20, 1472–1478 (2014).

Corces-Zimmerman, M.R., et al., Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Natl Acad Sci U S A, 2014. 111(7): p. 2548-53.

Okano, M., Xie, S.Li, E. Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nature Genet. 19, 219–220 (1998).

Chen, T., Tsujimoto, N. & Li, E. The PWWP domain of Dnmt3a and Dnmt3b is required for directing DNA methylation to the major satellite repeats at pericentric heterochromatin. Mol. Cell. Biol. 24, 9048–9058 (2004).

Jurkowska, R. Z., Jurkowski, T. P. & Jeltsch, A. Structure and function of mammalian DNA methyltransferases. Chembiochem 12, 206–222 (2011).

Xu, F. et al. Molecular and enzymatic profiles of mammalian DNA methyltransferases: structures and targets for drugs. Curr. Med. Chem. 17, 4052–4071 (2010).

Okano, M., Bell, D.W., Haber, D.A., Li, E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99:247–257

Chen, Z.X., Riggs, A.D. Maintenance and regulation of DNA methylation patterns in mammals. Biochem Cell Biol. 2005;83:438–44

Wienholz, B. L. et al. DNMT3L modulates significant and distinct flanking sequence preference for DNA methylation by DNMT3A and DNMT3B in vivo. PLoS Genet. 6, e1001106 (2010).

Jeltsch A. Reading and writing DNA methylation. Nat Struct Mol Biol. 2008;15(10):1003-1004.

Jasielec, J., V. Saloura, and L.A. Godley, The mechanistic role of DNA methylation in myeloid leukemogenesis. Leukemia, 2014. 28(9): p. 1765-73.

Li K.K., et al., DNA methyltransferases in hematologic malignancies. Semin Hematol, 2013. 50(1): p. 48-60.

Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13(7):484-492.

Abdel-Wahab O, Levine RL. Mutations in epigenetic modifiers in the pathogenesis and therapy of acute myeloid leukemia. Blood. 2013;121(18):3563-3572.

Rush M, Appanah R, Lee S, Lam LL, Goyal P, Lorincz MC. Targeting of EZH2 to a defined genomic site is sufficient for recruitment of Dnmt3a but not de novo DNA methylation. Epigenetics. 2009;16;4(6):404-14.

Yang, Y. et al. CRL4B promotes tumorigenesis by coordinating with SUV39H1/HP1/DNMT3A in DNA methylation-based epigenetic silencing. Oncogene 34, 104–118 (2013).

Muramatsu, D., Singh, P. B., Kimura, H.,Tachibana, M. & Shinkai, Y. Pericentric heterochromatin generated by HP1 protein interaction-defective histone methyltransferase Suv39h1. J. Biol. Chem. 288, 25285–25296 (2013).

Yang L, Rau R, Goodell M. DNMT3A in haematological malignancies. Nature Reviews. 2015;15:152-165

Couronne, L., Bastard, C. & Bernard, O. A. TET2 and DNMT3A mutations in human T-cell lymphoma.N. Engl. J. Med. 366, 95–96 (2012).

Grossmann, V. 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 52, 410–422 (2013).

Hou, Y. et al. Single-cell exome sequencing and monoclonal evolution of a JAK2-negative myeloproliferative neoplasm. Cell 148, 873–885 (2012).

Stegelmann, F. et al. DNMT3A mutations in myeloproliferative neoplasms. Leukemia 25, 1217–1219 (2011).

Walter, M.J., Ding, L., Shen, D., Shao, J., Grillot, M., McLellan, M. et al, Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia. 2011;25:1153–1158

Thol, F., Winschel, C., Ludeking, A., Yun, H., Friesen, I., Damm, F. et al, Rare occurrence of DNMT3A mutations in myelodysplastic syndromes. Haematologica. 2011;96:1870–1873;

Grossmann, V., Kohlmann, A., Haferlach, C., Alpermann, T., Wild, M., Weissmann, S. et al, Landmark analyses of DNMT3A mutations in hematological malignancies. Blood. 2011;:407

Yamashita, Y., Yuan, J., Suetake, I., Suzuki, H., Ishikawa, Y., Choi, Y.L. et al, Array-based genomic resequencing of human leukemia. Oncogene. 2010;29:3723–3731.

Yan, X.J., Xu, J., Gu, Z.H., Pan, C.M., Lu, G., Shen, Y. et al, Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat Genet. 2011;43:309–315.

Gaidzik, V.I., Schlenk, R.F., Paschka, P., Stolzle, A., Corbacioglu, A., Nachbaur, D. et al, DNMT3A mutations predict for inferior outcome in NPM1-wildtype and molecular unfavorable cytogenetically-normal acute myeloid leukemia: a study of the German-Austrian AMLSG. Blood. 2011;:415

O’Brien, E.C., J. Brewin, and T. Chevassut, DNMT3A: the DioNysian MonsTer of acute myeloid leukaemia. Therapeutic Advances in Hematology, 2014. 5(6): p. 187-196.

Yan, X. J. et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nature Genet. 43, 309–315 (2011)

Holz-Schietinger C, D.M. Matje DM, Reich NO. Mutations in DNA methyltransferase (DNMT3A) observed in acute myeloid leukemia patients disrupt processive methylation. J Biol Chem, 2012. 287(37): p. 30941-51.

Russler-Germain D.A., et al., The R882H DNMT3A mutation associated with AML dominantly inhibits wild-type DNMT3A by blocking its ability to form active tetramers. Cancer Cell, 2014. 25(4): p. 442-54.

McDevitt MA., Clinical applications of epigenetic markers and epigenetic profiling in myeloid malignancies. Semin Oncol, 2012. 39(1): p. 109-22.

Qu, Y. et al. Differential methylation in CN-AML preferentially targets non-CGI regions and is dictated by DNMT3A mutational status and associated with predominant hypomethylation of HOX genes. Epigenetics 9, 1108–1119 (2014).

Challen, G. A. et al. Dnmt3a and Dnmt3b have overlapping and distinct functions in hematopoietic stem cells. Cell Stem Cell 15, 350–364 (2014).

Chen T. Mechanistic and functional links between histone methylation and DNA methylation. Prog Mol Biol Transl Sci 2011;101:335-48.

Guo X, Wang L, Li J, et al. Structural insight into autoinhibition and histone H3-induced activation of DNMT3A. Nature 2015;517(7536):640-4.

Rau R, Rodriguez B, Luo M, et al. DOT1L as a therapeutic target for the treatment of DNMT3A-mutant acute myeloid leukemia. Blood. 2016 [Epub ahead of print]

National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology. Acute myeloid leukemia. V. 2. 2014

Ribeiro, A.F., et al., Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia. Blood, 2012. 119(24): p. 5824-31.

Hou, H.A., et al., DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications. Blood, 2012. 119(2): p. 559-68.

Figueroa, M. E. et al. DNA methylation signatures identify biologically distinct subtypes in acute myeloid leukemia. Cancer Cell 17, 13–27 (2010).

Wakita S, Yamaguchi H, Omori I, et al. Mutations of the epigenetics-modifying gene (DNMT3a, TET2, IDH1/2) at diagnosis may induce FLT3-ITD at relapse in de novo acute myeloid leukemia. Leukemia. 2013;27(5):1044-1052.

Fried, I. et al. Frequency, onset and clinical impact of somatic DNMT3A mutations in therapy-related and secondary acute myeloid leukemia. Haematologica 97, 246–250 (2012).

Zebisch, A., Hoefler, G., Quehenberger, F., Wolfler, A. & Sill, H. Mutant DNMT3A in acute myeloid leukemia: guilty of inducing genetic instability? Leukemia 27, 1777–1778 (2013).

Lu, C. et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 483, 474–478 (2012).

Jung SH, Kim YJ, Yim SH, Kim HJ, Kwon YR, Hur EH, Goo BK, Choi YS, Lee SH, Chung YJ, Lee JH. Somatic mutations predict outcomes of hypomethylating therapy in patients with myelodysplastic syndrome. Oncotarget. 2016 [Epub ahead of print]

Ploen, G.G., et al., Persistence of DNMT3A mutations at long-term remission in adult patients with AML. Br J Haematol, 2014. 167(4): p. 478-86.

Xu P, Hu G, Luo C, Liang Z. DNA methyltransferase inhibitirs: an updated patent review. Expert Opinion on theraputic patent 2016 [Epub ahead of print]




DOI: http://dx.doi.org/10.18103/imr.v2i9.172

Refbacks

  • There are currently no refbacks.
Copyright 2016. All rights reserved.